Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 693
Filtrar
1.
Adv Biol (Weinh) ; : e2400032, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39267218

RESUMEN

A wide range of cells respond to fibroblast growth factor 2 (FGF2) by proliferation via activation of the Ras/ERK1/2 pathway. In this study, the potential involvement of salt inducible kinase SIK2) in this cascade within retinal Müller glia is explored. It is found that SIK2 phosphorylation status and activity are modulated in an FGF2-dependent manner, possibly via ERK1/2. With SIK2 downregulation, enhanced ERK1/2 activation with delayed attenuation and increased cell proliferation is observed, while SIK2 overexpression hampers FGF2-dependent ERK1/2 activation. In vitro kinase and site-directed mutagenesis studies indicate that SIK2 targets the pathway element GRB2-associated-binding protein 1 (Gab1) on Ser266. This phosphorylation event weakens Gab1 interactions with its partners growth factor receptor-bound protein 2 (Grb2) and Src homology region 2 domain containing phosphatase 2 (Shp2). Collectively, these results suggest that during FGF2-dependent proliferation process ERK1/2-mediated activation of SIK2 targets Gab1, resulting in downregulation of the Ras/ERK1/2 cascade in a feedback loop.

2.
Cancers (Basel) ; 16(17)2024 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-39272961

RESUMEN

We showed previously that the autocrine activation of the FGFR-mediated pathway in GIST lacking secondary KIT mutations was a result of the inhibition of KIT signaling. We show here that the FGF2/FGFR pathway regulates VEGF-A/VEGFR signaling in IM-resistant GIST cells. Indeed, recombinant FGF2 increased the production of VEGF-A by IM-naive and resistant GIST cells. VEGF-A production was also increased in KIT-inhibited GIST, whereas the neutralization of FGF2 by anti-FGF2 mAb attenuated VEGFR signaling. Of note, BGJ 398, pan FGFR inhibitor, effectively and time-dependently inhibited VEGFR signaling in IM-resistant GIST T-1R cells, thereby revealing the regulatory role of the FGFR pathway in VEGFR signaling for this particular GIST cell line. This also resulted in significant synergy between BGJ 398 and VEGFR inhibitors (i.e., sunitinib and regorafenib) by enhancing their pro-apoptotic and anti-proliferative activities. The high potency of the combined use of VEGFR and FGFR inhibitors in IM-resistant GISTs was revealed by the impressive synergy scores observed for regorafenib or sunitinib and BGJ 398. Moreover, FGFR1/2 and VEGFR1/2 were co-localized in IM-resistant GIST T-1R cells, and the direct interaction between the aforementioned RTKs was confirmed by co-immunoprecipitation. In contrast, IM-resistant GIST 430 cells expressed lower basal levels of FGF2 and VEGF-A. Despite the increased expression VEGFR1 and FGFR1/2 in GIST 430 cells, these RTKs were not co-localized and co-immunoprecipitated. Moreover, no synergy between FGFR and VEGFR inhibitors was observed for the IM-resistant GIST 430 cell line. Collectively, the dual targeting of FGFR and VEGFR pathways in IM-resistant GISTs is not limited to the synergistic anti-angiogenic treatment effects. The dual inhibition of FGFR and VEGFR pathways in IM-resistant GISTs potentiates the proapoptotic and anti-proliferative activities of the corresponding RTKi. Mechanistically, the FGF2-induced activation of the FGFR pathway turns on VEGFR signaling via the overproduction of VEGF-A, induces the interaction between FGFR1/2 and VEGFR1, and thereby renders cancer cells highly sensitive to the dual inhibition of the aforementioned RTKs. Thus, our data uncovers the novel mechanism of the cross-talk between the aforementioned RTKs in IM-resistant GISTs lacking secondary KIT mutations and suggests that the dual blockade of FGFR and VEGFR signaling might be an effective treatment strategy for patients with GIST-acquired IM resistance via KIT-independent mechanisms.

3.
Adv Sci (Weinh) ; : e2404645, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39119899

RESUMEN

The lymph node is the most common site of distant metastasis of cervical squamous cell carcinoma (CSCC), which elicits dismal prognosis and limited efficiency for treatment. Elucidation of the mechanisms underlying CSCC lymphatic metastasis would provide potential therapeutic strategies for nodal metastatic of CSCC. Here, based on in vivo lymphatic metastasis screening model, a circular RNA is identified that is termed as lymph node metastasis associated circRNA (LNMAC), is markedly upregulated in lymphatic metastatic CSCC and correlated with lymph node metastasis. Overexpression of LNMAC dramatically augments the metastatic capability of CSCC cells to the lymph node via inducing lymphangiogenesis. Mechanistically, LNMAC epigenetically upregulates fibroblast growth factor 2 (FGF2) expression by directly associating with histone acacetylase 1 (HDAC1), preventing Importin α6/8-mediated nuclear translocation of HDAC1 and eliciting histone H3K27ac-induced FGF2 transcriptional activation. Treatment with 3F12E7, an anti-FGF2 monoclonal antibody, effectively inhibits LNMAC-induced CSCC lymphatic metastasis. Taken together, these findings indicate that LNMAC plays a crucial role in FGF2-mediated lymphangiogenesis and lymphatic metastasis, highlighting that LNMAC might be a therapeutic target for lymph node metastasis in CSCC patients.

4.
Cells ; 13(15)2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39120324

RESUMEN

High levels of vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2 and angiopoietin (ANG)-2 are found in tissues from oral squamous cell carcinoma (OSCC) and oral potentially malignant disorders (OPMDs). As might be expected, VEGF, FGF-2, and ANG-2 overexpression parallels the development of new blood and lymphatic vessels that nourish the growing OPMDs or OSCCs and provide the latter with metastatic routes. Notably, VEGF, FGF-2, and ANG-2 are also linked to the epithelial-to-mesenchymal transition (EMT), a trans-differentiation process that respectively promotes or exasperates the invasiveness of normal and neoplastic oral epithelial cells. Here, we have summarized published work regarding the impact that the interplay among VEGF, FGF-2, ANG-2, vessel generation, and EMT has on oral carcinogenesis. Results from the reviewed studies indicate that VEGF, FGF-2, and ANG-2 spark either protein kinase B (AKT) or mitogen-activated protein kinases (MAPK), two signaling pathways that can promote both EMT and new vessels' formation in OPMDs and OSCCs. Since EMT and vessel generation are key to the onset and progression of OSCC, as well as to its radio- and chemo-resistance, these data encourage including AKT or MAPK inhibitors and/or antiangiogenic drugs in the treatment of this malignancy.


Asunto(s)
Carcinoma de Células Escamosas , Transición Epitelial-Mesenquimal , Neoplasias de la Boca , Humanos , Neoplasias de la Boca/patología , Neoplasias de la Boca/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Progresión de la Enfermedad , Factor A de Crecimiento Endotelial Vascular/metabolismo , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Animales , Inductores de la Angiogénesis/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
5.
Heliyon ; 10(13): e34243, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39091947

RESUMEN

With the rapid development of biotechnology, long non-coding RNAs (lncRNAs) have shown promising potential for cancer treatment and may become novel therapeutic targets. This study aimed to explore the roles of lncRNAs in retinoblastoma (RB). It involves analysing differentially expressed lncRNAs in RB and normal tissues from the GSE111168 and GSE125903 datasets, further validating them in RB cells. Our findings determined that lncRNA MIMT1 was upregulated in RB cell lines and tissues. In WERI-Rb1 and Y79 cells, silencing MIMT1 significantly inhibited cell proliferation, whereas MIMT1 overexpression enhanced cell proliferation. Furthermore, in vivo xenograft experiments demonstrated that MIMT1 knockdown suppressed tumour volume and weight. Subsequent mechanistic investigations showed that MIMT1 upregulates fibroblast expression of FGF2 by binding to miR-153-5p, ultimately promoting RB cell proliferation. This suggest that MIMT1 functions as an oncogene in RB and potentially serves as a molecular marker for diagnostic and prognostic assessments. Thus, the MIMT1/miR-153-5p/FGF2 pathway is a promising therapeutic target for RB treatment.

6.
Phytomedicine ; 134: 155962, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39214017

RESUMEN

Vasculogenic mimicry (VM) is a novel model for supplying blood to multiple tumors, including gastric cancer (GC), and is a potential target for its treatment. Dihydroartemisinin (DHA) is a potential natural antitumor substance that inhibits the progression of tumors in many ways. The research aimed to evaluate the impact of DHA on VM formation and its mechanisms. The IC50 of DHA, DHA's effect on proliferation, invasion, and migration in GC cells and VM formation in both cell and animal models were determined through wound healing, MTT, EdU, colony formation, and Transwell assays. Genomics was employed to identify genes related to DHA inhibition of VM formation, and to analyze their relationship to VM formation. qRT‒PCR and western blot (WB) analysis were carried out to analyze the changes in protein and mRNA levels after DHA treatment and the changes in VM-associated protein biomarkers after blocking target gene-related pathways. The mechanism by which DHA inhibits VM in GC was elucidated in vivo. DHA reduced the invasion, proliferation, and migration of GC cells and inhibited VM in cells and in vivo. A total of 220 DEGs were identified in the DHA-treated HGC-27 cells. Among the 146 downregulated genes, fibroblast growth Factor 2 (FGF2) was most closely associated with angiogenesis and VM. The level of FGF2 in GC tissues with VM was markedly greater than in VM lacking tissues. Treatment with DHA or FGFR1 blockade suppressed VM formation and reduced VM-related biomarker proteins. DHA suppressed tumor progression and VM formation by reducing FGF2 in xenograft mouse models. Per our knowledge, this is the first study to demonstrate the inhibitory effect of DHA on VM, providing a novel strategy for the treatment of GC.

7.
Biotechnol J ; 19(8): e2400278, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39212202

RESUMEN

Skeletal muscle satellite cells (SCs) are essential for muscle regeneration. Their proliferation and differentiation are influenced by fibroblast growth factor (FGF)-2. In this study, we screened for FGF-2-derived peptides that promote SC proliferation. Utilizing photocleavable peptide array technology, a library of 7-residue peptides was synthesized, and its effect on SC proliferation was examined using a mixture of five peptides. The results showed that peptides 1-5 (136%), 21-25 (136%), 26-30 (141%), 31-35 (159%), 71-75 (135%), 76-80 (144%), and 126-130 (137%) significantly increased SC proliferation. Further experiments revealed that peptide 33, CKNGGFF, enhanced SC proliferation. Furthermore, its extended form, peptide 33-13, CKNGGFFLRIHPD, promoted SC proliferation and increased the percentage of Pax7-positive cells, indicating that SCs were maintained in an undifferentiated state. The addition of FGF-2 and peptide 33-13 further induced cell proliferation but did not increase the percentage of Pax7-positive cells. A proliferation assay using an FGF receptor (FGFR) inhibitor suggested that peptide 33-13 acts through the FGFR-mediated and other pathways. Although further research is necessary to explore the mechanisms of action of these peptides and their potential for in vivo and in vitro use, the high sequence conservation of peptides 33 and 33-13 in FGF-2 across multiple species suggests their broad application prospects in biomedical engineering and biotechnology.


Asunto(s)
Proliferación Celular , Factor 2 de Crecimiento de Fibroblastos , Péptidos , Células Satélite del Músculo Esquelético , Animales , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ratones , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Péptidos/química , Péptidos/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas
8.
Biomedicines ; 12(8)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39200192

RESUMEN

Sodium fluoride (NaF) is a fluoride application recommended by the World Health Organization for its efficacy and safety in preventing dental caries. Gingival fibroblasts that constitute the majority of connective tissue cells play a major role in wound healing via the expression of growth factors, including fibroblast growth factor-2 (FGF-2) and transforming growth factor-beta (TGF-ß). This study examined the effect of NaF mouthwash on FGF-2 and TGF-ß expression in human gingival fibroblasts (HGnFs). Fibroblasts were exposed to a medium with 225 ppmF NaF for 1 min, then switched to either 15 ppmF NaF for continuous stimulation or no NaF for transient stimulation. Continuous NaF stimulation significantly increased the gene and protein expression of FGF-2 and TGF-ß in HGnFs compared to controls, suggesting NaF's potential role in modulating periodontal tissue wound healing. Signaling pathway investigations showed the involvement of heterotrimeric GTP-binding proteins, calcium/calmodulin-dependent kinase II (CaMKII), and extracellular signal-regulated kinase (ERK) phosphorylation. Inhibiting CaMKII reduced NaF-induced FGF-2 and TGF-ß expression, while ERK phosphorylation increased after NaF stimulation. These results highlight NaF mouthwash's potential in promoting wound healing in extraction sockets, particularly during the mixed dentition period. Understanding NaF's effects is clinically relevant due to the common use of fluoride products.

9.
Biomedicines ; 12(8)2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39200370

RESUMEN

Hereditary neurodegenerative diseases (hNDDs) such as Alzheimer's, Parkinson's, Huntington's disease, and others are primarily characterized by their progressive nature, severely compromising both the cognitive and motor abilities of patients. The underlying genetic component in hNDDs contributes to disease risk, creating a complex genetic landscape. Considering the fact that growth factors play crucial roles in regulating cellular processes, such as proliferation, differentiation, and survival, they could have therapeutic potential for hNDDs, provided appropriate dosing and safe delivery approaches are ensured. This article presents a detailed overview of growth factors, and explores their therapeutic potential in treating hNDDs, emphasizing their roles in neuronal survival, growth, and synaptic plasticity. However, challenges such as proper dosing, delivery methods, and patient variability can hinder their clinical application.

10.
Biomolecules ; 14(8)2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39199276

RESUMEN

Several inflammatory cytokines bind to the allosteric site (site 2) and allosterically activate integrins. Site 2 is also a binding site for 25-hydroxycholesterol, an inflammatory lipid mediator, and is involved in inflammatory signaling (e.g., TNF and IL-6 secretion) in addition to integrin activation. FGF2 is pro-inflammatory and pro-thrombotic, and FGF1, homologous to FGF2, has anti-inflammatory and anti-thrombotic actions, but the mechanism of these actions is unknown. We hypothesized that FGF2 and FGF1 bind to site 2 of integrins and regulate inflammatory signaling. Here, we describe that FGF2 is bound to site 2 and allosterically activated ß3 integrins, suggesting that the pro-inflammatory action of FGF2 is mediated by binding to site 2. In contrast, FGF1 bound to site 2 but did not activate these integrins and instead suppressed integrin activation induced by FGF2, indicating that FGF1 acts as an antagonist of site 2 and that the anti-inflammatory action of FGF1 is mediated by blocking site 2. A non-mitogenic FGF1 mutant (R50E), which is defective in binding to site 1 of αvß3, suppressed ß3 integrin activation by FGF2 as effectively as WT FGF1.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos , Factor 2 de Crecimiento de Fibroblastos , Integrina beta3 , Humanos , Integrina beta3/metabolismo , Integrina beta3/genética , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Factor 1 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación Alostérica , Antiinflamatorios/farmacología , Sitio Alostérico , Animales , Unión Proteica , Sitios de Unión
11.
Endocrinology ; 165(9)2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39041201

RESUMEN

Breast cancer progression involves intricate interactions between cancer cells and the tumor microenvironment (TME). This study elucidates the critical role of progesterone receptor (PR) signaling in mediating the protumorigenic effects of cancer-associated fibroblasts (CAFs) on estrogen receptor-positive (ER+) luminal breast cancer cells. We demonstrate that CAFs produce physiologically relevant levels of estrogen and progesterone, which significantly contribute to breast cancer tumorigenicity. Specifically, CAF conditioned media (CM) promoted PR-dependent anchorage-independent growth, tumorsphere formation/stem cell expansion, and CD44 upregulation. CAF cells formed co-clusters more frequently with PR+ breast cancer cells relative to PR-null models. While both PR isoforms mediated these actions, PR-A was a dominant driver of tumorsphere formation/stemness, while PR-B induced robust CD44 expression and CAF/tumor cell co-cluster formation. CD44 knockdown impaired CAF/tumor cell co-clustering. Fibroblast growth factor 2 (FGF2), also secreted by CAFs, phosphorylated PR (Ser294) in a MAPK-dependent manner and activated PR to enhance CD44 expression and breast cancer tumorigenicity. The FGF receptor (FGFR) inhibitor PD173074 diminished CAF- and FGF2-dependent PR activation, tumorsphere formation, and co-clustering. In summary, this study reveals a novel mechanism through which stromal CAFs orchestrate elevated PR signaling in ER+ luminal breast cancer via secretion of both progesterone and FGF2, a potent activator of ERK1/2. Understanding tumor cell/TME interactions provides insights into potential therapeutic strategies aimed at disrupting PR- and/or FGF2/FGFR-dependent signaling pathways to prevent early metastasis in patients with ER+ breast cancer.


Asunto(s)
Neoplasias de la Mama , Fibroblastos Asociados al Cáncer , Receptores de Hialuranos , Receptores de Estrógenos , Receptores de Progesterona , Transducción de Señal , Humanos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Receptores de Progesterona/metabolismo , Receptores de Progesterona/genética , Femenino , Receptores de Hialuranos/metabolismo , Receptores de Hialuranos/genética , Receptores de Estrógenos/metabolismo , Animales , Microambiente Tumoral , Línea Celular Tumoral , Ratones , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Carcinogénesis/metabolismo , Progesterona/farmacología , Progesterona/metabolismo , Medios de Cultivo Condicionados/farmacología
12.
Int J Periodontics Restorative Dent ; 0(0): 1-21, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39058938

RESUMEN

Endodontic-periodontal lesions are characterized by the involvement of the pulp and periodontal disease in the same tooth. Despite successful root canal treatment, if the majority of bone support has been lost from periodontitis, the tooth may have a poor prognosis. In severe endodontic-periodontal lesions, the periodontal tissue regenerates poorly because of the significant loss of the periodontal ligament and cementum, poor tooth stability, and bone defect morphology unfavorable for bone regeneration. To overcome these difficult situations, in this case, osteotomy of the replantation bed and tooth replantation with horizontal rotation and deep placement were performed. To improve periodontal regeneration, fibroblast growth factor (FGF) 2 was applied to the artificially made periodontal defect. In addition, orthodontic extrusion of the deeply replaced tooth was performed for potential coronal migration of the periodontal tissue. This case presents a unique multidisciplinary method of treating severe endodontic-periodontal lesions using intentional replantation combined with FGF 2 application and orthodontic extrusion.

13.
Clin Transl Med ; 14(7): e1777, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39039912

RESUMEN

N-methyladenosine (m6A) represents a prevalent RNA modification observed in colorectal cancer. Despite its abundance, the biological implications of m6A methylation on the lncRNA CARMN remain elusive in colorectal cancer, especially for mutant p53 gain-of-function. Here, we elucidate that CARMN exhibits diminished expression levels in colorectal cancer patients with mutant p53, attributed to its rich m6A methylation, which promotes cancer proliferation, invasion and metastasis in vitro and in vivo. Further investigation illustrates that ALKBH5 acts as a direct demethylase of CARMN, targeting 477 methylation sites, thereby preserving CARMN expression. However, the interaction of mutant p53 with the ALKBH5 promoter impedes its transcription, enhancing m6A methylation levels on CARMN. Subsequently, YTHDF2/YTHDF3 recognise and degrade m6A-modified CARMN. Concurrently, overexpressing CARMN significantly suppressed colorectal cancer progression in vitro and in vivo. Additionally, miR-5683 was identified as a direct downstream target of lncRNA CARMN, exerting an antitumour effect by cooperatively downregulating FGF2 expression. Our findings revealed the regulator and functional mechanism of CARMN in colorectal cancer with mutant p53, potentially offering insights into demethylation-based strategies for cancer diagnosis and therapy. The m6A methylation of CARMN that is prime for mutant p53 gain-of-function-induced malignant progression of colorectal cancer, identifying a promising approach for cancer therapy.


Asunto(s)
Desmetilasa de ARN, Homólogo 5 de AlkB , Neoplasias Colorrectales , MicroARNs , ARN Largo no Codificante , Proteína p53 Supresora de Tumor , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Animales , Ratones , Progresión de la Enfermedad , Desmetilación , Línea Celular Tumoral , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/genética , Ratones Desnudos , Regulación Neoplásica de la Expresión Génica
14.
Artículo en Inglés | MEDLINE | ID: mdl-38980409

RESUMEN

Oral lichen planus (OLP) is a carcinogenic chronic inflammatory oral disease, which lacks effective treatments. Fraxin is an active ingredient of the traditional Chinese medicine Qin Pi, which has an anti-inflammatory effect, but its effect on OLP is unclear. The aim of this study was to investigate the therapeutic effect of fraxin on OLP and the underlying mechanism. Human immortalized keratinocytes (HaCat) were incubated with fraxin (10, 20, or 40 µM) for 48 h and then treated with 10 µg/mL LPS for 24 h. Cell viability and apoptosis were detected. Next, the interaction between OCT3 and FGF2 was predicted by online database and verified by Co-IP analysis. Fraxin, Ad-OCT3, sh-OCT3, and sh-FGF2 were, respectively, applied to treat LPS-incubated HaCat cells, and cell viability, apoptosis, and secretion of inflammatory factors were detected with MTT, flow cytometry, and ELISA assays. Then, the involvement of OCT3 and FGF2 in the prevention of fraxin on HaCat cells from LPS-induced cell apoptosis and inflammation was investigated through multiple rescue experiments. In addition, OLP models were constructed in VDR-/- mice and NOD/SCID mice by injecting with human OLP pathological tissue homogenates to verify the therapeutic effect of fraxin on OLP. Fraxin treatment increased cell viability and reduced cell apoptosis and the secretion of IL-6 and TNF-α in a dose-dependent manner. OCT3 was significantly upregulated in oral mucosa tissues of OLP mice. OCT3 silencing inhibited LPS-induced cell apoptosis and secretion of inflammatory factors. Fraxin incubation reduced the expression of OCT3, and OCT3 interacted with FGF2 to upregulate FGF2 protein. FGF2 silencing reduced the expression of p-p65/NF-κB protein and improved LPS-induced cell apoptosis and secretion of inflammatory factors. OCT3 overexpression increased the expression of FGF2 and p-p65/NF-κB proteins, rh-FGF2 aggravated this effect, while FGF2-Neu-Ab reversed this effect. The results of in vivo experiments showed that fraxin alleviated cell apoptosis and inflammation in oral buccal mucosa tissues of OLP mice. Fraxin inhibited cell apoptosis and inflammation by suppressing OCT3-mediated activation of the FGF2/NF-κB pathway, alleviating the progression of OLP.

15.
Cancer Lett ; 598: 217124, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39059573

RESUMEN

We previously reported that combined therapy with epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) osimertinib and AXL inhibitor ONO-7475 is effective in preventing the survival of drug-tolerant cells in high-AXL-expressing EGFR-mutated non-small cell lung cancer (NSCLC) cells. Nevertheless, certain residual cells are anticipated to eventually develop acquired resistance to this combination therapy. In this study, we attempted to establish a multidrug combination therapy from the first-line setting to overcome resistance to this combination therapy in high-AXL-expressing EGFR-mutated NSCLC. siRNA screening assay showed that fibroblast growth factor receptor 1 (FGFR1) knockdown induced pronounced inhibition of cell viability in the presence of the osimertinib-ONO-7475 combination, which activates FGFR1 by upregulating FGF2 via the c-Myc pathway. Cell-based assays showed that triple therapy with osimertinib, ONO-7475, and the FGFR inhibitor BGJ398 significantly increased apoptosis by increasing expression of proapoptotic factor Bim and reduced cell viability compared with that observed for the osimertinib-ONO-7475 therapy. Xenograft models showed that triple therapy considerably suppressed tumor regrowth. A novel therapeutic strategy of additional initial FGFR1 inhibition may be highly effective in suppressing the emergence of osimertinib- and ONO-7475-resistant cells.


Asunto(s)
Acrilamidas , Compuestos de Anilina , Protocolos de Quimioterapia Combinada Antineoplásica , Tirosina Quinasa del Receptor Axl , Carcinoma de Pulmón de Células no Pequeñas , Receptores ErbB , Neoplasias Pulmonares , Proteínas Proto-Oncogénicas , Pirimidinas , Proteínas Tirosina Quinasas Receptoras , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Animales , Femenino , Humanos , Ratones , Acrilamidas/farmacología , Compuestos de Anilina/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Benzocicloheptenos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Indoles , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/administración & dosificación , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Pirimidinas/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Triazoles , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Clin Med ; 13(11)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38892751

RESUMEN

Background: The risk of impaired bone-pin interface strength in titanium (Ti) pins coated with fibroblast growth factor (FGF)-calcium phosphate (CP) composite layers is yet to be evaluated in a clinical study. This retrospective study used Weibull plot analysis to evaluate bone-pin interface strength in Ti pins coated with FGF-CP layers for external distal radius fracture fixation. Methods: The distal radial fractures were treated with external fixation. The FGF-CP group comprised five patients (all women, aged 70.4 ± 5.9 (range: 62-77) years), and the uncoated pin group comprised ten patients (eight women and two men, aged 64.4 ± 11.7 (range: 43-83) years). The pins were removed after six weeks. The insertion and extraction peak torques were measured. The extraction peak torque was evaluated using Weibull plot analysis. Results: We compared the extraction torque of the two groups at or below 506 Nmm for a fair comparison using Weibull plot analysis. The Weibull plots were linear for both the FGF-CP and uncoated pin groups. The slope of the regression line was significantly higher in the FGF-CP group (1.7343) than in the uncoated pin group (1.5670) (p = 0.011). The intercept of the regression line was significantly lower in the FGF-CP group (-9.847) than in the uncoated pin group (-8.708) (p = 0.002). Thus, the two regression lines significantly differed. Conclusions: Ti pins coated with FGF-CP layers exhibit the potential to reduce the risk of impaired bone-pin interface strength in the external fixation of distal radius fractures.

17.
Sci Rep ; 14(1): 14892, 2024 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-38937503

RESUMEN

Accurate screening of COVID-19 infection status for symptomatic patients is a critical public health task. Although molecular and antigen tests now exist for COVID-19, in resource-limited settings, screening tests are often not available. Furthermore, during the early stages of the pandemic tests were not available in any capacity. We utilized an automated machine learning (ML) approach to train and evaluate thousands of models on a clinical dataset consisting of commonly available clinical and laboratory data, along with cytokine profiles for patients (n = 150). These models were then further tested for generalizability on an out-of-sample secondary dataset (n = 120). We were able to develop a ML model for rapid and reliable screening of patients as COVID-19 positive or negative using three approaches: commonly available clinical and laboratory data, a cytokine profile, and a combination of the common data and cytokine profile. Of the tens of thousands of models automatically tested for the three approaches, all three approaches demonstrated > 92% sensitivity and > 88 specificity while our highest performing model achieved 95.6% sensitivity and 98.1% specificity. These models represent a potential effective deployable solution for COVID-19 status classification for symptomatic patients in resource-limited settings and provide proof-of-concept for rapid development of screening tools for novel emerging infectious diseases.


Asunto(s)
COVID-19 , Citocinas , Aprendizaje Automático , Humanos , COVID-19/diagnóstico , Citocinas/sangre , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/inmunología , Tamizaje Masivo/métodos , Masculino , Femenino , Sensibilidad y Especificidad , Persona de Mediana Edad , Adulto , Anciano
18.
Heliyon ; 10(11): e32272, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38873667

RESUMEN

To delve into the expression and functions of FGF2 in patient with thyroid cancer (THCA), we conducted a systematic analysis of the association of FGF2 with immune cell infiltration, and prognosis in THCA patients. The transcription and protein levels, methylation, and biological properties of FGF2 were examined, along with its correlation with prognosis and immune cell infiltration in THCA patients using online databases UALCAN, Human Protein Atlas, Kaplan-Meier Plotter, DNMIVD, cBioPortal, GEPIA, Metascape, Linkedomics and TIMER. Clinical samples were collected for Western blot analyses. FGF2 was substantially overexpressed in the tumor group and shown correlations with age, tumor histology, nodal metastasis, and cancer stages. Moreover, higher expression of FGF2 (HR = 3.42, 95 % CI:1.57-7.44, p = 0.00099) was greatly correlated with poorer relapse-free survival in THCA patients, particularly in female patients. FGF2 methylation level was increased in the tumor group (p = 1.29E-6), and higher methylation levels of FGF2 were positively correlated with the poorer progression-free interval in THCA patients (p = 0.015). FGF2 mutations were markedly associated with shorter disease-free survival, with a mutation rate of 6 % among the total 498 THCA patients. FGF2 functions were potentially related to cell adhesion, cytokine-cytokine receptor interaction and angiogenesis. FGF2 expression showed positive correlations with the infiltration of B cells (Cor = 0.569, p = 1.04e-42), CD4+ T cells (Cor = 0.555, p = 9.43e-41), macrophages (Cor = 0.438, p = 2.94e-42), neutrophils (Cor = 0.578, p = 9.354e-45), and dendritic cells (Cor = 0.591, p = 5.00e-47). FGF2 is a potential prognostic marker in THCA patients, with its functions possibly related to cell adhesion, interaction of the cytokine-cytokine receptor, angiogenesis, and the promotion of multiple immune cell infiltration.

19.
Biotechnol Bioeng ; 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38877726

RESUMEN

Despite various clinical options, human anterior cruciate ligament (ACL) lesions do not fully heal. Biomaterial-guided gene therapy using recombinant adeno-associated virus (rAAV) vectors may improve the intrinsic mechanisms of ACL repair. Here, we examined whether poly(sodium styrene sulfonate)-grafted poly(ε-caprolactone) (pNaSS-grafted PCL) films can deliver rAAV vectors coding for the reparative basic fibroblast growth factor (FGF-2) and transforming growth factor beta (TGF-ß) in human mesenchymal stromal cells (hMSCs) as a source of implantable cells in ACL lesions. Efficient and sustained rAAV-mediated reporter (red fluorescent protein) and therapeutic (FGF-2 and TGF-ß) gene overexpression was achieved in the cells for at least 21 days in particular with pNaSS-grafted PCL films relative to all other conditions (up to 5.2-fold difference). Expression of FGF-2 and TGF-ß mediated by rAAV using PCL films increased the levels of cell proliferation, the DNA contents, and the deposition of proteoglycans and of type-I and -III collagen (up to 2.9-fold difference) over time in the cells with higher levels of transcription factor expression (Mohawk, Scleraxis) (up to 1.9-fold difference), without activation of inflammatory tumor necrosis alpha especially when using pNaSS-grafted PCL films compared with the controls. Overall, the effects mediated by TGF-ß were higher than those promoted by FGF-2, possibly due to higher levels of gene expression achieved upon rAAV gene transfer. This study shows the potential of using functionalized PCL films to apply rAAV vectors for ACL repair.

20.
Sci Rep ; 14(1): 11372, 2024 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762650

RESUMEN

The aim of this study was to identify angiogenic microRNAs (miRNAs) that could be used in the treatment of hindlimb ischemic tissues. miRNAs contained in extracellular vesicles (EVs) deriving from the plasma were analyzed in C57BL/6 mice, which have ischemia tolerance, and in BALB/c mice without ischemia tolerance as part of a hindlimb ischemia model; as a result 43 angiogenic miRNA candidates were identified. An aortic ring assay was employed by using femoral arteries isolated from BALC/c mice and EVs containing miRNA; as a result, the angiogenic miRNA candidates were limited to 14. The blood flow recovery was assessed after injecting EVs containing miRNA into BALB/c mice with hindlimb ischemia, and miR-709 was identified as a promising angiogenic miRNA. miR-709-encapsulating EVs were found to increase the expression levels of the fibroblast growth factor 2 (FGF2) mRNA in the thigh tissues of hindlimb ischemia model BALB/c mice. miR-709 was also found to bind to the 3'UTR of glycogen synthase kinase 3 beta (GSK3B) in three places. GSK3B-knockdown human artery-derived endothelial cells were found to express high levels of FGF2, and were characterized by increased cell proliferation. These findings indicate that miR-709 induces an upregulation of FGF2 through the downregulation of GSK3B.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos , Glucógeno Sintasa Quinasa 3 beta , Miembro Posterior , Isquemia , Ratones Endogámicos BALB C , MicroARNs , Neovascularización Fisiológica , Animales , Humanos , Masculino , Ratones , Regiones no Traducidas 3' , Proliferación Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Miembro Posterior/irrigación sanguínea , Isquemia/metabolismo , Isquemia/genética , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Neovascularización Fisiológica/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA