Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 475
Filtrar
1.
Stem Cell Res Ther ; 15(1): 120, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38659015

RESUMEN

BACKGROUND: Diabetic cardiomyopathy (DCM) is a serious health-threatening complication of diabetes mellitus characterized by myocardial fibrosis and abnormal cardiac function. Human umbilical cord mesenchymal stromal cells (hUC-MSCs) are a potential therapeutic tool for DCM and myocardial fibrosis via mechanisms such as the regulation of microRNA (miRNA) expression and inflammation. It remains unclear, however, whether hUC-MSC therapy has beneficial effects on cardiac function following different durations of diabetes and which mechanistic aspects of DCM are modulated by hUC-MSC administration at different stages of its development. This study aimed to investigate the therapeutic effects of intravenous administration of hUC-MSCs on DCM following different durations of hyperglycemia in an experimental male model of diabetes and to determine the effects on expression of candidate miRNAs, target mRNA and inflammatory mediators. METHODS: A male mouse model of diabetes was induced by multiple low-dose streptozotocin injections. The effects on severity of DCM of intravenous injections of hUC-MSCs and saline two weeks previously were compared at 10 and 18 weeks after diabetes induction. At both time-points, biochemical assays, echocardiography, histopathology, polymerase chain reaction (PCR), immunohistochemistry and enzyme-linked immunosorbent assays (ELISA) were used to analyze blood glucose, body weight, cardiac structure and function, degree of myocardial fibrosis and expression of fibrosis-related mRNA, miRNA and inflammatory mediators. RESULTS: Saline-treated diabetic male mice had impaired cardiac function and increased cardiac fibrosis after 10 and 18 weeks of diabetes. At both time-points, cardiac dysfunction and fibrosis were improved in hUC-MSC-treated mice. Pro-fibrotic indicators (α-SMA, collagen I, collagen III, Smad3, Smad4) were reduced and anti-fibrotic mediators (FGF-1, miRNA-133a) were increased in hearts of diabetic animals receiving hUC-MSCs compared to saline. Increased blood levels of pro-inflammatory cytokines (IL-6, TNF, IL-1ß) and increased cardiac expression of IL-6 were also observed in saline-treated mice and were reduced by hUC-MSCs at both time-points, but to a lesser degree at 18 weeks. CONCLUSION: Intravenous injection of hUC-MSCs ameliorated key functional and structural features of DCM in male mice with diabetes of shorter and longer duration. Mechanistically, these effects were associated with restoration of intra-myocardial expression of miRNA-133a and its target mRNA COL1AI as well as suppression of systemic and localized inflammatory mediators.


Asunto(s)
Diabetes Mellitus Experimental , Cardiomiopatías Diabéticas , Fibrosis , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , MicroARNs , Miocardio , Cordón Umbilical , Animales , Humanos , Masculino , Ratones , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Experimental/metabolismo , Cardiomiopatías Diabéticas/terapia , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/genética , Fibrosis/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Miocardio/metabolismo , Miocardio/patología , Cordón Umbilical/citología , Cordón Umbilical/metabolismo
2.
Ocul Surf ; 32: 13-25, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38191093

RESUMEN

PURPOSE: Corneal fibrosis and neovascularization (CNV) after ocular trauma impairs vision. This study tested therapeutic potential of tissue-targeted adeno-associated virus5 (AAV5) mediated decorin (DCN) and pigment epithelium-derived factor (PEDF) combination genes in vivo. METHODS: Corneal fibrosis and CNV were induced in New Zealand White rabbits via chemical trauma. Gene therapy in stroma was delivered 30-min after chemical-trauma via topical AAV5-DCN and AAV5-PEDF application using a cloning cylinder. Clinical eye examinations and multimodal imaging in live rabbits were performed periodically and corneal tissues were collected 9-day and 15-day post euthanasia. Histological, cellular, and molecular and apoptosis assays were used for efficacy, tolerability, and mechanistic studies. RESULTS: The AAV5-DCN and AAV5-PEDF combination gene therapy significantly reduced corneal fibrosis (p < 0.01 or p < 0.001) and CNV (p < 0.001) in therapy-given (chemical-trauma and AAV5-DCN + AAV5-PEDF) rabbit eyes compared to the no-therapy given eyes (chemical-trauma and AAV5-naked vector). Histopathological analyses demonstrated significantly reduced fibrotic α-smooth muscle actin and endothelial lectin expression in therapy-given corneas compared to no-therapy corneas on day-9 (p < 0.001) and day-15 (p < 0.001). Further, therapy-given corneas showed significantly increased Fas-ligand mRNA levels (p < 0.001) and apoptotic cell death in neovessels (p < 0.001) compared to no-therapy corneas. AAV5 delivered 2.69 × 107 copies of DCN and 2.31 × 107 copies of PEDF genes per µg of DNA. AAV5 vector and delivered DCN and PEDF genes found tolerable to the rabbit eyes and caused no significant toxicity to the cornea. CONCLUSION: The combination AAV5-DCN and AAV5-PEDF topical gene therapy effectively reduces corneal fibrosis and CNV with high tolerability in vivo in rabbits. Additional studies are warranted.


Asunto(s)
Neovascularización de la Córnea , Dependovirus , Modelos Animales de Enfermedad , Proteínas del Ojo , Fibrosis , Terapia Genética , Factores de Crecimiento Nervioso , Serpinas , Animales , Conejos , Terapia Genética/métodos , Fibrosis/terapia , Neovascularización de la Córnea/terapia , Neovascularización de la Córnea/genética , Neovascularización de la Córnea/patología , Neovascularización de la Córnea/metabolismo , Dependovirus/genética , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Serpinas/genética , Serpinas/metabolismo , Decorina/genética , Decorina/metabolismo , Córnea/patología , Córnea/metabolismo , Vectores Genéticos
3.
Ultrasound Med Biol ; 49(12): 2510-2518, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37714800

RESUMEN

OBJECTIVE: The aim of the work described here was to investigate the efficacy and potential mechanisms of low-intensity pulsed ultrasound (LIPUS) for the treatment of arthrogenic contracture induced by immobilization in rabbits. METHODS: The left knee joint of rabbits was immobilized for 6 wk to establish the model of extending knee joint contracture. The rabbits were divided into a control group (C), a group immobilized for 6 wk (IM-6w), a group remobilized for 1 wk (RM-1w), a group subjected to LIPUS intervention for 1 wk (LIPUS-1w), a group remobilized for 2 wk (RM-2w) and a group subjected to LIPUS intervention for 2 wk (LIPUS-2w). The degrees of arthrogenic contracture and joint capsule fibrosis were assessed, as were the levels of reactive oxygen species (ROS) and the activation status of the TGF-ß1/Smad signaling pathway in the joint capsule. RESULTS: After immobilization for 6 wk, the degrees of arthrogenic contracture and joint capsule fibrosis increased. The ROS level increased, as evidenced by an increase in malondialdehyde content and a decrease in superoxide dismutase content. In addition, the TGF-ß1/Smad signaling pathway was significantly activated. The degrees of knee joint contracture increased in the first week after remobilization and decreased in the second week. Furthermore, joint capsule fibrosis continued to develop during the 2 wk of remobilization, and the ROS level increased, while the TGF-ß1/Smad signaling pathway was significantly activated. LIPUS effectively reduced the level of ROS in the joint capsule, which further inhibited activation of the TGF-ß1/Smad signaling pathway, thereby improving joint capsule fibrosis and reducing arthrogenic contracture. CONCLUSION: The high ROS levels and overactivation of the TGF-ß1/Smad signaling pathway may be reasons why immobilization induces knee joint capsule fibrosis. LIPUS can alleviate the degree of knee joint capsule fibrosis induced by immobilization by inhibiting the production of ROS and the activation of the TGF-ß1/Smad signaling pathway.


Asunto(s)
Contractura , Factor de Crecimiento Transformador beta1 , Animales , Conejos , Contractura/metabolismo , Contractura/patología , Fibrosis/terapia , Cápsula Articular/metabolismo , Cápsula Articular/patología , Articulación de la Rodilla/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Ondas Ultrasónicas , Proteínas Smad/metabolismo
4.
Ger Med Sci ; 21: Doc04, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37405192

RESUMEN

Background: Lymphedema is a chronic, progressive clinical condition that evolves with intense fibrosis, the most advanced stage of which is stage III (lymphostatic fibrosclerosis). Aim: The aim of the present study was to show the possibility to reconstruct the dermal layers with the intensive treatment of fibrosis using the Godoy method. Case description: A 55-year-old patient with an eight-year history of edema of the lower limb of the leg had constant episodes of erysipelas, despite regular treatments. The edema progressed continually, associated with a change in the color of the skin and the formation of a crust. Intensive treatment (eight hours per day for three weeks) was proposed with the Godoy method. The ultrasound was performed and results revealed substantial improvement in the skin, with the onset of the reconstruction of the dermal layers. Conclusion: It is possible to reconstruct the layers of the skin in fibrotic conditions caused by lymphedema.


Asunto(s)
Dermis , Fibrosis , Linfedema , Enfermedades de la Piel , Humanos , Persona de Mediana Edad , Enfermedad Crónica , Fibrosis/diagnóstico por imagen , Fibrosis/etiología , Fibrosis/patología , Fibrosis/terapia , Linfedema/complicaciones , Linfedema/diagnóstico por imagen , Linfedema/patología , Linfedema/terapia , Piel/diagnóstico por imagen , Piel/patología , Enfermedades de la Piel/complicaciones , Enfermedades de la Piel/diagnóstico por imagen , Enfermedades de la Piel/patología , Enfermedades de la Piel/terapia , Dermis/diagnóstico por imagen , Dermis/patología , Ultrasonografía/métodos
5.
Nature ; 619(7971): 707-715, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37495877

RESUMEN

Engineering a patient's own T cells to selectively target and eliminate tumour cells has cured patients with untreatable haematologic cancers. These results have energized the field to apply chimaeric antigen receptor (CAR) T therapy throughout oncology. However, evidence from clinical and preclinical studies underscores the potential of CAR T therapy beyond oncology in treating autoimmunity, chronic infections, cardiac fibrosis, senescence-associated disease and other conditions. Concurrently, the deployment of new technologies and platforms provides further opportunity for the application of CAR T therapy to noncancerous pathologies. Here we review the rationale behind CAR T therapy, current challenges faced in oncology, a synopsis of preliminary reports in noncancerous diseases, and a discussion of relevant emerging technologies. We examine potential applications for this therapy in a wide range of contexts. Last, we highlight concerns regarding specificity and safety and outline the path forward for CAR T therapy beyond cancer.


Asunto(s)
Envejecimiento , Enfermedades Autoinmunes , Fibrosis , Cardiopatías , Inmunoterapia Adoptiva , Infecciones , Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/terapia , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Inmunoterapia Adoptiva/tendencias , Neoplasias/inmunología , Neoplasias/terapia , Receptores Quiméricos de Antígenos/uso terapéutico , Linfocitos T/inmunología , Enfermedades Autoinmunes/terapia , Infecciones/terapia , Fibrosis/terapia , Envejecimiento/patología , Cardiopatías/terapia
6.
BMC Urol ; 22(1): 69, 2022 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-35462546

RESUMEN

INTRODUCTION: Bladder outlet obstruction (BOO) was caused by a series of histological and biochemical changes in the bladder wall, through the inflammation process in the bladder wall, hypertrophy and fibrosis. ADSC has an important role in bladder regeneration. METHODS AND MATERIALS: This study was an experimental randomized study using male Wistar rats which were monitored at 2 and 4 weeks to determine the effect of ADSC therapy on TGF-ß1 type I collagen, and degree of fibrosis. RESULT: Rats were divided into 5 groups. In the week 2 BOO group, 1 sample included in the category of moderate fibrosis, 1 sample that was given ADSC with mild fibrosis category, 3 samples included in severe fibrosis category, 3 samples that were given ADSC included in the category of moderate fibrosis. The concentration of TGF-ß1 in the hADSC therapy group was significantly lower than the control group at the 2nd and 4th week of monitoring (p2 = 0.048, p4 = 0.048), and also with more type I collagen on 2nd and the 4th week (p2 = 0.048, p4 = 0.048). CONCLUSION: ADSC therapy can reduce the concentration of TGF-ß1, type I collagen, and degree of fibrosis in the male Wistar BOO model.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Obstrucción del Cuello de la Vejiga Urinaria , Animales , Colágeno Tipo I/análisis , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibrosis/metabolismo , Fibrosis/terapia , Humanos , Masculino , Células Madre Mesenquimatosas , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Células Madre/patología , Factor de Crecimiento Transformador beta1/análisis , Factor de Crecimiento Transformador beta1/metabolismo , Vejiga Urinaria/patología , Obstrucción del Cuello de la Vejiga Urinaria/metabolismo , Obstrucción del Cuello de la Vejiga Urinaria/patología , Obstrucción del Cuello de la Vejiga Urinaria/terapia
8.
Science ; 375(6576): 91-96, 2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-34990237

RESUMEN

Fibrosis affects millions of people with cardiac disease. We developed a therapeutic approach to generate transient antifibrotic chimeric antigen receptor (CAR) T cells in vivo by delivering modified messenger RNA (mRNA) in T cell­targeted lipid nanoparticles (LNPs). The efficacy of these in vivo­reprogrammed CAR T cells was evaluated by injecting CD5-targeted LNPs into a mouse model of heart failure. Efficient delivery of modified mRNA encoding the CAR to T lymphocytes was observed, which produced transient, effective CAR T cells in vivo. Antifibrotic CAR T cells exhibited trogocytosis and retained the target antigen as they accumulated in the spleen. Treatment with modified mRNA-targeted LNPs reduced fibrosis and restored cardiac function after injury. In vivo generation of CAR T cells may hold promise as a therapeutic platform to treat various diseases.


Asunto(s)
Ingeniería Celular , Endopeptidasas/inmunología , Cardiopatías/terapia , Inmunoterapia Adoptiva , Liposomas , Proteínas de la Membrana/inmunología , Nanopartículas , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Antígenos CD5/inmunología , Endopeptidasas/metabolismo , Fibroblastos/inmunología , Fibroblastos/patología , Fibrosis/terapia , Células HEK293 , Cardiopatías/patología , Insuficiencia Cardíaca/terapia , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , ARN Mensajero/genética , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Bazo/inmunología , Trogocitosis
9.
J Histochem Cytochem ; 70(1): 53-81, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34751050

RESUMEN

Immune checkpoint inhibitors have become the mainstay of treatment for hepatocellular carcinoma (HCC). However, they are ineffective in some cases. Previous studies have reported that genetic alterations in oncogenic pathways such as Wnt/ß-catenin are the important triggers in HCC for primary refractoriness. T-cell exhaustion has been reported in various tumors and is likely to play a prominent role in the emergence of HCC due to chronic inflammation and cirrhosis-associated immune dysfunction. Immunosuppressive cells including regulatory T-cells and tumor-associated macrophages infiltrating the tumor are associated with hyperprogressive disease in the early stages of immune checkpoint inhibitor treatment. In addition, stellate cells and tumor-associated fibroblasts create an abundant desmoplastic environment by producing extracellular matrix. This strongly contributes to epithelial to mesenchymal transition via signaling activities including transforming growth factor beta, Wnt/ß-catenin, and Hippo pathway. The abundant desmoplastic environment has been demonstrated in pancreatic ductal adenocarcinoma and cholangiocarcinoma to suppress cytotoxic T-cell infiltration, PD-L1 expression, and neoantigen expression, resulting in a highly immunosuppressive niche. It is possible that a similar immunosuppressive environment is created in HCC with advanced fibrosis in the background liver. Although sufficient understanding is required for the establishment of immune therapies of HCC, further investigations are still required in this field.


Asunto(s)
Carcinoma Hepatocelular/terapia , Fibrosis/terapia , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia , Neoplasias Hepáticas/terapia , Nicho de Células Madre/efectos de los fármacos , Animales , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Fibrosis/inmunología , Fibrosis/patología , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Nicho de Células Madre/inmunología
10.
Exp Cell Res ; 411(1): 112983, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34921827

RESUMEN

After the severe initial insults of acute kidney injury, progressive kidney tubulointerstitial fibrosis may occur, the peritubular capillary (PTC) rarefaction plays a key role in the disease progression. However, the mechanisms of PTC damage were not fully understood and potential therapeutic interventions were not explored. Previous studies of our research team and others in this field suggested that bone marrow-derived mesenchymal stem cells (BMSCs) transplanted into the AKI rat model may preserve the kidney function and pathological changes. In the current study, with the ischemia/reperfusion AKI rat model, we revealed that BMSCs transplantation attenuated the renal function decrease in the AKI model through preserving the peritubular capillaries (PTCs) function. The density of PTCs is maintained by BMSCs transplantation in the AKI model, detachment and relocation of pericytes in the PTCs diminished. Then we established that BMSCs transplantation may attenuate the renal fibrosis and preserve the kidney function after AKI by repairing the PTCs. Improving the vitality of pericytes, suppressing the detachment and trans-differentiation of pericytes, directly differentiation of BMSCs into pericytes by BMSCs transplantation all participate in the PTC repair. Through these processes, BMSCs rescued the microvascular damage and improved the density of PTCs. As a result, a preliminary conclusion can be reached that BMSCs transplantation can be an effective therapy for delaying renal fibrosis after AKI.


Asunto(s)
Lesión Renal Aguda/complicaciones , Endotelio Vascular/citología , Fibrosis/terapia , Enfermedades Renales/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Pericitos/citología , Animales , Fibrosis/etiología , Fibrosis/metabolismo , Fibrosis/patología , Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Masculino , Ratas , Ratas Sprague-Dawley
11.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-34884805

RESUMEN

Excessive cardiac fibrosis plays a crucial role in almost all types of heart disease. Generally, cardiac fibrosis is a scarring process triggered in response to stress, injury, or aging and is characterized by the accumulation of activated myofibroblasts that deposit high levels of extracellular matrix proteins in the myocardium. While it is beneficial for cardiac repair in the short term, it can also result in pathological remodeling, tissue stiffening, and cardiac dysfunction, contributing to the progression of heart failure, arrhythmia, and sudden cardiac death. Despite its high prevalence, there is a lack of effective and safe therapies that specifically target myofibroblasts to inhibit or even reverse pathological cardiac fibrosis. In the past few decades, cell therapy has been under continuous evaluation as a potential treatment strategy, and several studies have shown that transplantation of mesenchymal stromal cells (MSCs) can reduce cardiac fibrosis and improve heart function. Mechanistically, it is believed that the heart benefits from MSC therapy by stimulating innate anti-fibrotic and regenerative reactions. The mechanisms of action include paracrine signaling and cell-to-cell interactions. In this review, we provide an overview of the anti-fibrotic properties of MSCs and approaches to enhance them and discuss future directions of MSCs for the treatment of cardiac fibrosis.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Fibrosis/terapia , Cardiopatías/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Humanos , Células Madre Mesenquimatosas/citología , Miocardio/patología , Miofibroblastos/metabolismo , Medicina Regenerativa/métodos
12.
Biomolecules ; 11(10)2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34680180

RESUMEN

This study aimed to analyze the effects of fibrin constructs enhanced with laminin-nidogen, implanted in the wounded rat soft palate. Fibrin constructs with and without laminin-nidogen were implanted in 1 mm excisional wounds in the soft palate of 9-week-old rats and compared with the wounded soft palate without implantation. Collagen deposition and myofiber formation were analyzed at days 3, 7, 28 and 56 after wounding by histochemistry. In addition, immune staining was performed for a-smooth muscle actin (a-SMA), myosin heavy chain (MyHC) and paired homeobox protein 7 (Pax7). At day 56, collagen areas were smaller in both implant groups (31.25 ± 7.73% fibrin only and 21.11 ± 6.06% fibrin with laminin-nidogen)) compared to the empty wounds (38.25 ± 8.89%, p < 0.05). Moreover, the collagen area in the fibrin with laminin-nidogen group was smaller than in the fibrin only group (p ˂ 0.05). The areas of myofiber formation in the fibrin only group (31.77 ± 10.81%) and fibrin with laminin-nidogen group (43.13 ± 10.39%) were larger than in the empty wounds (28.10 ± 11.68%, p ˂ 0.05). Fibrin-based constructs with laminin-nidogen reduce fibrosis and improve muscle regeneration in the wounded soft palate. This is a promising strategy to enhance cleft soft palate repair and other severe muscle injuries.


Asunto(s)
Fibrina/genética , Fibrosis/genética , Paladar Blando/lesiones , Cicatrización de Heridas/genética , Actinas/genética , Animales , Colágeno/genética , Fibrina/farmacología , Fibrosis/patología , Fibrosis/terapia , Humanos , Laminina/genética , Laminina/farmacología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/farmacología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/crecimiento & desarrollo , Miofibrillas/genética , Cadenas Pesadas de Miosina/genética , Factores de Transcripción Paired Box/genética , Paladar Blando/efectos de los fármacos , Paladar Blando/patología , Ratas , Regeneración/genética
13.
Nat Genet ; 53(12): 1649-1663, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34635846

RESUMEN

Initiation of cyst formation in autosomal dominant polycystic kidney disease (ADPKD) occurs when kidney tubule cells are rendered null for either PKD1 or PKD2 by somatic 'second hit' mutations. Subsequent cyst progression remodels the organ through changes in tubule cell shape, proliferation and secretion. The kidney develops inflammation and fibrosis. We constructed a mouse model in which adult inactivation of either Pkd gene can be followed by reactivation of the gene at a later time. Using this model, we show that re-expression of Pkd genes in cystic kidneys results in rapid reversal of ADPKD. Cyst cell proliferation is reduced, autophagy is activated and cystic tubules with expanded lumina lined by squamoid cells revert to normal lumina lined by cuboidal cells. Increases in inflammation, extracellular matrix deposition and myofibroblast activation are reversed, and the kidneys become smaller. We conclude that phenotypic features of ADPKD are reversible and that the kidney has an unexpected capacity for plasticity controlled at least in part by ADPKD gene function.


Asunto(s)
Adaptación Fisiológica , Riñón/fisiología , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/terapia , Animales , Plasticidad de la Célula , Femenino , Fibrosis/terapia , Silenciador del Gen , Terapia Genética , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades Renales Poliquísticas/patología , Proteína Quinasa C/genética , Canales Catiónicos TRPP/genética , Activación Transcripcional
14.
Int J Mol Sci ; 22(19)2021 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-34639055

RESUMEN

Organ fibrosis often ends in eventual organ failure and leads to high mortality. Although researchers have identified many effector cells and molecular pathways, there are few effective therapies for fibrosis to date and the underlying mechanism needs to be examined and defined further. Epoxyeicosatrienoic acids (EETs) are endogenous lipid metabolites of arachidonic acid (ARA) synthesized by cytochrome P450 (CYP) epoxygenases. EETs are rapidly metabolized primarily via the soluble epoxide hydrolase (sEH) pathway. The sEH pathway produces dihydroxyeicosatrienoic acids (DHETs), which have lower activity. Stabilized or increased EETs levels exert several protective effects, including pro-angiogenesis, anti-inflammation, anti-apoptosis, and anti-senescence. Currently, intensive investigations are being carried out on their anti-fibrotic effects in the kidney, heart, lung, and liver. The present review provides an update on how the stabilized or increased production of EETs is a reasonable theoretical basis for fibrosis treatment.


Asunto(s)
Susceptibilidad a Enfermedades , Eicosanoides/efectos adversos , Fibrosis/etiología , Animales , Ácido Araquidónico/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Manejo de la Enfermedad , Eicosanoides/metabolismo , Fibrosis/metabolismo , Fibrosis/patología , Fibrosis/terapia , Humanos , Redes y Vías Metabólicas , Especificidad de Órganos
15.
Medicine (Baltimore) ; 100(35): e26774, 2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34477116

RESUMEN

ABSTRACT: Umbilical Vein Recanalization (UVR) may occur in patients with long-standing portal hypertension and liver cirrhosis. This study aimed to investigate the clinical significance of UVR.Medical records of a cohort of patients with cirrhosis (n = 247) who were hospitalized at the Digestive Medicine Center of the Second Affiliated Hospital of Nanchang University from January 2012 to October 2015 were accessed. The UVR diagnosis was made by ultrasound examination and was confirmed by computerized tomography scan.The UVR incidence was 20.2% (50/247) in the cohort. The size of UVR was 9.9 ±â€Š4.7 mm (range: 5-26.5 mm) in diameter. The UVR and non-UVR groups showed no difference in grades of hepatic encephalopathy (P = .496), Child-Pugh classification (P = .401), the incidence of moderately severe ascites (26% vs 26%, P = 1), the esophageal variceal bleeding rate (32% vs 39%, P = .402), or portal vein thrombosis (8% vs 12%, P = .580). However, the incidence of cavernous transformation of the portal vein was statistically different, that there was 0 case in the UVR group and 8 cases in the non-UVR group (P < .05).Our results suggested that UVR had little impact on the clinical manifestations of patients with liver cirrhosis, the significance of UVR as an intervention method requires further studies.


Asunto(s)
Cateterismo/estadística & datos numéricos , Fibrosis/fisiopatología , Venas Umbilicales , Adulto , Cateterismo/métodos , Femenino , Fibrosis/clasificación , Fibrosis/terapia , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Tomografía Computarizada por Rayos X/métodos
16.
Biomolecules ; 11(8)2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34439762

RESUMEN

Myofibroblasts are contractile, α-smooth muscle actin-positive cells with multiple roles in pathophysiological processes. Myofibroblasts mediate wound contractions, but their persistent presence in tissues is central to driving fibrosis, making them attractive cell targets for the development of therapeutic treatments. However, due to shared cellular markers with several other phenotypes, the specific targeting of myofibroblasts has long presented a scientific and clinical challenge. In recent years, myofibroblasts have drawn much attention among scientific research communities from multiple disciplines and specialisations. As further research uncovers the characterisations of myofibroblast formation, function, and regulation, the realisation of novel interventional routes for myofibroblasts within pathologies has emerged. The research community is approaching the means to finally target these cells, to prevent fibrosis, accelerate scarless wound healing, and attenuate associated disease-processes in clinical settings. This comprehensive review article describes the myofibroblast cell phenotype, their origins, and their diverse physiological and pathological functionality. Special attention has been given to mechanisms and molecular pathways governing myofibroblast differentiation, and updates in molecular interventions.


Asunto(s)
Fibrosis/terapia , Miofibroblastos/metabolismo , Piel/patología , Actinas/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Cicatriz , Receptores ErbB/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Ligandos , Ratones , Fenotipo , Transducción de Señal/fisiología , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Wnt/metabolismo , Cicatrización de Heridas , beta Catenina/metabolismo
17.
Int J Mol Sci ; 22(14)2021 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-34299066

RESUMEN

Stem cell therapy is a promising alternative approach to heart diseases. The most prevalent source of multipotent stem cells, usually called somatic or adult stem cells (mesenchymal stromal/stem cells, MSCs) used in clinical trials is bone marrow (BM-MSCs), adipose tissue (AT-MSCs), umbilical cord (UC-MSCs) and placenta. Therapeutic use of MSCs in cardiovascular diseases is based on the benefits in reducing cardiac fibrosis and inflammation that compose the cardiac remodeling responsible for the maintenance of normal function, something which may end up causing progressive and irreversible dysfunction. Many factors lead to cardiac fibrosis and failure, and an effective therapy is lacking to reverse or attenuate this condition. Different approaches have been shown to be promising in surpassing the poor survival of transplanted cells in cardiac tissue to provide cardioprotection and prevent cardiac remodeling. This review includes the description of pre-clinical and clinical investigation of the therapeutic potential of MSCs in improving ventricular dysfunction consequent to diverse cardiac diseases.


Asunto(s)
Fibrosis/terapia , Cardiopatías/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Animales , Humanos
18.
Int J Mol Sci ; 22(11)2021 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-34205870

RESUMEN

Diabetic patients are predisposed to diabetic cardiomyopathy, a specific form of cardiomyopathy which is characterized by the development of myocardial fibrosis, cardiomyocyte hypertrophy, and apoptosis that develops independently of concomitant macrovascular and microvascular diabetic complications. Its pathophysiology is multifactorial and poorly understood and no specific therapeutic guideline has yet been established. Diabetic cardiomyopathy is a challenging diagnosis, made after excluding other potential entities, treated with different pharmacotherapeutic agents targeting various pathophysiological pathways that need yet to be unraveled. It has great clinical importance as diabetes is a disease with pandemic proportions. This review focuses on the potential mechanisms contributing to this entity, diagnostic options, as well as on potential therapeutic interventions taking in consideration their clinical feasibility and limitations in everyday practice. Besides conventional therapies, we discuss novel therapeutic possibilities that have not yet been translated into clinical practice.


Asunto(s)
Diabetes Mellitus/fisiopatología , Cardiomiopatías Diabéticas/fisiopatología , Fibrosis/fisiopatología , Miocardio/patología , Apoptosis/genética , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/terapia , Cardiomiopatías Diabéticas/diagnóstico , Cardiomiopatías Diabéticas/terapia , Fibrosis/diagnóstico , Fibrosis/terapia , Humanos
19.
Life Sci ; 281: 119756, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34175316

RESUMEN

Duchenne Muscular Dystrophy (DMD) is caused by mutations in the dystrophin gene, accompanied by aberrant extracellular matrix synthesis and muscle damage. ADAMTS1 metalloproteinase was reported increased in dystrophin-deficient mdx mouse. The aim of this study was to explore the role of ADAMTS1 in muscle function, fibrosis and damage, and respiratory function of mdx mice. 102 DMD patients and their mothers were included in this study. Multiplex ligation dependent probe amplification (MLPA) assay and Next-generation sequencing (NGS) were adopted to do genetic diagnosis. Dystrophin-deficient mdx mice were treated with anti-ADAMTS1 antibody (anti-ADAMTS1) for three weeks. The results showed that ADAMTS1 was increased in gastrocnemius muscle of mdx mice and serum of DMD patients. Anti-ADAMTS1 treatment increased Versican transcription but suppressed versican protein expression. Besides, we found anti-ADAMTS1 improved muscle strength, diaphragm and extensor digitorum longus muscles functions in mdx mice. Meanwhile, muscle fibrosis and damage were attenuated in anti-ADAMTS1 treated dystrophic mice. In summary, anti-ADAMTS1 antibody relieved muscle dysfunction and fibrosis in dystrophic mice. It is suggested that ADAMTS1 is a potential target for developing new biological therapies for DMD.


Asunto(s)
Proteína ADAMTS1/antagonistas & inhibidores , Anticuerpos Monoclonales/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/terapia , Proteína ADAMTS1/genética , Proteína ADAMTS1/inmunología , Proteína ADAMTS1/metabolismo , Animales , Modelos Animales de Enfermedad , Distrofina/genética , Fibrosis/terapia , Humanos , Masculino , Ratones , Ratones Endogámicos mdx , Proteínas Musculares/metabolismo , Fuerza Muscular/inmunología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/genética , ARN Mensajero/metabolismo , Versicanos/inmunología
20.
J Nanobiotechnology ; 19(1): 156, 2021 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-34039349

RESUMEN

BACKGROUND: Despite the dramatic advances in modern medicine, efficient therapeutic measures for renal fibrosis remain limited. Celastrol (CLT) is effective in treating renal fibrosis in rat models, while causing severe systemic toxicity. Thus, we designed a tubule-specific nanocage (K3-HBc NCs) that effectively deliver CLT to tubular epithelial cell in a virus-like manner. The targeting ligand (K3) to tubular epithelial cells was displayed on the surface of Hepatitis B core protein (HBc) NCs by genetic fusion to the major immunodominant loop region. Ultra-small CLT nanodots were subtly encapsulated into the cavity through electrostatic interaction with the disassembly and reassembly of K3-HBc NCs, to yield K3-HBc/CLT complex. The efficacy of K3-HBc/CLT NCs were demonstrated in Unilateral ureteral obstruction (UUO)-induced renal fibrosis. RESULTS: The self-assembled K3-HBc/CLT could specifically target tubular epithelial cells via affinity with K3 ligand binding to the megalin receptor, significantly attenuating renal fibrosis. Remarkably, K3-HBc/CLT NCs significantly increased therapeutic efficacy and reduced the systemic toxicity in comparison with free CLT in UUO-induced mouse renal fibrosis model. Importantly, analysis of RNA sequencing data suggested that the anti-fibrotic effect of K3-HBc/CLT could be attributed to suppression of premature senescence in tubular epithelial cells via p21Cip1 and p16Ink4a pathway. CONCLUSION: The tubule-specific K3-HBc/CLT represented a promising option to realize precise treatment for renal fibrosis.


Asunto(s)
Fibrosis/terapia , Túbulos Renales/patología , Riñón/metabolismo , Riñón/patología , Animales , Apoptosis , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales , Masculino , Ratones , Ratones Endogámicos BALB C , Triterpenos Pentacíclicos , Ratas , Ratas Sprague-Dawley , Obstrucción Ureteral/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...