Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Biochem Pharmacol ; 221: 116039, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38301966

RESUMO

Translocator protein (18 kDa) (TSPO) plays an important role in retinal neuroinflammation in the early stage of diabetic retinopathy (DR). Studies have found that a FGF1 variant (FGF1ΔHBS) with reduced proliferative potency exerts excellent anti-inflammatory effects and potential therapeutic value for diabetic complications. In this study, intravitreal injection of FGF1ΔHBS was administrated every week for one month in db/db mice, which are genetically predisposed to develop type 2 diabetes mellitus and early retinopathy. Changes in retinal function and structure in the animal models were detected by electrophysiology (ERG) and optical tomography coherence (OCT). TSPO expression and retinal inflammation were analyzed by immunofluorescence, Western blot and real-time qPCR. In the retina of T2D (db/db) mice, FGF1 was significantly down-regulated while FGFR1 was up-regulated (both p < 0.05). TSPO and retinal inflammatory factors were all up-regulated. TSPO and FGFR1 were mainly co-stained in the inner retina. After FGF1ΔHBS treatment, ERG showed that the total amplitude of dark-adapted b-wave and oscillating potentials (Ops) was significantly improved, and OCT showed that the thickness of the retina around the optical nerve head was significantly preserved in T2D mice (all p < 0.05). The TSPO signal was significantly suppressed by FGF1ΔHBS. The activation of NF-κB p65 and the expression of inflammatory factors such as TNF-α, IL-1ß, IL-6, COX-2, MIP-1α, and iNOS were all significantly down-regulated (all p < 0.05). Collectively, our current data demonstrated that intravitreal FGF1ΔHBS treatment can effectively inhibit retinal inflammation via suppressing TSPO signal and to preserve retinal function and structure in a T2D mouse model.


Assuntos
Diabetes Mellitus Tipo 2 , Retinopatia Diabética , Camundongos , Animais , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Retina/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/metabolismo , Modelos Animais de Doenças , Proteínas de Transporte/metabolismo
2.
Am J Otolaryngol ; 44(4): 103895, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37075695

RESUMO

BACKGROUND: Postoperative nasal treatment is an important factor affecting the outcomes of endoscopic sinus surgery (ESS) in patients with chronic rhinosinusitis (CRS). This study aimed to determine the effect of recombinant human acidic fibroblast growth factor (rh-aFGF) on nasal mucosal healing after ESS. METHODS: This study is a prospective, single-blind, and randomized controlled clinical study. Fifty-eight CRS patients with nasal polyps (CRSwNP) with bilateral ESS were enrolled and randomly given 1 mL of budesonide nasal spray and 2 mL of rh-aFGF solution (rh-aFGF group) or 1 mL of budesonide nasal spray and 2 mL of rh-aFGF solvent (budesonide group)-infiltrated Nasopore nasal packing after ESS. Preoperative and postoperative scores for Sino-Nasal Outcome Test (SNOT-22), Visual Analogue Scale (VAS), and Lund-Kennedy were collected and analyzed. RESULTS: Forty-two patients completed the 12-week follow-up. Postoperative SNOT-22 scores and VAS scores showed no significant differences between the two groups. In terms of the Lund-Kennedy scores, there was a statistically significant difference between the two groups at the 2-, 4-, 8-, and 12-week postoperative visits, but not at the 1-week visit. Twelve weeks after surgery, the nasal mucosa had completely epithelialized in 18 patients in the rh-aFGF group and in 12 patients in the budesonide group (χ2 = 4.200, P = 0.040). CONCLUSION: The combined application of rh-aFGF and budesonide significantly improved postoperative endoscopic appearance in the nasal mucosal healing process.


Assuntos
Pólipos Nasais , Seios Paranasais , Rinite , Sinusite , Humanos , Seios Paranasais/cirurgia , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Sprays Nasais , Estudos Prospectivos , Método Simples-Cego , Rinite/tratamento farmacológico , Rinite/cirurgia , Sinusite/tratamento farmacológico , Sinusite/cirurgia , Mucosa Nasal , Pólipos Nasais/tratamento farmacológico , Pólipos Nasais/cirurgia , Budesonida , Endoscopia , Doença Crônica , Resultado do Tratamento
3.
ACS Appl Mater Interfaces ; 15(5): 6397-6410, 2023 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-36625595

RESUMO

Drugs for metabolic diseases usually require systemic administration and act on multiple tissues, which may produce some unpredictable side effects. There have been many successful studies on targeted drugs, especially antitumor drugs. However, there is still little research on metabolic disease drugs targeting specific tissues. Fibroblast growth factor 1 (FGF1) is a potential therapy for type 2 diabetes (T2D) without the risk of hypoglycemia. However, the major impediment to the clinical application of FGF1 is its mitogenic potential. We previously engineered an FGF1 variant (named FGF1ΔHBS) to tune down its mitogenic activity via reducing the heparin-binding ability. However, other notable side effects still remained, including severe appetite inhibition, pathogenic loss of body weight, and increase in fatality rate. In this study, we used AlphaFold2 and PyMOL visualization tools to construct a novel FGF1ΔHBS conjugate fused with skeletal muscle-targeted (MT) peptide through a flexible peptide linker termed MT-FGF1ΔHBS. We found that MT-FGF1ΔHBS specifically homed to skeletal muscle tissue after systemic administration and induced a potent glucose-lowering effect in T2D mice without hypoglycemia. Mechanistically, MT-FGF1ΔHBS elicits the glucose-lowering effect via AMPK activation to promote the GLUT4 expression and translocation in skeletal muscle cells. Notably, compared with native FGF1ΔHBS, MT-FGF1ΔHBS had minimal effects on food intake and body weight and did not induce any hyperplasia in major tissues of both T2D and normal mice, indicating that this muscle-homing protein may be a promising candidate for T2D treatment. Our targeted peptide strategy based on computer-aided structure prediction in this study could be effectively applied for delivering agents to functional tissues to treat metabolic or other diseases, offering enhanced efficacy and reducing systemic off-target side effects.


Assuntos
Diabetes Mellitus Tipo 2 , Hipoglicemia , Camundongos , Animais , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Músculo Esquelético , Peptídeos/metabolismo , Glucose/metabolismo , Hipoglicemia/metabolismo , Peso Corporal
4.
Int J Mol Sci ; 23(21)2022 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-36361940

RESUMO

In our previous study, human fibroblast growth factor 1 was successfully fused with oleosomes, energy-storing organelles of seeds, which are considered to be excellent "expression carriers" for substances with a convenient purification process. The present work aimed to explore the beneficial effects of oleosomes fused with human fibroblast growth factor 1 (OLAF) on wound healing. The data showed marked improvements in terms of the angiogenesis, vascular integrity, collagen and inflammation on the wound sites of rats with a full-thickness skin defect. Moreover, the positive role of OLAF in promoting angiogenesis and its possible pathways were clarified in vivo and in vitro. The results showed that the number, length and branches of the blood vessels of the chick embryo chorioallantoic membrane were markedly increased after OLAF treatment. Meanwhile, the in vitro results also revealed that 100 ng/mL OLAF exhibited a promoting effect on the proliferation, migration and tube formation of human umbilical vein endothelial cells. In addition, the potential of OLAF to improve wound angiogenesis was demonstrated to be associated with an up-regulated PI3K/Akt pathway by transcriptome sequencing analysis and the introduction of a PI3K/Akt pathway inhibitor (LY294002). These findings suggest that OLAF has many prospects in the development of drugs for wound healing.


Assuntos
Fator 1 de Crescimento de Fibroblastos , Gotículas Lipídicas , Cicatrização , Animais , Embrião de Galinha , Humanos , Ratos , Inibidores da Angiogênese/farmacologia , Movimento Celular , Proliferação de Células , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Células Endoteliais da Veia Umbilical Humana/metabolismo , Gotículas Lipídicas/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/metabolismo , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologia
5.
Stem Cell Res Ther ; 13(1): 341, 2022 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-35883153

RESUMO

BACKGROUND: Traumatic brain injury (TBI) leads to cell and tissue impairment, as well as functional deficits. Stem cells promote structural and functional recovery and thus are considered as a promising therapy for various nerve injuries. Here, we aimed to investigate the role of ectoderm-derived frontal bone mesenchymal stem cells (FbMSCs) in promoting cerebral repair and functional recovery in a murine TBI model. METHODS: A murine TBI model was established by injuring C57BL/6 N mice with moderate-controlled cortical impact to evaluate the extent of brain damage and behavioral deficits. Ectoderm-derived FbMSCs were isolated from the frontal bone and their characteristics were assessed using multiple differentiation assays, flow cytometry and microarray analysis. Brain repairment and functional recovery were analyzed at different days post-injury with or without FbMSC application. Behavioral tests were performed to assess learning and memory improvements. RNA sequencing analysis, immunofluorescence staining, and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) were used to examine inflammation reaction and neural regeneration. In vitro co-culture analysis and quantification of glutamate transportation were carried out to explore the possible mechanism of neurogenesis and functional recovery promoted by FbMSCs. RESULTS: Ectoderm-derived FbMSCs showed fibroblast like morphology and osteogenic differentiation capacity. FbMSCs were CD105, CD29 positive and CD45, CD31 negative. Different from mesoderm-derived MSCs, FbMSCs expressed the ectoderm-specific transcription factor Tfap2ß. TBI mice showed impaired learning and memory deficits. Microglia and astrocyte activation, as well as neural damage, were significantly increased post-injury. FbMSC application ameliorated the behavioral deficits of TBI mice and promoted neural regeneration. RNA sequencing analysis showed that signal pathways related to inflammation decreased, whereas those related to neural activation increased. Immunofluorescence staining and qRT-PCR data revealed that microglial activation and astrocyte polarization to the A1 phenotype were suppressed by FbMSC application. In addition, FGF1 secreted from FbMSCs enhanced glutamate transportation by astrocytes and alleviated the cytotoxic effect of excessive glutamate on neurons. CONCLUSIONS: Ectoderm-derived FbMSC application significantly alleviated neuroinflammation, brain injury, and excitatory toxicity to neurons, improved cognition and behavioral deficits in TBI mice. Therefore, ectoderm-derived FbMSCs could be ideal therapeutic candidates for TBI which mostly affect cells from the same embryonic origins as FbMSCs.


Assuntos
Lesões Encefálicas Traumáticas , Lesões Encefálicas , Células-Tronco Mesenquimais , Animais , Lesões Encefálicas/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Ectoderma/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Osso Frontal/metabolismo , Ácido Glutâmico/metabolismo , Ácido Glutâmico/farmacologia , Ácido Glutâmico/uso terapêutico , Inflamação/metabolismo , Inflamação/terapia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Doenças Neuroinflamatórias , Osteogênese
6.
Br J Cancer ; 127(7): 1340-1351, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35778553

RESUMO

BACKGROUND: Ovarian cancer patients frequently develop chemotherapy resistance, limiting treatment options. We have previously shown that individuality in fibroblast growth factor 1 (FGF1) expression influences survival and chemotherapy response. METHODS: We used MTT assays to assess chemosensitivity to cisplatin and carboplatin following shRNA-mediated knockdown or heterologous over-expression of FGF1 (quantified by qRT-PCR and immunoblot analysis), and in combination with the FGFR inhibitors AZD4547 and SU5402, the ATM inhibitor KU55933 and DNA-PK inhibitor NU7026. Immunofluorescence microscopy was used to quantify the FGF1-dependent timecourse of replication protein A (RPA) and γH2AX foci formation. RESULTS: Pharmacological inhibition of FGF signalling reversed drug resistance in immortalised cell lines and in primary cell lines from drug-resistant ovarian cancer patients, while FGF1 over-expression induced resistance. Ataxia telangiectasia mutated (ATM) phosphorylation, but not DNA adduct formation was FGF1 dependent, following cisplatin or carboplatin challenge. Combining platinum drugs with the ATM inhibitor KU55933, but not with the DNA-PK inhibitor NU7026 re-sensitised resistant cells. FGF1 expression influenced the timecourse of damage-induced RPA and γH2AX nuclear foci formation. CONCLUSION: Drug resistance arises from FGF1-mediated differential activation of high-fidelity homologous recombination DNA damage repair. FGFR and ATM inhibitors reverse platinum drug resistance, highlighting novel combination chemotherapy approaches for future clinical trial evaluation.


Assuntos
Cisplatino , Neoplasias Ovarianas , Proteínas Mutadas de Ataxia Telangiectasia , Carboplatina/uso terapêutico , Carcinoma Epitelial do Ovário/tratamento farmacológico , Linhagem Celular Tumoral , Cisplatino/uso terapêutico , Dano ao DNA , Reparo do DNA , Proteína Quinase Ativada por DNA/metabolismo , Resistência a Medicamentos , Feminino , Fator 1 de Crescimento de Fibroblastos/genética , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Fatores de Crescimento de Fibroblastos , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Platina/uso terapêutico , RNA Interferente Pequeno , Reparo de DNA por Recombinação , Proteína de Replicação A/genética
7.
Biochim Biophys Acta Mol Basis Dis ; 1868(8): 166414, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35447340

RESUMO

Elderly adults are at higher risk for developing diabetic complications including diabetic nephropathy (DN), contributing to excess morbidity and mortality in elderly individuals. A non-mitogenic variant of fibroblast growth factor 1 (FGF1ΔHBS) was demonstrated to prevent DN in an early-stage (2-month-old) type 2 diabetes (T2D) mouse model. The present study aimed to investigate the potential therapeutic effects of FGF1ΔHBS against the progression of renal dysfunction in a late-stage T2D mouse model with established DN. Nine-month-old db/db mice were administered FGF1ΔHBS every other day for 3 months. db/db mice at 12-month-old without FGF1ΔHBS treatment exhibited high blood glucose level and elevated urine albumin-to-creatinine ratio. FGF1ΔHBS treatment effectively reversed hyperglycemia, delayed the development of renal dysfunction, and reduced kidney size and weight. Furthermore, FGF1ΔHBS treatment significantly prevented the progression of renal morphologic impairment. FGF1ΔHBS treatment demonstrated anti-inflammatory and anti-fibrotic effects, with significantly decreased protein levels of key pro-inflammatory cytokines and pro-fibrotic factors in kidney. Moreover, FGF1ΔHBS treatment greatly decreased apoptosis of renal tubular cells, accompanied by significant downregulation of the proapoptotic protein and upregulation of the antiapoptotic protein and peroxisome proliferator-activated receptor α (PPARα) expression in kidney. Mechanistically, FGF1ΔHBS treatment directly protected mouse proximal tubule cells against palmitate-induced apoptosis, which was abolished by PPARα inhibition. In conclusion, this study demonstrated that FGF1ΔHBS delays the progression of renal dysfunction likely through activating PPARα to prevent renal tubule cell death in late-stage T2D, exhibiting a promising translational potential in treating DN in elderly T2D individuals by ameliorating renal inflammation, fibrosis and apoptosis.


Assuntos
Diabetes Mellitus Tipo 2 , Nefropatias Diabéticas , Fator 1 de Crescimento de Fibroblastos , Animais , Apoptose , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/metabolismo , Modelos Animais de Doenças , Fator 1 de Crescimento de Fibroblastos/genética , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Fibrose , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Camundongos , PPAR alfa/metabolismo
8.
Neoplasia ; 28: 100788, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35378464

RESUMO

BACKGROUND: Considerable evidence implicates myeloid-derived suppressor cells (MDSCs) promote tumor progression and drug resistance. Sorafenib is the standard first-line therapy for advanced hepatocellular carcinoma (HCC). Clinical evidence indicates that sorafenib resistance is associated with increased MDSCs, by which MDSCs exerts these effects is obscure. This study aimed to investigate the mechanism of sorafenib resistance mediated by MDSCs. METHODS: A syngeneic mouse-liver cancer cell line BNL was subcutaneously injected to build a tumor-bearing mouse model, and syngeneic MDSCs were adoptive transferred into the tumor-bearing mouse. Tumor tissue was obtained, and transcriptomic analysis of the tumor was carried out on RNAseq data. A coculture system was used to verify the crosstalk between MDSCs and BNL cells. RESULTS: Adoptive MDSCs transfer into tumor-bearing mice induced an increase of tumor-infiltrating MDSCs, which led to tumor growth and impaired antitumor activity of sorafenib in BNL HCC models. MDSCs transfer contributed to tumor fibrosis and tumor-associated fibroblast (CAF) activation, associated with fibroblast growth factor (FGF1) upregulation. In contrast, MDSC depletion by anti-Ly6G+ reduced fibrosis and increased sorafenib antitumor efficacy. Intriguingly, tumor-infiltrating MDSCs barely expressed FGF1. IL-6 derived from MDSCs increased FGF1 expression in BNL liver cancer cells, and anti-IL-6 attenuated this effect in vitro. MAPK pathway, one of the sorafenib targets, is the downstream signaling of FGF1 and is reactivated by MDSCs-mediated FGF1 upregulation. CONCLUSIONS: Our finding demonstrated that MDSCs led to tumor growth and sorafenib resistance via FGF1 upregulation and subsequent indirect CAF activation. We offered a novel mechanism of MDSCs-driven HCC progression and sorafenib resistance.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Células Supressoras Mieloides , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Fibrose , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Camundongos , Células Supressoras Mieloides/metabolismo , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico , Regulação para Cima
9.
Arch Pediatr ; 28(4): 296-300, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33715934

RESUMO

INTRODUCTION: Infantile hemangioma (IH) is a common vascular tumor in children. It is reported that IHs are associated with immunochemical markers such as vascular endothelial growth factor (VEGF)-A, glucose transporter isoform 1 (GLUT1), and insulin-like growth factor-2 (IGF-2). MATERIAL AND METHODS: This cross-sectional study focused on pediatric patients with IH. A total of 46 patients (mean age 14.2±21.9 months) with IH and 45 healthy controls (mean age 21.8±15.08 months) were enrolled. Demographic data, clinical findings, and laboratory parameters were recorded. Blood samples were collected. Serum GLUT1, IGF-2, VEGF-A, fibroblast growth factor 1 (FGF1), and angiopoietin 2 levels were assessed by enzyme-linked immunosorbent assay. RESULTS: Serum GLUT1, IGF-2, and VEGF-A levels were significantly higher in patients with IH than in healthy controls (8.80±4.07pg/mL vs. 5.66±4.34pg/mL, 281.10±84.12pg/mL vs. 234.19±75.38pg/mL, 1196.99±389.34pg/mL vs. 996.99±349.16pg/mL, respectively, p=0.026, p=0.030, and p=0.036). Serum GLUT1, IGF-2, and VEGF-A levels in patients with complicated hemangioma were significantly higher than in healthy controls (9.69±3.94pg/mL vs. 5.66±4.34pg/mL, 289.94±83.18pg/mL vs. 234.19±75.38pg/mL, 1276.22±388.24pg/mL vs. 996.99±349.16pg/mL, respectively, p=0.017, p=0.022, and p=0.011). Serum GLUT1, IGF-2, and VEGF-A levels in patients with hemangioma receiving propranolol treatment were significantly higher than in healthy controls. Serum FGF1 levels were higher in patients with IH, complicated hemangioma, and hemangioma receiving propranolol treatment than in healthy controls but the difference was not statistically significantly. CONCLUSION: Serum GLUT1, IGF-2, and VEGF-A levels were positively correlated with disease severity in patients with hemangioma, for example, in complicated hemangioma and hemangioma requiring propranolol treatment. However, further research on larger and different age subgroups is warranted to assess these markers.


Assuntos
Angiopoietina-2/sangue , Fator 1 de Crescimento de Fibroblastos/sangue , Transportador de Glucose Tipo 1/sangue , Hemangioma/tratamento farmacológico , Fator de Crescimento Insulin-Like II/análise , Propranolol/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/sangue , Neoplasias Vasculares/tratamento farmacológico , Angiopoietina-2/uso terapêutico , Biomarcadores/sangue , Criança , Pré-Escolar , Estudos Transversais , Ensaio de Imunoadsorção Enzimática , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Hemangioma/sangue , Hemangioma/patologia , Humanos , Lactente , Masculino , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Neoplasias Vasculares/sangue , Neoplasias Vasculares/patologia
10.
Cell Death Dis ; 12(1): 107, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33479232

RESUMO

Prolonged type 2 diabetes mellitus (T2DM) produces a common complication, peripheral neuropathy, which is accompanied by nerve fiber disorder, axon atrophy, and demyelination. Growing evidence has characterized the beneficial effects of acidic fibroblast growth factor (aFGF) and shown that it relieves hyperglycemia, increases insulin sensitivity, and ameliorates neuropathic impairment. However, there is scarce evidence on the role of aFGF on remodeling of aberrant myelin under hyperglycemia condition. Presently, we observed that the expression of aFGF was rapidly decreased in a db/db T2DM mouse model. Administration of exogenous aFGF was sufficient to block acute demyelination and nerve fiber disorganization. Furthermore, this strong anti-demyelinating effect was most likely dominated by an aFGF-mediated increase of Schwann cell (SC) proliferation and migration as well as suppression of its apoptosis. Mechanistically, the beneficial biological effects of aFGF on SC behavior and abnormal myelin morphology were likely due to the inhibition of hyperglycemia-induced oxidative stress activation, which was most likely activated by kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid-derived-like 2 (Nrf2) signaling. Thus, this evidence indicates that aFGF is a promising protective agent for relieving myelin pathology through countering oxidative stress signaling cascades under diabetic conditions.


Assuntos
Diabetes Mellitus Tipo 2/terapia , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Animais , Modelos Animais de Doenças , Fator 1 de Crescimento de Fibroblastos/farmacologia , Masculino , Camundongos , Estresse Oxidativo , Ratos , Espécies Reativas de Oxigênio
11.
Nat Metab ; 2(10): 1025-1033, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32895577

RESUMO

We recently showed that perineuronal nets (PNNs) enmesh glucoregulatory neurons in the arcuate nucleus (Arc) of the mediobasal hypothalamus (MBH)1, but whether these PNNs play a role in either the pathogenesis of type 2 diabetes (T2D) or its treatment remains unclear. Here we show that PNN abundance within the Arc is markedly reduced in the Zucker diabetic fatty (ZDF) rat model of T2D, compared with normoglycaemic rats, correlating with altered PNN-associated sulfation patterns of chondroitin sulfate glycosaminoglycans in the MBH. Each of these PNN-associated changes is reversed following a single intracerebroventricular (icv) injection of fibroblast growth factor 1 (FGF1) at a dose that induces sustained diabetes remission in male ZDF rats. Combined with previous work localizing this FGF1 effect to the Arc area2-4, our finding that enzymatic digestion of Arc PNNs markedly shortens the duration of diabetes remission following icv FGF1 injection in these animals identifies these extracellular matrix structures as previously unrecognized participants in the mechanism underlying diabetes remission induced by the central action of FGF1.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/fisiopatologia , Matriz Extracelular , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Hipotálamo/fisiopatologia , Neurônios , Idoso , Animais , Glicemia , Peso Corporal , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Ingestão de Alimentos , Fator 1 de Crescimento de Fibroblastos/administração & dosagem , Humanos , Injeções Intraventriculares , Masculino , Pessoa de Meia-Idade , Ratos , Ratos Wistar , Ratos Zucker , Adulto Jovem
12.
Biochem Biophys Res Commun ; 528(2): 249-255, 2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32482386

RESUMO

Type-2 diabetes (T2D) is a common metabolic disorder, which causes several physiological and pathological complications. Spleen is regarded as an important organ, which regulates immune system and iron metabolism in the body. Precious few studies have been conducted to explore the pathological and deleterious roles of diabetes on spleen. In our current study, we have explored and confirmed the pathological effects of diabetes on spleen in db/db experimental mice model. In our current study, 0.5 mg/kg fibroblast growth factor 1 (FGF1) dose was intraperitoneally administrated to db/db mice. We found that diabetes evidently induced spleen enlargement and fibrosis progression in the db/db mice. Additionally, our studies demonstrate that iron has hugely deposited in the spleen in db/db mice. Several studies have documented that diabetes largely disrupts the inflammatory cells distribution, immune homeostasis, proliferation and oxidative stress with the down-regulation of anti-inflammatory cytokines and antioxidant activities. Moreover, we have observed that FGF1 administration significantly reversed the deleterious effect of diabetes on spleen enlargement and dysfunction. In summary, these substantial findings clearly demonstrate that diabetes plays deleterious roles in maintaining the spleen structure and functions. Therefore, our investigations suggest that FGF1 can effectively prevent diabetes-mediated splenomegaly progression.


Assuntos
Diabetes Mellitus Experimental/complicações , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Inflamação/patologia , Estresse Oxidativo , Esplenomegalia/tratamento farmacológico , Esplenomegalia/etiologia , Animais , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Fator 1 de Crescimento de Fibroblastos/administração & dosagem , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fibrose , Inflamação/complicações , Ferro/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , Baço/efeitos dos fármacos , Baço/patologia
13.
Diabetes ; 68(7): 1462-1472, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31048370

RESUMO

Fibroblast growth factor 1 (FGF1) has been shown to reverse hyperglycemia in diabetic rodent models through peripheral and central administration routes. Previous studies demonstrated that insulin is required for central and peripheral FGF1 metabolic improvements; however, it is unknown if FGF1 targets insulin secretion at the islet level. Here we show for the first time that FGF1 increases islet insulin secretion in diabetic mouse models. FGF1 was administered via a single intracerebroventricular or multiple subcutaneous injections to leptin receptor-deficient (db/db), diet-induced obese, and control mice; pancreatic islets were isolated 7 days later for analysis of insulin secretion. Central and peripheral FGF1 significantly lowered blood glucose in vivo and increased ex vivo islet insulin secretion from diabetic, but not control, mice. FGF1 injections to the cisterna magna mimicked intracerebroventricular outcomes, pointing to a novel therapeutic potential. Central effects of FGF1 appeared dependent on reductions in food intake, whereas peripheral FGF1 had acute actions on islet function prior to significant changes in food intake or blood glucose. Additionally, peripheral, but not central, FGF1 increased islet ß-cell density, suggesting that peripheral FGF1 may induce long-term changes in islet structure and function that are not present with central treatment.


Assuntos
Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Fator 1 de Crescimento de Fibroblastos/administração & dosagem , Teste de Tolerância a Glucose , Imuno-Histoquímica , Infusões Intraventriculares , Injeções Subcutâneas , Secreção de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-fos/metabolismo
14.
World Neurosurg ; 129: e343-e351, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31132502

RESUMO

OBJECTIVE: Repair of spinal cord injury (SCI) using peripheral nerve graft (PNG) and acidic fibroblast growth factor (aFGF) has shown promising results in rats and a few human patients, but not in nonhuman primates. The aim of this study was to verify the effective use of PNG and aFGF for repairing incomplete SCI in nonhuman primates. METHODS: Six adult rhesus macaques received spinal cord hemisection at T8 level and were grouped into repair and control groups (n = 3 in each). Animals in the repair group underwent nerve repair with autologous PNG plus aFGF immediately after lesioning. The control group received exactly the same operation for lesioning but no treatment. Postoperative behavioral evaluations, electrophysiologic tests (including motor and somatosensory evoked potentials), and magnetic resonance imaging were performed and compared between the 2 groups as well as histologic examination of the spinal cord cephalic to, at, and caudal to the lesion site after sacrifice. RESULTS: Animals in the repair group had better motor function in the lower limbs at every observed time point and demonstrated more improvement on electrophysiologic examinations than the control group. The repair group had smaller areas of myelomalacia on magnetic resonance imaging around the lesion compared with the control group, suggesting diminished inflammatory responses with the repair strategy. CONCLUSIONS: PNG plus aFGF for SCI in nonhuman primates yielded improvements in clinical behavior, electrophysiologic tests, and magnetic resonance imaging. This study suggests that the repair strategy is feasible and effective for nonhuman primate SCI. Further investigations are warranted to corroborate its effectiveness for clinical application.


Assuntos
Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Regeneração Nervosa/fisiologia , Transferência de Nervo/métodos , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/terapia , Animais , Potencial Evocado Motor/fisiologia , Potenciais Somatossensoriais Evocados/fisiologia , Macaca mulatta , Masculino , Modelos Animais , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/cirurgia
15.
Clin Res Hepatol Gastroenterol ; 43(6): 707-714, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31029643

RESUMO

Acetaminophen (APAP) overdose/abuse is the leading cause of acute liver failure in many countries. Fibroblast growth factor 1 (FGF 1) is a metabolic regulator with several physiological functions. Previous studies suggested that FGF1 promotes differentiation and maturation of liver-derived stem cells. In this study, we investigated the protective effects of FGF1 against APAP-induced hepatotoxicity in mice. APAP markedly increased circulating levels of ALT and AST, while FGF1 significantly inhibited increases in the serum levels of ALT and AST, as compared to littermates. In addition, histopathological evaluation of the livers revealed that FGF1 prevented APAP-induced centrilobular necrosis. Livers exhibited severe inflammation, apoptosis, oxidative stress and endoplasmic reticulum stress in response to APAP toxicity, whereas these changes were reversed by a single injection of FGF1. In conclusion, our findings suggest that FGF1 protects mice from APAP-induced hepatotoxicity through suppression of inflammation, apoptosis, and oxidative and endoplasmic reticulum stress. Therefore, FGF1 may represent a promising therapeutic agent for APAP-induced acute liver injury.


Assuntos
Acetaminofen/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Estresse do Retículo Endoplasmático/fisiologia , Fator 1 de Crescimento de Fibroblastos/fisiologia , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Estresse Oxidativo/fisiologia , Animais , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fator 1 de Crescimento de Fibroblastos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos
16.
Diabetes ; 68(3): 654-664, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30523024

RESUMO

We recently reported that in rodent models of type 2 diabetes (T2D), a single intracerebroventricular (icv) injection of fibroblast growth factor 1 (FGF1) induces remission of hyperglycemia that is sustained for weeks. To clarify the peripheral mechanisms underlying this effect, we used the Zucker diabetic fatty fa/fa rat model of T2D, which, like human T2D, is characterized by progressive deterioration of pancreatic ß-cell function after hyperglycemia onset. We report that although icv FGF1 injection delays the onset of ß-cell dysfunction in these animals, it has no effect on either glucose-induced insulin secretion or insulin sensitivity. These observations suggest that FGF1 acts in the brain to stimulate insulin-independent glucose clearance. On the basis of our finding that icv FGF1 treatment increases hepatic glucokinase gene expression, we considered the possibility that increased hepatic glucose uptake (HGU) contributes to the insulin-independent glucose-lowering effect of icv FGF1. Consistent with this possibility, we report that icv FGF1 injection increases liver glucokinase activity by approximately twofold. We conclude that sustained remission of hyperglycemia induced by the central action of FGF1 involves both preservation of ß-cell function and stimulation of HGU through increased hepatic glucokinase activity.


Assuntos
Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Animais , Glicemia/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Glucoquinase/genética , Glucoquinase/metabolismo , Teste de Tolerância a Glucose , Humanos , Hipoglicemiantes/uso terapêutico , Insulina/metabolismo , Resistência à Insulina , Masculino , Ratos , Ratos Zucker , Reação em Cadeia da Polimerase em Tempo Real
17.
Invest Ophthalmol Vis Sci ; 59(11): 4720-4730, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30267094

RESUMO

Purpose: Organ cultures of rabbit corneas have been used to ascertain the effectiveness of a human fibroblast growth factor (FGF)-1 derivative (TTHX1114), lacking cysteine residues, to protect against and/or repair epithelial lesions following exposure to nitrogen mustard (NM). Methods: Rabbit corneas were exposed to NM and cultured for up to 14 days, with or without drug (TTHX1114). At specified times, tissue was examined by histopathology and graded by a novel composite scale. Proliferation was measured by 5-ethynyl-2'-deoxyuridine (EdU) incorporation, and the expression of native FGF-1 and ADAM-17 after NM exposure was determined by immunofluorescence. Results: Rabbit corneas, exposed to a single dose of NM, showed a nearly complete loss of epithelial cells by day 6 but were significantly regenerated by day 14. When treated continuously with TTHX1114 following vesicant exposure, the losses remained at day 2 levels. The loss of keratocytes in the stroma was not affected by TTHX1114. EdU incorporation over the same time course showed a steady increase in tissue that had not been treated with TTHX1114, while corneas that were treated with the drug showed a higher percent incorporation initially, which then decreased, indicating the strong proliferative response to TTHX1114. ADAM-17 was not significantly altered by TTHX1114 treatment. Corneal epithelial FGF-1 disappeared after only 1 day following exposure to NM. Conclusions: TTHX1114 is protective against NM-induced damage of the corneal epithelium, possibly by supplying an NM-resistant source of trophic support and by stimulating regeneration of new epithelial cells. These responses underscore the potential value of TTHX1114 as an anti-vesicant therapeutic.


Assuntos
Substâncias para a Guerra Química/toxicidade , Córnea/efeitos dos fármacos , Lesões da Córnea/prevenção & controle , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Mecloretamina/toxicidade , Proteína ADAM17/metabolismo , Animais , Córnea/metabolismo , Córnea/patologia , Lesões da Córnea/induzido quimicamente , Lesões da Córnea/metabolismo , Dano ao DNA , Fator 1 de Crescimento de Fibroblastos/análogos & derivados , Fator 1 de Crescimento de Fibroblastos/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Técnicas de Cultura de Órgãos , Engenharia de Proteínas , Coelhos
18.
Sci Rep ; 8(1): 12691, 2018 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-30139947

RESUMO

Few treatments have proven effective for patients with chronic spinal cord injury (SCI). This study aimed to evaluate the efficacy and safety of acidic fibroblast growth factor (aFGF) in human SCI. This was an open-label prospective clinical trial of aFGF with an extended follow-up to 48 months. All patients were treated with aFGF 3 times, including once directly applied to the injured spinal cord during neurolysis surgery, and twice via lumbar punctures at 3- and 6-months post-operation. Every patient was evaluated with standardized measurements of neurological functions. The trial initially enrolled 60 patients (30 cervical and 30 thoracolumbar SCI), but only 46 (21 cervical- and 25 thoracolumbar-SCI) completed the follow-up. The ASIA impairment scales, motor, pin prick, light touch, and FIM motor subtotal scores were all improved in both groups, except that the ASIA scores of light touch only demonstrated tendency of increase in the cervical-SCI group. All patients had a decrease in dependence, and there were no major adverse events or other oncological problems throughout the follow-up. At 48 months, the study demonstrated that aFGF was safe, feasible, and could yield modest functional improvement in chronic SCI patients. Further randomized control investigations are warranted for validation of its optimal dosage.


Assuntos
Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/metabolismo , Adolescente , Adulto , Idoso , Doença Crônica/tratamento farmacológico , Feminino , Fator 1 de Crescimento de Fibroblastos/efeitos adversos , Humanos , Masculino , Pessoa de Meia-Idade , Regeneração Nervosa/efeitos dos fármacos , Estudos Prospectivos , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/reabilitação , Adulto Jovem
19.
J Cell Mol Med ; 22(5): 2727-2738, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29512938

RESUMO

Fibroblast growth factor 1 (FGF1) is thought to exert protective and regenerative effects on neurons following spinal cord injury (SCI), although the mechanism of these effects is not well understood. The use of FGF1 as a therapeutic agent is limited by its lack of physicochemical stability and its limited capacity to cross the blood-spinal cord barrier. Here, we demonstrated that overexpression of FGF1 in spinal cord following SCI significantly reduced tissue loss, protected neurons in the ventricornu, ameliorated pathological morphology of the lesion, dramatically improved tissue recovery via neuroprotection, and promoted axonal regeneration and remyelination both in vivo and in vivo. In addition, the autophagy and the expression levels of PRDX1 (an antioxidant protein) were induced by AAV-FGF1 in PC12 cells after H2 O2 treatment. Furthermore, the autophagy levels were not changed in PRDX1-suppressing cells that were treated by AAV-FGF1. Taken together, these results suggest that FGF1 improves functional recovery mainly through inducing PRDX1 expression to increase autophagy and anti-ROS activity after SCI.


Assuntos
Autofagia , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Peroxirredoxinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/fisiopatologia , Animais , Autofagia/efeitos dos fármacos , Axônios/efeitos dos fármacos , Axônios/metabolismo , Polaridade Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Dependovirus/genética , Feminino , Fator 1 de Crescimento de Fibroblastos/farmacologia , Vetores Genéticos/metabolismo , Atividade Motora/efeitos dos fármacos , Regeneração Nervosa/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Células PC12 , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Remielinização/efeitos dos fármacos , Traumatismos da Medula Espinal/patologia
20.
Int J Nanomedicine ; 12: 7103-7119, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29026304

RESUMO

The present study seeks to observe the preventive effects of doxorubicin-induced cardiomyopathy (DOX-CM) in rats using targeted non-mitogenic acidic fibroblast growth factor (MaFGF) mediated by nanoparticles (NP) combined with ultrasound-targeted MB destruction (UTMD). DOX-CM rats were induced by intraperitoneally injected doxorubicin. Six weeks after intervention, the indices from the transthoracic echocardiography and velocity vector imaging showed that the left ventricular function in the MaFGF-loaded NP (MaFGF-NP) + UTMD group was significantly improved compared with the DOX-CM group. The increased malondialdehyde and decreased superoxide dismutase were observed in the DOX-CM group, while a significant increase in superoxide dismutase and a decrease in malondialdehyde were detected in the groups treated with MaFGF-NP + UTMD. From the Masson staining, the MaFGF-NP + UTMD group showed a significant difference from the DOX-CM group. The cardiac collagen volume fraction and the ratio of the perivascular collagen area to the luminal area number of terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labelling positive cells in the MaFGF-NP + UTMD group decreased to 8.9%, 0.55-fold, compared with the DOX-CM group (26.5%, 1.7-fold). From terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labelling staining, the results showed the strongest inhibition of apoptosis progress in MaFGF-NP + UTMD group. The immunohistochemical staining of the TGF-ß1 in MaFGF-NP + UTMD group reached 3.6%, which was much lower than that of the DOX-CM group (12.6%). These results confirmed that the abnormalities, including left ventricular dysfunction, myocardial fibrosis, cardiomyocytes apoptosis and oxidative stress, could be suppressed by twice weekly MaFGF treatments for 6 consecutive weeks (free MaFGF or MaFGF-NP+/UTMD), with the strongest improvements observed in the MaFGF-NP + UTMD group. Western blot analyses of the heart tissue further revealed the highest pAkt levels, highest anti-apoptosis protein (Bcl-2) levels and strongest reduction in proapoptosis protein (Bax) levels in the MaFGF-NP + UTMD group. This study confirmed the preventive effects of DOX-CM in the rats with MaFGF-NP and UTMD by retarding myocardial fibrosis, inhibiting oxidative stress, and decreasing cardiomyocyte apoptosis.


Assuntos
Cardiomiopatias/induzido quimicamente , Cardiomiopatias/prevenção & controle , Doxorrubicina/efeitos adversos , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Microbolhas , Nanopartículas/química , Ultrassom , Animais , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Cardiomiopatias/diagnóstico por imagem , Cardiomiopatias/patologia , Fator 1 de Crescimento de Fibroblastos/farmacologia , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Ventrículos do Coração/ultraestrutura , Masculino , Malondialdeído/metabolismo , Mitógenos , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Nanopartículas/ultraestrutura , Tamanho do Órgão/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Disfunção Ventricular Esquerda/prevenção & controle , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA