RESUMEN
Although many studies have addressed the regulatory circuits affecting neuronal activities, local non-synaptic mechanisms that determine neuronal excitability remain unclear. Here, we found that microglia prevented overactivation of pre-sympathetic neurons in the hypothalamic paraventricular nucleus (PVN) at steady state. Microglia constitutively released platelet-derived growth factor (PDGF) B, which signaled via PDGFRα on neuronal cells and promoted their expression of Kv4.3, a key subunit that conducts potassium currents. Ablation of microglia, conditional deletion of microglial PDGFB, or suppression of neuronal PDGFRα expression in the PVN elevated the excitability of pre-sympathetic neurons and sympathetic outflow, resulting in a profound autonomic dysfunction. Disruption of the PDGFBMG-Kv4.3Neuron pathway predisposed mice to develop hypertension, whereas central supplementation of exogenous PDGFB suppressed pressor response when mice were under hypertensive insult. Our results point to a non-immune action of resident microglia in maintaining the balance of sympathetic outflow, which is important in preventing cardiovascular diseases.
Asunto(s)
Hipertensión , Microglía , Animales , Hipertensión/metabolismo , Ratones , Neuronas/fisiología , Potasio/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismoRESUMEN
Immunotherapy is a promising cancer therapeutic strategy. However, the "cold" tumor immune microenvironment (TIME), characterized by insufficient immune cell infiltration and immunosuppressive status, limits the efficacy of immunotherapy. Tumor vascular abnormalities due to defective pericyte coverage are gradually recognized as a profound determinant in "cold" TIME establishment by hindering immune cell trafficking. Recently, several vascular normalization strategies by improving pericyte coverage have been reported, whereas have unsatisfactory efficacy and high rates of resistance. Herein, a combinatorial strategy to induce tumor vasculature-targeted pericyte recruitment and zinc ion-mediated immune activation with a platelet-derived growth factor B (PDGFB)-loaded, cyclo (Arg-Gly-Asp-D-Phe-Lys)-modified zeolitic imidazolate framework 8 (PDGFB@ZIF8-RGD) nanoplatform is proposed. PDGFB@ZIF8-RGD effectively induced tumor vascular normalization, which facilitated trafficking and infiltration of immune effector cells, including natural killer (NK) cells, M1-like macrophages and CD8+ T cells, into tumor microenvironment. Simultaneously, vascular normalization promoted the accumulation of zinc ions inside tumors to trigger effector cell immune activation and effector molecule production. The synergy between these two effects endowed PDGFB@ZIF8-RGD with superior capabilities in reprogramming the "cold" TIME to a "hot" TIME, thereby initiating robust antitumor immunity and suppressing tumor growth. This combinatorial strategy for improving immune effector cell infiltration and activation is a promising paradigm for solid tumor immunotherapy.
Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Proteínas Proto-Oncogénicas c-sis/farmacología , Proteínas Proto-Oncogénicas c-sis/uso terapéutico , Neoplasias/terapia , Inmunoterapia , Oligopéptidos/uso terapéutico , Zinc/farmacología , Microambiente TumoralRESUMEN
Although advances in the prediction and management of ovarian hyperstimulation syndrome (OHSS) have been introduced, complete prevention is not yet possible. Previously, we and other authors have shown that vascular endothelial growth factor, angiopoietins (ANGPTs) and sphingosine-1-phosphate are involved in OHSS etiology. In addition, we have demonstrated that ovarian protein levels of platelet-derived growth factor (PDGF) ligands -B and -D decrease in an OHSS rat model, whilst PDGFR-ß and ANGPT2 remain unchanged. In the present work, we investigated the role of PDGF-B in OHSS by evaluating ligand protein levels in follicular fluid (FF) from women at risk of developing OHSS and by using an immature rat model of OHSS. We demonstrated that PDGF-B and PDGF-D are lower in FF from women at risk of developing OHSS compared to control patients (P < 0.05). In the OHSS rat model, PDGF-B (0.5 µg/ovary) administration decreased ovarian weight (P < 0.05), reduced serum progesterone (P < 0.05) and lowered the percentage of cysts (P < 0.05), compared to untreated OHSS rats, but had no effect on the proportion of follicles or corpora lutea (CL). PDGF-B treatment also restored the expression of steroidogenic acute regulatory protein (P < 0.05) and P450 cholesterol side-chain cleavage enzyme (P < 0.01) to control levels. In addition, PDGF-B increased the peri-endothelial cell area in CL and cystic structures, and reduced vascular permeability compared to untreated OHSS ovaries. Lastly, PDGF-B increased the levels of junction proteins claudin-5 (P < 0.05), occludin (P < 0.05) and ß-catenin (P < 0.05), while boosting the extracellular deposition of collagen IV surrounding the ovarian vasculature (PP < 0.01), compared to OHSS alone. In conclusion, our findings indicate that PDGF-B could be another crucial mediator in the onset and development of OHSS, which may lead to the development of novel prediction markers and therapeutic strategies.
Asunto(s)
Síndrome de Hiperestimulación Ovárica/tratamiento farmacológico , Síndrome de Hiperestimulación Ovárica/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , Proteínas Proto-Oncogénicas c-sis/uso terapéutico , Adulto , Animales , Western Blotting , Femenino , Humanos , Inmunohistoquímica , Ratas Sprague-DawleyRESUMEN
RESEARCH QUESTION: Endometriosis, an inflammatory disease, is assumed to be associated with an increased production of growth-related cytokines. Based on the emerging immunomodulatory role of vitamin D3 in different inflammatory conditions, this study aimed to examine its modulatory effect on the expression levels of the genes for platelet-derived growth factor-B (PDGFB), monocyte/macrophage-derived growth factor (MDGF, also known as PPBP) and epidermal growth factor (EGF) in peritoneal fluid mononuclear cells (PFMC) in women with and without endometriosis. DESIGN: PFMC from 10 women with endometriosis and 10 control participants were treated with vitamin D3.The gene expression levels of PDGFB, MDGF and EGF were measured 6, 24 and 48 h following vitamin D3 administration using real-time PCR. RESULTS: Gene expression levels of EGF and PDGFB were higher in the PFMC of women with endometriosis than the control group (Pâ¯=â¯0.006, P < 0.001, respectively). Although MDGF expression showed an increase in the endometriosis group compared with non-endometriotic controls, no significant difference was found. Vitamin D3 significantly decreased EGF expression at 6, 24 and 48 h (P < 0.001, P < 0.001 and Pâ¯=â¯0.007, respectively), MDGF at 24 and 48 h (P < 0.001 and Pâ¯=â¯0.009, respectively) and PDGFB at 6 h (Pâ¯=â¯0.047) in the endometriosis group. Vitamin D3 treatment had no significant effect on expression of the genes in the PFMC of non-endometriotic women. CONCLUSIONS: The study concluded that PDGFB and EGF gene expression increases in endometriosis, and vitamin D3 could markedly decrease this expression, suggesting its therapeutic potential in endometriosis.
Asunto(s)
Colecalciferol/farmacología , Endometriosis/genética , Factor de Crecimiento Epidérmico/genética , Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/genética , Leucocitos Mononucleares/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis/genética , Adulto , Líquido Ascítico/efectos de los fármacos , Líquido Ascítico/metabolismo , Endometriosis/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leucocitos Mononucleares/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Adulto JovenRESUMEN
Platelet-derived growth factor (PDGF) is a family of growth factors with mitogenic and chemotactic activity. However, uncontrolled and overactivated PDGF signaling has been implicated in a variety of diseases, such as cancers and atherosclerosis. In this context, inhibition of PDGF-PDGFR signaling is of paramount importance in progression of such diseases. The purpose of the current study was to identify novel PDGF-B inhibitors using virtual screening methods. To this end, a combination of molecular modeling techniques such as molecular docking and dynamics simulation, as well as drug likeness filtering criteria, was applied to select anti-PDGF peptidomimetic candidates based on crystallography solved structure of an anti-PDGF-B monoclonal antibody named, MOR8457. In vitro biological assays of the selected compounds revealed two of them being active at micromolar IC50 concentrations. The presented work can provide a framework for systematic peptidomimetic identification for anti-PDGF-B agents from large chemical libraries.
Asunto(s)
Anticuerpos Monoclonales/farmacología , Descubrimiento de Drogas , Proteínas Proto-Oncogénicas c-sis/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Anticuerpos Monoclonales/química , Células Cultivadas , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-ActividadRESUMEN
Cellular responses to platelet-derived growth factor (PDGF) are altered in a variety of pathological conditions, including cancers, fibroses, and vascular diseases, making PDGF-induced signaling pathways important therapeutic targets. The limited success of therapies designed to impact PDGF pathways may be overcome with a clearer understanding of how cells integrate signals from PDGF and the extracellular matrix (ECM). Here, we assessed the effects of fibronectin matrix assembly on the responsiveness of mesenchymal cells to PDGF. Our results indicate that fibroblast-mediated assembly of fibronectin fibrils attenuates intracellular calcium release in response to PDGF. The dose-dependent inhibition of PDGF-induced intracellular calcium release was specific to the ECM form of fibronectin. Further, a recombinant protein engineered to mimic ECM fibronectin similarly attenuated intracellular calcium release in response to PDGF. Of note, fibronectin attenuated the PDGF-calcium signaling axis at the level of phosphoinositide 3-kinase (PI3K) activation. Interestingly, ECM fibronectin did not alter other intracellular signals activated by PDGF, including activation of PDGF receptor ß, AKT Ser/Thr kinase, phospholipase Cγ1, and extracellular signal-regulated kinase 1/2 (ERK1/2). Rather, fibronectin inhibited activation of the p55 regulatory subunit of PI3K in response to a variety of stimuli, indicating that ECM fibronectin selectively attenuates the intracellular calcium release cascade while leaving intact other PDGF signaling pathways. Selective regulation of calcium signaling by ECM fibronectin via the p55 regulatory subunit of PI3K represents a mechanism by which cells tune their response to PDGF and may therefore serve as a target to selectively regulate one branch of PDGF signaling.
Asunto(s)
Calcio/metabolismo , Fibronectinas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Activación Enzimática , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Especificidad por SustratoRESUMEN
OBJECTIVE: Unruptured intracranial aneurysms (UIAs) are relatively common lesions that may cause devastating intracranial hemorrhage, thus producing considerable suffering and anxiety in those affected by the disease or an increased likelihood of developing it. Advances in the knowledge of the pathobiology behind intracranial aneurysm (IA) formation, progression, and rupture have led to preclinical testing of drug therapies that would prevent IA formation or progression. In parallel, novel biologically based diagnostic tools to estimate rupture risk are approaching clinical use. Arterial wall remodeling, triggered by flow and intramural stresses and mediated by inflammation, is relevant to both. METHODS: This review discusses the basis of flow-driven vessel remodeling and translates that knowledge to the observations made on the mechanisms of IA initiation and progression on studies using animal models of induced IA formation, study of human IA tissue samples, and study of patient-derived computational fluid dynamics models. RESULTS: Blood flow conditions leading to high wall shear stress (WSS) activate proinflammatory signaling in endothelial cells that recruits macrophages to the site exposed to high WSS, especially through macrophage chemoattractant protein 1 (MCP1). This macrophage infiltration leads to protease expression, which disrupts the internal elastic lamina and collagen matrix, leading to focal outward bulging of the wall and IA initiation. For the IA to grow, collagen remodeling and smooth muscle cell (SMC) proliferation are essential, because the fact that collagen does not distend much prevents the passive dilation of a focal weakness to a sizable IA. Chronic macrophage infiltration of the IA wall promotes this SMC-mediated growth and is a potential target for drug therapy. Once the IA wall grows, it is subjected to changes in wall tension and flow conditions as a result of the change in geometry and has to remodel accordingly to avoid rupture. Flow affects this remodeling process. CONCLUSIONS: Flow triggers an inflammatory reaction that predisposes the arterial wall to IA initiation and growth and affects the associated remodeling of the UIA wall. This chronic inflammation is a putative target for drug therapy that would stabilize UIAs or prevent UIA formation. Moreover, once this coupling between IA wall remodeling and flow is understood, data from patient-specific flow models can be gathered as part of the diagnostic workup and utilized to improve risk assessment for UIA initiation, progression, and eventual rupture.
Asunto(s)
Arterias Cerebrales/patología , Circulación Cerebrovascular , Inflamación/patología , Aneurisma Intracraneal/patología , Humanos , Hidrodinámica , Inflamación/complicaciones , Aneurisma Intracraneal/etiología , Modelos Biológicos , Estrés FisiológicoRESUMEN
DNA binding protein A (DbpA) is a member of the human cold shock domain-containing protein superfamily, with known functions in cell proliferation, differentiation, and stress responses. DbpA mediates tight junction-associated activities in tubular epithelial cells, but the function of DbpA in mesangial cells is unknown. Here, we found DbpA protein expression restricted to vascular smooth muscle cells in healthy human kidney tissue but profound induction of DbpA protein expression within the glomerular mesangial compartment in mesangioproliferative nephritis. In vitro, depletion or overexpression of DbpA using lentiviral constructs led to inhibition or promotion, respectively, of mesangial cell proliferation. Because platelet-derived growth factor B (PDGF-B) signaling has a pivotal role in mesangial cell proliferation, we examined the regulatory effect of PDGF-B on DbpA. In vitro studies of human and rat mesangial cells confirmed a stimulatory effect of PDGF-B on DbpA transcript numbers and protein levels. Additional in vivo investigations showed DbpA upregulation in experimental rat anti-Thy1.1 nephritis and murine mesangioproliferative nephritis models. To interfere with PDGF-B signaling, we injected nephritic rats with PDGF-B neutralizing aptamers or the MEK/ERK inhibitor U0126. Both interventions markedly decreased DbpA protein expression. Conversely, continuous PDGF-B infusion in healthy rats induced DbpA expression predominantly within the mesangial compartment. Taken together, these results indicate that DbpA is a novel target of PDGF-B signaling and a key mediator of mesangial cell proliferation.
Asunto(s)
Proteínas y Péptidos de Choque por Frío/fisiología , Proteínas de Unión al ADN/fisiología , Mesangio Glomerular/patología , Mesangio Glomerular/fisiología , Glomerulonefritis/etiología , Células Mesangiales/patología , Animales , Proliferación Celular , Células Cultivadas , Humanos , Nefritis Lúpica/etiología , RatasRESUMEN
Mammary epithelial cells (MECs) play an important role in immune responses and inflammatory diseases such as mastitis, which is mainly attributed to the activation of Toll-like receptors and the release of cytokines. However, the overall change of gene expression and biological pathways of MECs to microbial factors stimulation remains unknown. Here, we analyzed the gene expression profile in mouse MECs treated with lipopolysaccharide (LPS) for 24 h. Microarray analysis revealed that about 1548 genes differentially expressed, these genes mainly involved in 346 gene ontology terms and 128 molecular pathways, and particularly, some innate immune-associated pathways were significant. By analyzing data for pathway relation network, we prioritized differentially expressed genes with respect to LPS. The importance of changes, indicating that RNA interference-mediated inhibition of two genes identified in this analysis, transforming growth factor beta 1 (Tgf-ß1) and platelet-derived growth factor B (Pdgfb), reduced interleukin (IL)-6 and tumor necrosis factor α production respectively, in gene expression was verified. These findings delineate mouse MECs gene response patterns induced by LPS and identify Tgf-ß1 and Pdgfb that have been closely related to innate immunity.
RESUMEN
The purpose of this study was to elucidate the transfection of chitosan nanoparticle carrying small interfering RNA against platelet-derived growth factor B (PDGF-B) to inhibit the expression of PDGF-B mRNA and proliferation of smooth muscle cells. A rabbit iliac artery injury model was constructed. A small interfering RNA (siRNA) against PDGF-B mRNA expression vector was constructed and packaged by chitosan nanoparticle to transfect into the vascular smooth muscle cells (vSMCs) of balloon catheter-injured rabbit iliac artery wall, using a therapeutic ultrasound for the gene delivery. The experiment was divided into two groups: experimental group, denudation and nano-PDGF-B siRNA treated, and only single denudation as control. Effects of the siRNA on the expressions of proliferating cell nuclear antigen (PCNA) and PDGF-B mRNA by vSMCs and the proliferation of vSMCs were observed with the methods of routine pathological, immunohistochemical staining, in situ hybridization and morphometry. The nano siRNA against PDGF-B was successfully transfected. The nano siRNA significantly inhibited the expressions of PCNA and PDGF-B mRNA in intimal vSMCs. The local intimal thickness and area were also reduced remarkably. In conclusion, transfection of chitosan nanoparticle carrying siRNA against PDGF-B mRNA could inhibit proliferation of vSMCs in the rabbit iliac artery injury model.
Asunto(s)
Proliferación Celular , Quitosano/química , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Nanopartículas , Proteínas Proto-Oncogénicas c-sis/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Tratamiento con ARN de Interferencia/métodos , Transfección/métodos , Lesiones del Sistema Vascular/terapia , Angioplastia de Balón , Animales , Modelos Animales de Enfermedad , Arteria Ilíaca/lesiones , Arteria Ilíaca/metabolismo , Arteria Ilíaca/patología , Masculino , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , ARN Interferente Pequeño/química , ARN Interferente Pequeño/metabolismo , Conejos , Ultrasonido , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patologíaRESUMEN
AIM OF THE STUDY: Platelet-derived growth factor B (PDGF-B), a vital growth factor which can induce angiogenesis and epithelial-mesenchymal transition (EMT), is important in the metastasis of many tumors. However, the roles of PDGF-B in gastric carcinoma are largely unknown. We investigated the correlation between PDGF-B, PDGFR-ß and E-cadherin expression with the clinical features of gastric carcinoma patients to evaluate the relationship between PDGF-B signaling, E-cadherin and metastasis of gastric carcinoma, the correlation between PDGF-B and E-cadherin expression to assess the roles of PDGF-B signaling in metastasis of gastric carcinoma.. MATERIAL AND METHODS: We detected expressions of PDGF-B, PDGFR-ß and E-cadherin in gastric carcinoma tissues and normal gastric mucosa tissues of 64 patients with gastric carcinoma who had undergone surgical resection, and investigated their relationships with clinical features and the relationships between PDGF-B and E-cadherin expression in gastric carcinoma. RESULTS: In surgical specimens, tumor cells expressed PDGF-B, and PDGFR-ß was expressed by tumor stromal cells. E-cadherin was expressed by both tumor cells and normal gastric mucosa cells. Expressions of PDGF-B and PDGFR-ß were increased in gastric carcinoma tissues (p < 0.05) and were positively correlated with the depth of cancer invasion, lymph node metastasis and TNM stage (p < 0.05). The expression of E-cadherin was reduced in gastric carcinoma tissues (p < 0.05) and was negatively correlated with the depth of cancer invasion, lymph node metastasis and TNM stage (p < 0.05). The correlation between PDGF-B and E-cadherin expression was negative (p < 0.05). CONCLUSION: Our data indicate that either the overexpression of PDGF-B and PDGFR-ß or the underexpression of E-cadherin is correlated with cancer progression and lymphogenous metastasis of gastric carcinoma. The PDGF-B signal pathway might induce EMT by down-regulating expression of E-cadherin to promote metastasis of gastric carcinoma.
RESUMEN
Debilitating abdominal pain is a common symptom affecting most patients with chronic pancreatitis (CP). There are multiple underlying mechanisms that contribute to CP-related pain, which makes successful treatment difficult. The identification of biomarkers for subtypes of pain could provide viable targets for nonopioid interventions and the development of mechanistic approaches to pain management in CP. Nineteen inflammation- and nociception-associated proteins were measured in serum collected from 358 subjects with definite CP enrolled in PROspective Evaluation of Chronic Pancreatitis for EpidEmiologic and Translational StuDies, a prospective observational study of pancreatitis in US adult subjects. First, serum levels of putative biomarkers were compared between CP subjects with and without pain. Only platelet-derived growth factor B (PDGF-B) stood out, with levels significantly higher in the CP pain group as compared to subjects with no pain. Subjects with pain were then stratified into 4 pain subtypes (Neuropathic, Nociceptive, Mixed, and Unclassified). A comparison of putative biomarker concentration among 5 groups (no pain and 4 pain subtypes) identified unique proteins that were correlated with pain subtypes. Serum transforming growth factor beta 1 (TGFß1) level was significantly higher in the Nociceptive pain group compared to the No pain group, suggesting that TGFß1 may be a biomarker for nociceptive pain. The Neuropathic pain only group was too small to detect statistical differences. However, glycoprotein 130 (GP130), a coreceptor for interleukin 6, was significantly higher in the Mixed pain group compared to the groups lacking a neuropathic pain component. These data suggest that GP130 may be a biomarker for neuropathic pain in CP. PERSPECTIVE: Serum TGFß1 and GP130 may be biomarkers for nociceptive and neuropathic CP pain, respectively. Preclinical data suggest inhibiting TGFß1 or GP130 reduces CP pain in rodent models, indicating that additional translational and clinical studies may be warranted to develop a precision medicine approach to the management of pain in CP.
Asunto(s)
Dolor Crónico , Neuralgia , Dolor Nociceptivo , Pancreatitis Crónica , Adulto , Humanos , Biomarcadores , Receptor gp130 de Citocinas , Neuralgia/diagnóstico , Neuralgia/etiología , Neuralgia/tratamiento farmacológico , Nocicepción , Pancreatitis Crónica/complicaciones , Pancreatitis Crónica/diagnósticoRESUMEN
Gynecological sarcomas are rare and their location in the vulva and vagina has an incidence of 5% of all malignant neoplasms in the female genital tract. We present the case of a 54-year-old patient with a diagnosis of dermatofibrosarcoma protuberans in the vulva, an infrequent pathology with less than 60 cases reported worldwide in this anatomical location. Clinically, it is locally aggressive, due to the proliferation of spindle cells with pleomorphism and frequent mitotic figures infiltrating the reticular dermis and subcutaneous cellular tissue, giving rise to variable size tumors with high local recurrence rates. The first-line treatment is surgical excision of the tumor with Mohs micrographic surgery among other surgical techniques for complete circumferential peripheral and deep margin assessment. However, identification of carcinogenesis mechanisms where the chromosomal translocation t (17; 22) (q22; q13) is recognized, forming the COL1A1-PDGFB fusion gene, which participates in stimulating tumor cell proliferation, allowing treatment with tyrosine kinase inhibitors such as imatinib for neoadjuvant therapy of surgically unresectable tumors and local recurrences.
Los sarcomas ginecológicos son infrecuentes y la localización de estos en vulva y vagina tienen una incidencia del 5% de todas las neoplasias malignas del tracto genital femenino. Presentamos una paciente de 54 años con diagnóstico de dermatofibrosarcoma protuberans en vulva, el cual es una patología infrecuente con menos de 60 casos reportados a nivel mundial en esta localización anatómica. Clínicamente tiene un comportamiento localmente agresivo, debido a la proliferación de células fusiformes con pleomorfismo y frecuentes figuras de mitosis que infiltran la dermis reticular y tejido celular subcutáneo, dando origen a lesiones tumorales de tamaño variable y con altas tasas de recurrencia local. El tratamiento en primera elección es la escisión quirúrgica del tumor con técnicas como cirugía micrográfica de Mohs u otras técnicas quirúrgicas para evaluación completa del margen periférico circunferencial y profundo. Sin embargo, la identificación de mecanismos de carcinogénesis donde se reconoce la translocación cromosómica t (17; 22) (q22; q13), formando al gen de fusión COL1A1-PDGFB el cual participa estimulando la proliferación celular tumoral, ha permitido la utilización de los inhibidores de la tirosina quinasa como el imatinib para la realización de terapia neoadyuvante en casos de tumores irresecables quirúrgicamente y en recurrencias locales.
Asunto(s)
Dermatofibrosarcoma , Neoplasias Cutáneas , Dermatofibrosarcoma/diagnóstico , Dermatofibrosarcoma/genética , Dermatofibrosarcoma/cirugía , Femenino , Humanos , Mesilato de Imatinib/uso terapéutico , Persona de Mediana Edad , Neoplasias Cutáneas/diagnóstico , Translocación Genética , Vulva/patologíaRESUMEN
We developed a composite wound dressing that provides multifunctional wound care. Alginate and polycaprolactone (PCL) were coelectrospun as composite fibers. Highly absorbent alginate provided a moist environment for wounds and PCL increased cell adhesion. Silver nanoparticles embedded in PCL fibers for long-term release inhibited the growth of microorganisms. In addition, plasmid DNA encoding platelet-derived growth factor-B (PDGF-B) were complexed with polyethylenimine (PEI) to form cationic nanoparticles which were then adsorbed on anionic alginate fibers through electrostatic interaction. As wound cells adhered to composite fibers, they were in situ transfected to express PDGF-B continuously. Moreover, calcium ions in alginate fibers were released into the wound site through ion exchange to accelerate hemostasis. Wound healing experiments demonstrated that PDGF-B gene-loaded composite fibers accelerated wound closure and promoted collagen formation. We expect this comprehensive study offers an ideal multifunctional solution to facilitate wound healing.
Asunto(s)
Alginatos , Nanopartículas del Metal , Alginatos/farmacología , Vendajes , Poliésteres , Plata/farmacologíaRESUMEN
BACKGROUND: Primary familial brain calcification (PFBC) is a neurodegenerative disease characterized with calcium deposition in multiple brain regions. Mutations in PDGFB have been discovered in sporadic and familial PFBC cases. While several known variants displayed loss-of function, no complete deletion of platelet-derived growth factor B (PDGFB) has been reported. METHODS: For the diagnostic purpose, brain computerized tomography or magnetic resonance imaging scanning and whole-genome sequencing were performed on the proband and family members in the pedigree. RESULTS: We identified a heterozygous PDGFB complete deletion in a Chinese pedigree. The proband presented with paroxysmal kinesigenic dyskinesia (PKD), a rare symptom in PFBC. The proband's mother carrying the same mutation was asymptomatic. CONCLUSIONS: For the first time, we reported a PFBC with a heterozygous deletion of PDGFB, and provided evidence of haploinsufficiency in the pathogenesis of PFBC.
Asunto(s)
Encefalopatías/genética , Encéfalo/patología , Distonía/genética , Eliminación de Gen , Proteínas Proto-Oncogénicas c-sis/genética , Adolescente , Encefalopatías/patología , Calcinosis/genética , Distonía/patología , Heterocigoto , Humanos , Masculino , Mutación , LinajeRESUMEN
Elevated circulating activity of adenosine deaminase 2 (ADA2) is associated with liver fibrosis in nonalcoholic fatty liver disease (NAFLD). In the liver of NAFLD patients, ADA2-positive portal macrophages are significantly associated with the degree of liver fibrosis. These liver macrophages are CD14- and CD16-positive and co-express chemokine receptors CCR2, CCR5, and CXCR3, indicating infiltrative monocyte origin. Human circulatory monocytes release ADA2 upon macrophage differentiation in vitro. When stimulated by recombinant human ADA2 (rhADA2), human monocyte-derived macrophages demonstrate upregulation of pro-inflammatory and pro-fibrotic genes, including PDGF-B, a key pro-fibrotic cytokine. This PDGF-B upregulation is reproduced by inosine, the enzymatic product of ADA2, but not adenosine, and is abolished by E359N, a loss-of-function mutation in ADA2. Finally, rhADA2 also stimulates PDGF-B production from Kupffer cells in primary human liver spheroids. Together, these data suggest that infiltrative monocytes promote fibrogenesis in NAFLD via ADA2-mediated autocrine/paracrine signaling culminating in enhanced PDGF-B production.
Asunto(s)
Adenosina Desaminasa/metabolismo , Comunicación Autocrina , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Macrófagos del Hígado/enzimología , Hígado/enzimología , Monocitos/enzimología , Enfermedad del Hígado Graso no Alcohólico/enzimología , Comunicación Paracrina , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-sis/metabolismoRESUMEN
BACKGROUND: Recent advances in surgical and neuroprotective strategies could effectively manage the pathophysiological progression of subarachnoid hemorrhage (SAH). However, pulmonary dysfunction frequently occurs in SAH patients with an increased risk of unsatisfactory outcomes. Based on the similar microvascular structures in the blood-air barrier and blood-brain barrier and possible brain-lung crosstalks, we believe that pericytes may be involved in both neurological and pulmonary dysfunction after SAH. METHODS: In our experiments, platelet-derived growth factor B (PDGF-B) retention motif knockout (PDGF-Bret/ret) mice and adeno-associated virus PDGF-B were employed to show the involvement of pericyte deficiency and PDGF-B expression. Neurological score, SAH grade, hematoxylin-eosin staining, and PaO2/FiO2 ratio analysis were performed to evaluate the neurological deficits and pulmonary functions in endovascular perforation SAH models at 24 h after surgery, as well as western blotting and immunofluorescence staining for underlying molecular expressions. RESULTS: We found that neonatal PDGF-Bret/ret mice exhibited pulmonary atelectasis 12 h after birth. Further investigation showed a decrease in PaO2/FiO2 and lung-specific surfactant proteins in adult PDGF-Bret/ret mice. These dysfunctions were much worse than those in wild-type mice at 24 h after SAH. PDGF-B overexpression alleviated pulmonary dysfunction after SAH. CONCLUSIONS: These results suggested pulmonary dysfunction after SAH and the pivotal role of PDGF-B signaling for the pathophysiological process and future therapeutic targets of pulmonary injury treatment after SAH. Further studies are needed for pathophysiological investigations and translational studies on pulmonary injuries after SAH.
RESUMEN
Sepsis is a systemic inflammatory response during infection and remains a major clinical problem with high morbidity and mortality. Platelet-derived growth factor B (PDGF-B) is a member belongs to PDGF family and has been recently reported higher expressed in survivors of severe sepsis patients. However, the exact role and underlying mechanisms of PDGF-BB in sepsis remains unclear. In this study, we found that PDGF-BB levels were significantly elevated in patients with sepsis, and higher PDGF-BB levels were negatively correlated with the levels of proinflammatory cytokines (TNF-α, IL-6, IL-1ß, IL-8), and chemokines (CXCL-1 and CCL2). PDGF-BB was also found increased in experimental sepsis in mice. Blockade of PDGF-BB using Tyrphostin AG 1296 aggravated, whereas recombinant PDGF-BB treatment improved survival and tissues injury in both two murine models of CLP-induced sepsis and LPS- induced endotoxemia. PDGF-BB blockade increased, whereas PDGF-BB administration decreased the inflammatory responses, as reflected by proinflammatory cytokines (TNF-α, IL-6, IL-1ß, IL-8), and chemokines (CXCL-1 and CCL2). PDGF-BB also showed inhibitory effect on immune cell activation and cytokines production in vivo and in vitro. Therefore, our findings suggest that PDGF-BB plays a protective role in sepsis by decreasing the production of pro-inflammatory cytokines and chemokines. PDGF-BB thus may be a potential therapeutic strategy for treating sepsis.
Asunto(s)
Antiinflamatorios/uso terapéutico , Citocinas/inmunología , Proteínas Proto-Oncogénicas c-sis/uso terapéutico , Sepsis/tratamiento farmacológico , Adulto , Animales , Antiinflamatorios/farmacología , Citocinas/sangre , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-sis/sangre , Proteínas Proto-Oncogénicas c-sis/farmacología , Proteínas Recombinantes/sangre , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Sepsis/sangre , Sepsis/inmunologíaRESUMEN
The glymphatic system is a highly polarized cerebrospinal fluid (CSF) transport system that facilitates the clearance of neurotoxic molecules through a brain-wide network of perivascular pathways. Herein we have mapped the development of the glymphatic system in mice. Perivascular CSF transport first emerges in hippocampus in newborn mice, and a mature glymphatic system is established in the cortex at 2 weeks of age. Formation of astrocytic endfeet and polarized expression of aquaporin 4 (AQP4) consistently coincided with the appearance of perivascular CSF transport. Deficiency of platelet-derived growth factor B (PDGF-B) function in the PDGF retention motif knockout mouse line Pdgfbret/ret suppressed the development of the glymphatic system, whose functions remained suppressed in adulthood compared with wild-type mice. These experiments map the natural development of the glymphatic system in mice and define a critical role of PDGF-B in the development of perivascular CSF transport.
Asunto(s)
Astrocitos/metabolismo , Sistema Glinfático/crecimiento & desarrollo , Linfocinas/genética , Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Acuaporina 4/genética , Acuaporina 4/metabolismo , Astrocitos/citología , Femenino , Sistema Glinfático/metabolismo , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Linfocinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transporte de ProteínasRESUMEN
BACKGROUND: Neurofibromas in von Recklinghausen's disease (vRD) can develop in the dermis. Therefore, we hypothesized that a dermal niche exists that promotes the development of these neurofibromas in subjects with vRD. OBJECTIVE: The purpose of this study is to examine the function of polydom, known as a ligand for integrin, mediating cell adhesion, and expressed in mouse nerve tissue, in promotion of neurofibroma. METHODS: Molecular, transcriptome and immunohistochemical analysis were performed to investigate the association between polydom expression and neurofibroma development. RESULTS: Polydom mRNA levels were significantly higher in neurofibroma tissue than in control tissue. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis of RNA purified from primary cultured dermal neurofibroma cells demonstrated significantly higher polydom mRNA expression in cells derived from the surrounding dermis of neurofibromas compared to those from normal human dermal fibroblasts. RNA sequencing was used to compare gene expression between cultured cells derived from dermal neurofibroma-surrounding tissue with or without polydom knockdown. Subsequent gene ontology assays revealed that expression of integrinß8 (ITGB8), a factor that releases transforming growth factor-ß (TGF-ß) from pro-TGF-ß, was downregulated following polydom knockdown, suggesting upregulation of polydom-mediated TGF-ß production. Furthermore, we observed a strong association between polydom expression and the increase in platelet-derived growth factor B (PDGFB) expression in primary cultured cells from the surrounding dermis of neurofibromas exposed to TGF-ß1. CONCLUSION: Our results suggest that increased polydom expression in the dermis surrounding neurofibromas may promote dermal neurofibroma development by activating the TGF-ß signaling pathway.