Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.471
Filtrar
1.
Front Endocrinol (Lausanne) ; 15: 1410433, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39359415

RESUMEN

Background: Sex steroid hormones, primarily synthesized by gonadal somatic cells, are pivotal for sexual development and reproduction. Mice studies have shown that two transcription factors, steroidogenic factor 1 (SF-1) and Wilms' tumor 1 (WT1), are involved in gonadal development. However, their role in human gonadal somatic differentiation remains unclear. We therefore aimed to investigate the roles of SF-1 and WT1 in human gonadal steroidogenic cell differentiation. Methods: Using a transient lentivirus-mediated gene expression system, we assessed the effects of SF-1 and WT1 expression on the steroidogenic potential of human amniotic membrane-derived mesenchymal stem cells (hAmMSCs). Results: SF-1 and WT1-KTS, a splice variant of WT1, played distinct roles in human steroidogenic differentiation of hAmMSCs. SF-1 induced hAmMSC differentiation into progesterone- and androgen-producing cell lineages, whereas WT1-KTS promoted hAmMSC differentiation into estrogen-producing cell lineages. Conclusion: Our findings revealed that SF-1 and WT1-KTS play important roles in human gonadal steroidogenic cell differentiation, especially during ovarian development. These findings may pave the way for future studies on human ovarian differentiation and development.


Asunto(s)
Amnios , Andrógenos , Diferenciación Celular , Linaje de la Célula , Estrógenos , Células Madre Mesenquimatosas , Progesterona , Factor Esteroidogénico 1 , Proteínas WT1 , Humanos , Proteínas WT1/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Factor Esteroidogénico 1/metabolismo , Factor Esteroidogénico 1/genética , Progesterona/metabolismo , Progesterona/biosíntesis , Estrógenos/metabolismo , Andrógenos/metabolismo , Amnios/citología , Amnios/metabolismo , Femenino , Células Cultivadas , Factores de Empalme de ARN
2.
Sci Rep ; 14(1): 23717, 2024 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-39390042

RESUMEN

With the increasing demand for exercise, the population of patients with ankle sprain to anterior talofibular ligament injury has the characteristics of a large base and high requirements for returning to sports, and how to promote the repair of damaged ligaments from a microscopic perspective is an urgent problem to be solved. In many studies, human amniotic mesenchymal stem cells have strong differentiation ability, and can be induced to continuously differentiate into ligament cells to achieve the purpose of repairing damaged ligaments. Human amniotic stem cells were extracted and cultured from human amniotic tissues, evaluated by cell identification and other techniques, and evaluated into ligament differentiation by toluidine blue, alizarin red, oil red O staining and detection of ligament cell differentiation, protein detection by Western blot, mRNA level by qPCR, and finally, the targeted binding relationship between miR-16a-5p and mRNA FGF2 was verified by double luciferase reporter assay. The expression of collagen type 1 (COL 1), collagen type 3 (COL3), SCX and MKX was increased by overexpression of mRNA FGF2, respectively, and miR-16a-5p had a targeted effect on FGF2 and regulated the ligamentous differentiation of human amniotic mesenchymal stem cells. We found that the regulatory effect of overexpressed mRNA FGF2 on mesenchymal stem cells could be inhibited by up-regulation of miR-16a-5p, while the knockdown of FGF2 could reverse the regulatory effect of miR-16a-5p inhibition on ligament-forming differentiation of human amniotic mesenchymal stem cells. In this study, we discovered the existence of the miR-16a-5p-FGF2 axis in human amniotic mesenchymal stem cells, and the differentiation of human amniotic mesenchymal stem cells into ligamentous cells can be regulated by regulating various links in this axis.


Asunto(s)
Diferenciación Celular , Factor 2 de Crecimiento de Fibroblastos , Células Madre Mesenquimatosas , MicroARNs , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Regeneración , Amnios/citología , Amnios/metabolismo , Células Cultivadas , Tendones/metabolismo , Tendones/citología , Colágeno Tipo III/metabolismo , Colágeno Tipo III/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico
3.
Exp Clin Transplant ; 22(7): 576-578, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39223817

RESUMEN

We report the successful reconstruction of suture exposure with the oral mucosal graft in a patient with suture exposure after transscleral-sutured posterior chamber intraocular lens implantation. The 70-year-old patient had a history of vitreoretinal surgery and transscleral-sutured posterior chamber intraocular lens implantation after complicated cataract surgery. He was referred to our department because of suture exposure. The best-corrected visual acuity was 20/2000 OD and 20/50 OS. We observed exposed PC9 sutures from both the nasal and temporal conjunctiva in the right eye. The patient showed appearance of scleromalacia in the same regions, so scleral flap surgery was not considered. Despite both tenoplasty and amniotic membrane transplant procedures, exposure could not be controlled. Instead, the patient received oral (buccal) mucosal graft transplant to the resistant exposure areas. A single layer of protective amniotic membrane was transplanted over the buccal mucosal graft. This method resulted in effective control of the exposed area. In conclusion, an oral mucosal graft can be used in many ocular pathologies that require conjunctival reconstruction because of the simplicity of tissue excision from the mucosa, allowing adequate tissue excision, durability of the obtained tissue, and ease of use. Our case report highlights that resistant transscleral-sutured posterior chamber intraocular lens suture exposure can be successfully managed with oral mucosal grafting.


Asunto(s)
Implantación de Lentes Intraoculares , Mucosa Bucal , Técnicas de Sutura , Agudeza Visual , Humanos , Masculino , Implantación de Lentes Intraoculares/efectos adversos , Anciano , Mucosa Bucal/trasplante , Resultado del Tratamiento , Esclerótica/cirugía , Esclerótica/trasplante , Amnios/trasplante , Lentes Intraoculares , Extracción de Catarata
4.
Mol Med ; 30(1): 138, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39232672

RESUMEN

BACKGROUND: Premature rupture of the membranes (PROM) is a key cause of preterm birth and represents a major cause of neonatal mortality and morbidity. Natural products N-acetyl-d-galactosamine (GalNAc), which are basic building blocks of important polysaccharides in biological cells or tissues, such as chitin, glycoproteins, and glycolipids, may improve possible effects of wound healing. METHODS: An in vitro inflammation and oxidative stress model was constructed using tumor necrosis-α (TNF-α) and lipopolysaccharide (LPS) action on WISH cells. Human amniotic epithelial cells (hAECs) were primarily cultured by digestion to construct a wound model. The effects of GalNAc on anti-inflammatory and anti-oxidative stress, migration and proliferation, epithelial-mesenchymal transition (EMT), glycosaminoglycan (GAG)/hyaluronic acid (HA) production, and protein kinase B (Akt) pathway in hAECs and WISH cells were analyzed using the DCFH-DA fluorescent probe, ELISA, CCK-8, scratch, transwell migration, and western blot to determine the mechanism by which GalNAc promotes amniotic wound healing. RESULTS: GalNAc decreased IL-6 expression in TNF-α-stimulated WISH cells and ROS expression in LPS-stimulated WISH cells (P < 0.05). GalNAc promoted the expression of Gal-1 and Gal-3 with anti-inflammatory and anti-oxidative stress effects. GalNAc promoted the migration of hAECs (50% vs. 80%) and WISH cells through the Akt signaling pathway, EMT reached the point of promoting fetal membrane healing, and GalNAc did not affect the activity of hAECs and WISH cells (P > 0.05). GalNAc upregulated the expression of sGAG in WISH cells (P < 0.05) but did not affect HA levels (P > 0.05). CONCLUSIONS: GalNAc might be a potential target for the prevention and treatment of PROM through the galectin pathway, including (i) inflammation; (ii) epithelial-mesenchymal transition; (iii) proliferation and migration; and (iv) regression, remodeling, and healing.


Asunto(s)
Acetilgalactosamina , Movimiento Celular , Transición Epitelial-Mesenquimal , Rotura Prematura de Membranas Fetales , Galectinas , Transducción de Señal , Cicatrización de Heridas , Humanos , Rotura Prematura de Membranas Fetales/metabolismo , Acetilgalactosamina/metabolismo , Acetilgalactosamina/análogos & derivados , Galectinas/metabolismo , Embarazo , Células Epiteliales/metabolismo , Línea Celular , Estrés Oxidativo , Femenino , Amnios/metabolismo , Amnios/citología , Proliferación Celular , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo
5.
Transl Vis Sci Technol ; 13(9): 10, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39235403

RESUMEN

Purpose: Pterygium is an ocular surface disease characterized by the invasion of fibrovascular tissue from the bulbar conjunctiva to the cornea and is associated with abnormal tear function caused by changes in tear composition and osmolarity. In this study, the effect of two different surgical techniques to remove primary pterygium: conjunctival autograft surgery (CAG) and amniotic membrane transplantation (AMT), on changes in MUC2 and MUC5AC tear mucins concentration were evaluated. Methods: Forty-four patients (>18 years old) with primary unilateral pterygium (> 1.0 mm long, measured from the limbus to the apex on the cornea) were randomly enrolled, and assigned to the AMT or CAG group by using the permuted block technique. Patients with systemic inflammatory diseases or other eye comorbidities were excluded from the study. Tear break-up time (TBUT) and best-corrected visual acuity (BCVA) assessments were performed before surgery and at 1, 3, and 6 months after surgery. Tears were collected concurrently with the clinical evaluations, and MUC2 and MUC5AC concentrations were subsequently measured by means of ELISA. Results: At 6 months after CAG or AMT, TBUT and BCVA were significantly lower (P < 0.05) in comparison with the baseline values in the study subjects. The tear mucin concentrations of both MUC2 and MUC5AC were significantly higher (P < 0.0001) in patients with pterygium before any surgical procedure than in healthy individuals. The concentration of MUC2 increased at 1 and 3 months after CAG surgery and decreased at 6 months; however, the MUC2 concentration decreased on the AMT group in all time point measurements. Interestingly, the MUC5AC concentration significantly increased at 1 month after AMT or CAG and then decreased at 3 and 6 months after surgery. Finally, an inverse correlation was found between both MUC2 and MUC5AC tear mucins concentration and the TBUT. Conclusions: These results suggest that pterygium excision via both CAG or AMT changes the concentrations of the tear mucins MUC2 and MUC5AC during the evaluated times, and these changes could affect tear film stability and clinical recovery after pterygium treatment. Translational Relevance: The tear film stability during pterygium excision was evaluated to determine adequate treatments.


Asunto(s)
Amnios , Conjuntiva , Mucina 5AC , Mucina 2 , Pterigion , Lágrimas , Humanos , Masculino , Pterigion/cirugía , Pterigion/metabolismo , Femenino , Persona de Mediana Edad , Conjuntiva/metabolismo , Conjuntiva/trasplante , Mucina 2/metabolismo , Lágrimas/metabolismo , Amnios/trasplante , Amnios/metabolismo , Estudios de Seguimiento , Mucina 5AC/metabolismo , Anciano , Adulto , Autoinjertos , Agudeza Visual , Ensayo de Inmunoadsorción Enzimática , Trasplante Autólogo/métodos , Estudios Prospectivos
6.
Taiwan J Obstet Gynecol ; 63(5): 755-758, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39266160

RESUMEN

OBJECTIVE: We present low-level mosaic trisomy 14 at amniocentesis. CASE REPORT: A 37-year-old, gravida 2, para 1, woman underwent amniocentesis at 18 weeks of gestation because of advanced maternal age. This pregnancy was conceived by in vitro fertilization and embryo transfer (IVF-ET). Amniocentesis revealed a karyotype of 47,XX,+14 [4]/46,XX [27], consistent with 12.9% mosaicism for trisomy 14. Simultaneous array comparative genomic hybridization (aCGH) analysis on the DNA extracted from uncultured amniocytes revealed the result of arr (1-22, X) × 2 with no genomic imbalance. Prenatal ultrasound findings were unremarkable. She was referred for genetic counseling at 21 weeks of gestation and was offered expanded non-invasive prenatal testing (NIPT) which was positive for trisomy 14. At 24 weeks of gestation, she underwent repeat amniocentesis which revealed a karyotype of 47,XX,+14 [2]/46,XX [26], consistent with 7% mosaicism for trisomy 14. The parental karyotypes were normal. Simultaneous aCGH analysis on the DNA extracted from uncultured amniocytes revealed no genomic imbalance. Polymorphic marker analysis excluded uniparental disomy (UPD) 14. Interphase fluorescence in situ hybridization (FISH) analysis on 104 uncultured amniocytes detected no trisomy 14 cell. At 35 weeks of gestation, a 2315-g phenotypically normal baby was delivered. The umbilical cord and placenta had the karyotype of 46, XX (40/40 cells). aCGH analysis on the DNA extracted from peripheral blood and buccal mucosal cells at the age of three months revealed no genomic imbalance. The neonate was normal in phenotype and development during postnatal follow-ups. CONCLUSIONS: Low-level mosaic trisomy 14 at amniocentesis can be associated with cytogenetic discrepancy between cultured amniocytes and uncultured amniocytes, perinatal progressive decrease of the trisomy 14 cell line and a favorable fetal outcome.


Asunto(s)
Amniocentesis , Cromosomas Humanos Par 14 , Hibridación Genómica Comparativa , Mosaicismo , Trisomía , Disomía Uniparental , Humanos , Embarazo , Femenino , Mosaicismo/embriología , Trisomía/diagnóstico , Trisomía/genética , Adulto , Disomía Uniparental/diagnóstico , Disomía Uniparental/genética , Cromosomas Humanos Par 14/genética , Recién Nacido , Pruebas Prenatales no Invasivas/métodos , Nacimiento Vivo/genética , Amnios/citología , Resultado del Embarazo/genética , Cariotipificación/métodos
7.
Elife ; 132024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39344495

RESUMEN

A complex extracted from the amniotic membrane in humans reduces post-surgical pain in mice by directly inhibiting pain-sensing neurons.


Asunto(s)
Analgésicos Opioides , Dolor Postoperatorio , Animales , Ratones , Dolor Postoperatorio/tratamiento farmacológico , Humanos , Analgésicos Opioides/farmacología , Amnios , Neuronas/fisiología , Neuronas/efectos de los fármacos
8.
Front Immunol ; 15: 1459489, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39290694

RESUMEN

Introduction: Fetal membrane inflammation is an integral event of parturition. However, excessive pro-inflammatory cytokines can impose threats to the fetus. Coincidentally, the fetal membranes express abundant 11ß-hydroxysteroid dehydrogenase 1 (11ß-HSD1), which generates biologically active cortisol to promote labor through induction of prostaglandin synthesis. Given the well-recognized anti-inflammatory actions of glucocorticoids, we hypothesized that cortisol regenerated in the fetal membranes might be engaged in restraining fetus-hazardous pro-inflammatory cytokine production for the safety of the fetus, while reserving pro-labor effect on prostaglandin synthesis to ensure safe delivery of the fetus. Methods: The hypothesis was examined in human amnion tissue and cultured primary human amnion fibroblasts as well as a mouse model. Results: 11ß-HSD1 was significantly increased in the human amnion in infection-induced preterm birth. Studies in human amnion fibroblasts showed that lipopolysaccharide (LPS) induced 11ß-HSD1 expression synergistically with cortisol. Cortisol completely blocked NF-κB-mediated pro-inflammatory cytokine expression by LPS, but STAT3-mediated cyclooxygenase 2 expression, a crucial prostaglandin synthetic enzyme, remained. Further studies in pregnant mice showed that corticosterone did not delay LPS-induced preterm birth, but alleviated LPS-induced fetal organ damages, along with increased 11ß-HSD1, cyclooxygenase 2, and decreased pro-inflammatory cytokine in the fetal membranes. Discussion: There is a feed-forward cortisol regeneration in the fetal membranes in infection, and cortisol regenerated restrains pro-inflammatory cytokine expression, while reserves pro-labor effect on prostaglandin synthesis. This dual role of cortisol regeneration can prevent excessive pro-inflammatory cytokine production, while ensure in-time delivery for the safety of the fetus.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1 , Amnios , Fibroblastos , Glucocorticoides , Inflamación , Parto , Humanos , Animales , Femenino , Embarazo , Ratones , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Amnios/metabolismo , Inflamación/metabolismo , Fibroblastos/metabolismo , Citocinas/metabolismo , Regeneración , Lipopolisacáridos , Células Cultivadas , Nacimiento Prematuro/inmunología , Hidrocortisona
9.
BMC Ophthalmol ; 24(1): 409, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39300402

RESUMEN

BACKGROUND: Human amniotic membrane (AM) transplantation has been applied to treat ocular surface diseases, including corneal trauma. The focus of much deliberation is to balance the mechanical strength of the amniotic membrane, its resistance to biodegradation, and its therapeutic efficacy. It is commonly observed that the crosslinked human decellularized amniotic membranes lose the functional human amniotic epithelial cells (hAECs), which play a key role in curing the injured tissues. METHODS AND RESULTS: In this study, we crosslinked human decellularized amniotic membranes (dAM) with genipin and re-planted the hAECs onto the genipin crosslinked AM. The properties of the AM were evaluated based on optical clarity, biodegradation, cytotoxicity, and ultrastructure. The crosslinked AM maintained its transparency. The color of crosslinked AM deepened with increasing concentrations of genipin. And the extracts from low concentrations of genipin crosslinked AM had no toxic effect on human corneal epithelial cells (HCECs), while high concentrations of genipin exhibited cytotoxicity. The microscopic observation and H&E staining revealed that 2 mg/mL genipin-crosslinked dAM (2 mg/mL cl-dAM) was more favorable for the attachment, migration, and proliferation of hAECs. Moreover, the results of the CCK-8 assay and the transwell assay further indicated that the living hAECs' tissue-engineered amniotic membranes could facilitate the proliferation and migration of human corneal stromal cells (HCSCs) in vitro. CONCLUSIONS: In conclusion, the cl-dAM with living hAECs demonstrates superior biostability and holds significant promise as a material for ocular surface tissue repair in clinical applications.


Asunto(s)
Amnios , Proliferación Celular , Epitelio Corneal , Ingeniería de Tejidos , Humanos , Ingeniería de Tejidos/métodos , Epitelio Corneal/citología , Células Cultivadas , Enfermedades de la Córnea/cirugía , Iridoides/farmacología , Células Epiteliales
10.
Injury ; 55(11): 111901, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39341049

RESUMEN

Amniotic-derived products have been used for decades in various medical subspecialties and have proven to be a safe method of allograft tissue transplantation. These products have shown promising preclinical and early clinical results in the treatment of tendon/ligament injuries, cartilage defects, and osteoarthritis. The therapeutic benefits of amniotic-derived products are likely due to intrinsic properties, such as their structure as an extracellular matrix and concentration of growth factors, as well as anti-inflammatory, antifibrotic, and antimicrobial molecules. We performed a narrative review, evaluating the pre-clinical and clinical use of amniotic-derived products in musculoskeletal injuries such as osteoarthritis, Achilles tendinopathy, plantar fasciitis, lateral epicondylitis, chronic stenosing tenosynovitis, and nerve, cartilage and tendon repair or reconstruction, along with fracture healing treatment. In vitro and pre-clinical studies using amniotic-derived products for orthopedic treatments have shown promising results and provide the foundation for further human trials to be conducted. With the rise of commercially available biologics, incorporating amniotic products into orthopedic practice is becoming more accessible, while further studies investigating long-term outcomes and potential adverse events are necessary.


Asunto(s)
Amnios , Procedimientos Ortopédicos , Humanos , Amnios/trasplante , Procedimientos Ortopédicos/métodos , Productos Biológicos/uso terapéutico
11.
FASEB J ; 38(16): e70004, 2024 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-39190010

RESUMEN

Previous studies showed that the bladder extracellular matrix (B-ECM) could increase the differentiation efficiency of mesenchymal cells into smooth muscle cells (SMC). This study investigates the potential of human amniotic membrane-derived hydrogel (HAM-hydrogel) as an alternative to xenogeneic B-ECM for the myogenic differentiation of the rabbit adipose tissue-derived MSC (AD-MSC). Decellularized human amniotic membrane (HAM) and sheep urinary bladder (SUB) were utilized to create pre-gel solutions for hydrogel formation. Rabbit AD-MSCs were cultured on SUB-hydrogel or HAM-hydrogel-coated plates supplemented with differentiation media containing myogenic growth factors (PDGF-BB and TGF-ß1). An uncoated plate served as the control. After 2 weeks, real-time qPCR, immunocytochemistry, flow cytometry, and western blot were employed to assess the expression of SMC-specific markers (MHC and α-SMA) at both protein and mRNA levels. Our decellularization protocol efficiently removed cell nuclei from the bladder and amniotic tissues, preserving key ECM components (collagen, mucopolysaccharides, and elastin) within the hydrogels. Compared to the control, the hydrogel-coated groups exhibited significantly upregulated expression of SMC markers (p ≤ .05). These findings suggest HAM-hydrogel as a promising xenogeneic-free alternative for bladder tissue engineering, potentially overcoming limitations associated with ethical concerns and contamination risks of xenogeneic materials.


Asunto(s)
Amnios , Diferenciación Celular , Hidrogeles , Células Madre Mesenquimatosas , Miocitos del Músculo Liso , Animales , Amnios/citología , Amnios/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Conejos , Humanos , Hidrogeles/química , Vejiga Urinaria/citología , Vejiga Urinaria/metabolismo , Matriz Extracelular/metabolismo , Ovinos , Células Cultivadas , Ingeniería de Tejidos/métodos
12.
Med J Malaysia ; 79(Suppl 4): 68-71, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-39215418

RESUMEN

INTRODUCTION: Gastric perforation is a rare occurrence, particularly in neonates. This is an emergency case in this population. The incidence of spontaneous gastric perforation in neonates is 1:2900 live births, with high mortality and morbidity rates. The primary treatment is surgical debridement and repair of the perforation, which has a high incidence of anastomotic leakage. At present, there is a plethora of studies investigating the efficacy of human dried amniotic membrane (H-DAM) technology in promoting wound healing. Consequently, researchers sought to ascertain whether there were differences in the number of adhesion and abscess classifications for the macroscopic evaluation of gastric perforation repair with HDAM as a biomaterial in New Zealand white rabbits. MATERIAL AND METHODS: A total of 30 male New Zealand rabbits underwent laparotomy and gastric perforation. These animals were then divided into three groups, with each group comprising 10 rabbits. Group 1 underwent primary repair, group 2 underwent omental patch repair, and group 3 underwent H-DAM patch repair. The rabbits were euthanised on the 7th day and the adhesion score and abscess classification were evaluated. RESULT: A total of 30 samples of rabbits were homogeneous. On macroscopic evaluation, it was found that the H-DAM had the lowest mean adhesion score and the lowest incidence of abscess formation compared to all other groups. CONCLUSIONS: It can be concluded that the utilisation of HDAM as a biomaterial patch in the treatment of gastric perforation in the rabbit model did not result in any instances of leakage, adhesion or infection.


Asunto(s)
Amnios , Cicatrización de Heridas , Animales , Conejos , Amnios/trasplante , Masculino , Humanos , Materiales Biocompatibles
13.
Int J Mol Sci ; 25(16)2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39201288

RESUMEN

The functioning of the human cornea heavily relies on the maintenance of its extracellular matrix (ECM) mechanical properties. Within this context, corneal stromal fibroblasts (CSFs) are essential, as they are responsible for remodeling the corneal ECM. In this study, we used a decellularized human amniotic membrane (dHAM) and a custom fibrillar collagen film (FCF) to explore the effects of fibrillar materials on human CSFs. Our findings indicate that substrates like FCF can enhance the early development of focal adhesions (FAs), leading to the activation and propagation of mechanotransduction signals. This is primarily achieved through FAK autophosphorylation and YAP1 nuclear translocation pathways. Remarkably, inhibiting FAK autophosphorylation negated the observed changes. Proteome analysis further confirmed the central role of FAs in mechanotransduction propagation in CSFs cultured on FCF. This analysis also highlighted complex signaling pathways, including chromatin epigenetic modifications, in response to fibrillar substrates. Overall, our research highlights the potential pathways through which CSFs undergo behavioral changes when exposed to fibrillar substrates, identifying FAs as essential mechanotransducers.


Asunto(s)
Sustancia Propia , Fibroblastos , Adhesiones Focales , Mecanotransducción Celular , Humanos , Adhesiones Focales/metabolismo , Fibroblastos/metabolismo , Sustancia Propia/citología , Sustancia Propia/metabolismo , Fosforilación , Matriz Extracelular/metabolismo , Células Cultivadas , Proteínas Señalizadoras YAP/metabolismo , Colágenos Fibrilares/metabolismo , Amnios/citología , Amnios/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo
14.
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
15.
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
16.
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
17.
J Mater Chem B ; 12(36): 8977-8992, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39140678

RESUMEN

Stem cell-based therapy implementation relies heavily on advancements in cell tracking. The present research has been designed to develop a gold nanorod (AuNR) labeling protocol applied to amniotic epithelial cells (AECs) leveraging the pro-regenerative properties of this placental stem cell source which is widely used for both human and veterinary biomedical regenerative applications, although not yet exploited with tracking technologies. Ovine AECs, in native or induced mesenchymal (mAECs) phenotypes via epithelial-mesenchymal transition (EMT), served as the model. Initially, various uptake methods validated on other sources of mesenchymal stromal cells (MSCs) were assessed on mAECs before optimization for AECs. Furthermore, the protocol was implemented by adopting the biological strategy of MitoCeption to improve endocytosis. The results indicate that the most efficient, affordable, and easy protocol leading to internalization of AuNRs in living mAECs recognized the combination of the one-step uptake condition (cell in suspension), centrifugation-mediated internalization method (G-force) and MitoCeption (mitochondrial isolated from mAECs). This protocol produced labeled vital mAECs within minutes, suitable for preclinical and clinical trials. The optimized protocol has the potential to yield feasible labeled amniotic-derived cells for biomedical purposes: up to 10 million starting from a single amniotic membrane. Similar and even higher efficiency was found when the protocol was applied to ovine and human AECs, thereby demonstrating the transferability of the method to cells of different phenotypes and species-specificity, hence validating its great potential for the development of improved biomedical applications in cell-based therapy and diagnostic imaging.


Asunto(s)
Amnios , Oro , Animales , Ovinos , Oro/química , Amnios/química , Amnios/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Humanos , Células Epiteliales/citología , Nanopartículas del Metal/química , Células Cultivadas , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Tamaño de la Partícula
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA