Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
J Oral Pathol Med ; 53(6): 366-375, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38763759

RESUMEN

BACKGROUND: Angiopoietin-like 4 is a molecular hallmark that correlates with the growth and metastasis of head and neck squamous cell carcinoma, one of the most prevalent cancers worldwide. However, the molecular mechanisms by which angiopoietin-like 4 promotes head and neck squamous cell carcinoma tumorigenesis are unclear. METHODS: Using well-characterized cell lines of head and neck squamous cell carcinoma development, including human normal oral keratinocytes, dysplastic oral keratinocytes, oral leukoplakia-derived oral keratinocytes, and head and neck squamous cell carcinoma cell lines, HN13, HN6, HN4, HN12, and CAL27, we investigated the signaling pathways upstream and downstream of angiopoietin-like 4-induced head and neck squamous cell carcinoma tumorigenesis. RESULTS: We found that both epidermal growth factor receptor ligands, epithelial growth factor, and amphiregulin led to angiopoietin-like 4 upregulation in normal oral keratinocytes and dysplastic oral keratinocytes and cooperated with the activation of hypoxia-inducible factor-1 in this effect. Interestingly, amphiregulin and angiopoietin-like 4 were increased in dysplastic oral keratinocytes and head and neck squamous cell carcinoma cell lines, and amphiregulin-induced activation of cell proliferation was dependent on angiopoietin-like 4. Although both p38 mitogen-activated protein kinases (p38 MAPK) and protein kinase B (AKT) were activated by angiopoietin-like 4, only pharmacological inhibition of p38 MAPK was sufficient to prevent head and neck squamous cell carcinoma cell proliferation and migration. We further observed that angiopoietin-like 4 promoted the secretion of interleukin 11 (IL-11), interleukin 12 (IL-12), interleukin-1 alpha (IL-1α), vascular endothelial growth factor, platelet-derived growth factor (PDGF), and tumour necrosis factor alpha (TNF-α), cytokines and chemokines previously implicated in head and neck squamous cell carcinoma pathogenesis. CONCLUSION: Our results demonstrate that angiopoietin-like 4 is a downstream effector of amphiregulin and promotes head and neck squamous cell carcinoma development both through direct activation of p38 kinase as well as paracrine mechanisms.


Asunto(s)
Anfirregulina , Proteína 4 Similar a la Angiopoyetina , Movimiento Celular , Proliferación Celular , Neoplasias de Cabeza y Cuello , Carcinoma de Células Escamosas de Cabeza y Cuello , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos , Humanos , Anfirregulina/farmacología , Anfirregulina/metabolismo , Proliferación Celular/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Movimiento Celular/efectos de los fármacos , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Proteína 4 Similar a la Angiopoyetina/metabolismo , Línea Celular Tumoral , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Transducción de Señal , Queratinocitos/metabolismo , Queratinocitos/efectos de los fármacos , Receptores ErbB/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(50): e2302845120, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38055741

RESUMEN

It has previously been reported that antioxidant vitamins can help reduce the risk of vision loss associated with progression to advanced age-related macular degeneration (AMD), a leading cause of visual impairment among the elderly. Nonetheless, how oxidative stress contributes to the development of choroidal neovascularization (CNV) in some AMD patients and geographic atrophy (GA) in others is poorly understood. Here, we provide evidence demonstrating that oxidative stress cooperates with hypoxia to synergistically stimulate the accumulation of hypoxia-inducible factor (HIF)-1α in the retinal pigment epithelium (RPE), resulting in increased expression of the HIF-1-dependent angiogenic mediators that promote CNV. HIF-1 inhibition blocked the expression of these angiogenic mediators and prevented CNV development in an animal model of ocular oxidative stress, demonstrating the pathological role of HIF-1 in response to oxidative stress stimulation in neovascular AMD. While human-induced pluripotent stem cell (hiPSC)-derived RPE monolayers exposed to chemical oxidants resulted in disorganization and disruption of their normal architecture, RPE cells proved remarkably resistant to oxidative stress. Conversely, equivalent doses of chemical oxidants resulted in apoptosis of hiPSC-derived retinal photoreceptors. Pharmacologic inhibition of HIF-1 in the mouse retina enhanced-while HIF-1 augmentation reduced-photoreceptor apoptosis in two mouse models for oxidative stress, consistent with a protective role for HIF-1 in photoreceptors in patients with advanced dry AMD. Collectively, these results suggest that in patients with AMD, increased expression of HIF-1α in RPE exposed to oxidative stress promotes the development of CNV, but inadequate HIF-1α expression in photoreceptors contributes to the development of GA.


Asunto(s)
Neovascularización Coroidal , Atrofia Geográfica , Degeneración Macular Húmeda , Ratones , Animales , Humanos , Anciano , Epitelio Pigmentado de la Retina/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Inhibidores de la Angiogénesis , Degeneración Macular Húmeda/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Agudeza Visual , Neovascularización Coroidal/genética , Neovascularización Coroidal/prevención & control , Neovascularización Coroidal/metabolismo , Oxidantes/metabolismo , Hipoxia/metabolismo
3.
J Oral Pathol Med ; 52(10): 1004-1012, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37817274

RESUMEN

BACKGROUND: Compelling evidence implicates diabetes-associated hyperglycemia as a promoter of tumor progression in oral potentially malignant disorders (OPMD). Yet, information on hyperglycemia-induced cell signaling networks in oral oncology remains limited. Our group recently reported that glucose-rich conditions significantly enhance oral dysplastic keratinocyte viability and migration through epidermal growth factor receptor (EGFR) activation, a pathway strongly linked to oral carcinogenesis. Here, we investigated the basal metabolic phenotype in these cells and whether specific glucose-responsive EGFR ligands mediate these responses. METHODS: Cell energy phenotype and lactate concentration were evaluated via commercially available assays. EGFR ligands in response to normal (5 mM) or high (20 mM) glucose were analyzed by quantitative real-time PCR, ELISA, and western blotting. Cell viability and migration assays were performed in the presence of pharmacological inhibitors or RNA interference. RESULTS: When compared to normal keratinocytes, basal glycolysis in oral dysplastic keratinocytes was significantly elevated. In highly glycolytic cells, high glucose-activated EGFR increasing viability and migration. Notably, we identified amphiregulin (AREG) as the predominant glucose-induced EGFR ligand. Indeed, enhanced cell migration in response to high glucose was blunted by EGFR inhibitor cetuximab and AREG siRNA. Conversely, AREG treatment under normal glucose conditions significantly increased cell viability, migration, lactate levels, and expression of glycolytic marker pyruvate kinase M2. CONCLUSION: These novel findings point to AREG as a potential high glucose-induced EGFR activating ligand in highly glycolytic oral dysplastic keratinocytes. Future studies are warranted to gain more insight into the role of AREG in hyperglycemia-associated OPMD tumor progression.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Neoplasias , Humanos , Anfirregulina/genética , Anfirregulina/metabolismo , Glucosa/farmacología , Glucosa/metabolismo , Ligandos , Receptores ErbB/metabolismo , Familia de Proteínas EGF/metabolismo , Queratinocitos/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/metabolismo , Carcinogénesis/metabolismo , Lactatos/metabolismo
4.
Cell Signal ; 108: 110697, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37169211

RESUMEN

OBJECTIVES: The molecular mechanisms whereby angiopoietin-like 4 (ANGPTL4), a pluripotent protein implicated in cancer development, contributes to head and neck squamous cell carcinoma (HNSCC) growth and dissemination are unclear. MATERIALS AND METHODS: We investigated ANGPTL4 expression in human normal oral keratinocytes (NOKs), dysplastic oral keratinocytes (DOKs), oral leukoplakia cells (LEUK1), and HNSCC cell lines, as well as in tissue biopsies from patients with oral dysplasia, and primary and metastatic HNSCC. We further examined the contribution of ANGPTL4 cancer progression in an HNSCC orthotopic floor-of mouth tumor model and the signaling pathways linking ANGPTL4 to cancer cell migration. RESULTS: ANGPTL4 expression was upregulated in premalignant DOKs and HNSCC cell lines compared to NOKs and was increased in tissue biopsies from patients with oral dysplasia, as well as in primary and metastatic HNSCC. We also observed that downregulation of ANGPTL4 expression inhibited primary and metastatic cancer growth in an HNSCC orthotopic tumor model. Interestingly, ANGPTL4 binding to the neuropilin1 (NRP1) receptor led to phosphorylation of the focal adhesion protein, paxillin (PXN), and tumor cell migration; this was dependent on the tyrosine kinase ABL1. Treatment with the ABL1 inhibitor, dasatinib and small interfering RNA silencing of NRP1 or ABL1 expression blocked PXN phosphorylation and tumor cell migration. CONCLUSION: Our findings suggest an early, sustained, and angiogenesis-independent autocrine role for ANGPTL4 in HNSCC progression and expose ANGPTL4/NRP1/ABL1/PXN as an early molecular marker and vulnerable target for the prevention of HNSCC growth and metastasis.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Humanos , Angiopoyetinas/genética , Angiopoyetinas/metabolismo , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Neuropilina-1/metabolismo , Paxillin/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello
5.
J Clin Invest ; 133(13)2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37227777

RESUMEN

Many patients with diabetic eye disease respond inadequately to anti-VEGF therapies, implicating additional vasoactive mediators in its pathogenesis. We demonstrate that levels of angiogenic proteins regulated by HIF-1 and -2 remain elevated in the eyes of people with diabetes despite treatment with anti-VEGF therapy. Conversely, by inhibiting HIFs, we normalized the expression of multiple vasoactive mediators in mouse models of diabetic eye disease. Accumulation of HIFs and HIF-regulated vasoactive mediators in hyperglycemic animals was observed in the absence of tissue hypoxia, suggesting that targeting HIFs may be an effective early treatment for diabetic retinopathy. However, while the HIF inhibitor acriflavine prevented retinal vascular hyperpermeability in diabetic mice for several months following a single intraocular injection, accumulation of acriflavine in the retina resulted in retinal toxicity over time, raising concerns for its use in patients. Conversely, 32-134D, a recently developed HIF inhibitor structurally unrelated to acriflavine, was not toxic to the retina, yet effectively inhibited HIF accumulation and normalized HIF-regulated gene expression in mice and in human retinal organoids. Intraocular administration of 32-134D prevented retinal neovascularization and vascular hyperpermeability in mice. These results provide the foundation for clinical studies assessing 32-134D for the treatment of patients with diabetic eye disease.


Asunto(s)
Diabetes Mellitus Experimental , Retinopatía Diabética , Neovascularización Retiniana , Humanos , Ratones , Animales , Acriflavina/metabolismo , Acriflavina/farmacología , Acriflavina/uso terapéutico , Diabetes Mellitus Experimental/metabolismo , Retina/metabolismo , Neovascularización Retiniana/metabolismo , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo
6.
Cell Rep ; 42(1): 111976, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36640318

RESUMEN

Tight glycemic control (TGC), the cornerstone of diabetic management, reduces the incidence and progression of diabetic microvascular disease. However, TGC can also lead to transient episodes of hypoglycemia, which have been associated with adverse outcomes in patients with diabetes. Here, we demonstrate that low glucose levels result in hypoxia-inducible factor (HIF)-1-dependent expression of the glucose transporter, Glut1, in retinal cells. Enhanced nuclear accumulation of HIF-1α was independent of its canonical post-translational stabilization but instead dependent on stimulation of its translation and nuclear localization. In the presence of hypoxia, this physiologic response to low glucose resulted in a marked increase in the secretion of the HIF-dependent vasoactive mediators that promote diabetic retinopathy. Our results provide a molecular explanation for how early glucose control, as well as glycemic variability (i.e., oscillating serum glucose levels), contributes to diabetic eye disease. These observations have important implications for optimizing glucose management in patients with diabetes.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Hipoglucemia , Humanos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Retinopatía Diabética/metabolismo , Glucosa/metabolismo , Hipoglucemia/complicaciones , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia
7.
J Clin Invest ; 133(2)2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36413411

RESUMEN

BACKGROUNDStudies assessing the efficacy of therapies for neovascular age-related macular degeneration (nvAMD) have demonstrated that aflibercept may have a longer treatment interval than its less-expensive alternative, bevacizumab. However, whether this benefit justifies the additional cost of aflibercept remains under debate. We have recently reported that a treat-and-extend-pause/monitor approach can be used to successfully wean 31% of patients with nvAMD off anti-VEGF therapy. Here, we examined whether the choice of therapy influences the outcomes of this approach.METHODSIn this retrospective analysis, 122 eyes of 106 patients with nvAMD underwent 3 consecutive monthly injections with either aflibercept (n = 70) or bevacizumab (n = 52), followed by a treat-and-extend protocol, in which the decision to extend the interval between treatments was based on visual acuity, clinical exam, and the presence or absence of fluid on optical coherence tomography. Eyes that remained stable 12 weeks from their prior treatment were given a 6-week trial of holding further treatment, followed by quarterly monitoring. Treatment was resumed for worsening vision, clinical exam, or optical coherence tomography findings.RESULTSAt the end of 1 year, eyes receiving bevacizumab had similar vision but required more injections (8.7 ± 0.3 treatments vs. 7.2 ± 0.3 treatments) compared with eyes receiving aflibercept. However, eyes treated with aflibercept were almost 3 times more likely to be weaned off treatment (43% vs. 15%) compared with eyes treated with bevacizumab at the end of 1 year.CONCLUSIONThese observations expose an advantage of aflibercept over bevacizumab and have important clinical implications for the selection of therapy for patients with nvAMD.FUNDINGThis work was supported by the National Eye Institute, NIH grants R01EY029750 and R01EY025705, Research to Prevent Blindness, the Alcon Young Investigator Award from the Alcon Research Institute, and the Branna and Irving Sisenwein Professorship in Ophthalmology.


Asunto(s)
Inhibidores de la Angiogénesis , Degeneración Macular , Humanos , Bevacizumab/uso terapéutico , Inhibidores de la Angiogénesis/uso terapéutico , Ranibizumab/efectos adversos , Estudios Retrospectivos , Proteínas Recombinantes de Fusión/uso terapéutico , Degeneración Macular/tratamiento farmacológico , Resultado del Tratamiento , Estudios de Seguimiento
8.
JCI Insight ; 7(13)2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35653189

RESUMEN

Most patients with neovascular age-related macular degeneration (nvAMD), the leading cause of severe vision loss in elderly US citizens, respond inadequately to current therapies targeting a single angiogenic mediator, vascular endothelial growth factor (VEGF). Here, we report that aqueous fluid levels of a second vasoactive mediator, angiopoietin-like 4 (ANGPTL4), can help predict the response of patients with nvAMD to anti-VEGF therapies. ANGPTL4 expression was higher in patients who required monthly treatment with anti-VEGF therapies compared with patients who could be effectively treated with less-frequent injections. We further demonstrate that ANGPTL4 acts synergistically with VEGF to promote the growth and leakage of choroidal neovascular (CNV) lesions in mice. Targeting ANGPTL4 expression was as effective as targeting VEGF expression for treating CNV in mice, while simultaneously targeting both was more effective than targeting either factor alone. To help translate these findings to patients, we used a soluble receptor that binds to both VEGF and ANGPTL4 and effectively inhibited the development of CNV lesions in mice. Our findings provide an assay that can help predict the response of patients with nvAMD to anti-VEGF monotherapy and suggest that therapies targeting both ANGPTL4 and VEGF will be a more effective approach for the treatment of this blinding disease.


Asunto(s)
Neovascularización Coroidal , Degeneración Macular , Proteína 4 Similar a la Angiopoyetina , Animales , Humor Acuoso/metabolismo , Neovascularización Coroidal/tratamiento farmacológico , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/metabolismo , Ratones , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Sci Adv ; 8(9): eabm1896, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35235351

RESUMEN

For patients with proliferative diabetic retinopathy (PDR) who do not respond adequately to pan-retinal laser photocoagulation (PRP) or anti-vascular endothelial growth factor (VEGF) therapies, we hypothesized that vascular cells within neovascular tissue secrete autocrine/paracrine angiogenic factors that promote disease progression. To identify these factors, we performed multiplex ELISA angiogenesis arrays on aqueous fluid from PDR patients who responded inadequately to anti-VEGF therapy and/or PRP and identified plasminogen activator inhibitor-1 (PAI-1). PAI-1 expression was increased in vitreous biopsies and neovascular tissue from PDR eyes, limited to retinal vascular cells, regulated by the transcription factor hypoxia-inducible factor (HIF)-2α, and necessary and sufficient to stimulate angiogenesis. Using a pharmacologic inhibitor of HIF-2α (PT-2385) or nanoparticle-mediated RNA interference targeting Pai1, we demonstrate that the HIF-2α/PAI-1 axis is necessary for the development of retinal neovascularization in mice. These results suggest that targeting HIF-2α/PAI-1 will be an effective adjunct therapy for the treatment of PDR patients.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Neovascularización Retiniana , Inductores de la Angiogénesis/uso terapéutico , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/etiología , Retinopatía Diabética/metabolismo , Humanos , Ratones , Neovascularización Patológica , Inhibidor 1 de Activador Plasminogénico/genética , Neovascularización Retiniana/etiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
J Clin Invest ; 132(2)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34874918

RESUMEN

BackgroundTo reduce the treatment burden for patients with neovascular age-related macular degeneration (nvAMD), emerging therapies targeting vascular endothelial growth factor (VEGF) are being designed to extend the interval between treatments, thereby minimizing the number of intraocular injections. However, which patients will benefit from longer-acting agents is not clear.MethodsEyes with nvAMD (n = 122) underwent 3 consecutive monthly injections with currently available anti-VEGF therapies, followed by a treat-and-extend protocol. Patients who remained quiescent 12 weeks from their prior treatment entered a treatment pause and were switched to pro re nata (PRN) treatment (based on vision, clinical exam, and/or imaging studies). Proteomic analysis was performed on aqueous fluid to identify proteins that correlate with patients' response to treatment.ResultsAt the end of 1 year, 38 of 122 eyes (31%) entered a treatment pause (≥30 weeks). Conversely, 21 of 122 eyes (17%) failed extension and required monthly treatment at the end of year 1. Proteomic analysis of aqueous fluid identified proteins that correlated with patients' response to treatment, including proteins previously implicated in AMD pathogenesis. Interestingly, apolipoprotein-B100 (ApoB100), a principal component of drusen implicated in the progression of nonneovascular AMD, was increased in treated patients who required less frequent injections. ApoB100 expression was higher in AMD eyes compared with controls but was lower in eyes that develop choroidal neovascularization (CNV), consistent with a protective role. Accordingly, mice overexpressing ApoB100 were partially protected from laser-induced CNV.FundingThis work was supported by the National Eye Institute, National Institutes of Health grants R01EY029750, R01EY025705, and R01 EY27961; the Research to Prevent Blindness, Inc.; the Alcon Research Institute; and Johns Hopkins University through the Robert Bond Welch and Branna and Irving Sisenwein professorships in ophthalmology.ConclusionAqueous biomarkers could help identify patients with nvAMD who may not require or benefit from long-term treatment with anti-VEGF therapy.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Apolipoproteína B-100/metabolismo , Neovascularización Coroidal , Proteínas del Ojo/metabolismo , Degeneración Macular , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Anciano , Anciano de 80 o más Años , Animales , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/metabolismo , Femenino , Humanos , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/metabolismo , Masculino , Ratones , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
J Clin Invest ; 131(12)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34128478

RESUMEN

Therapies targeting VEGF have proven only modestly effective for the treatment of proliferative sickle cell retinopathy (PSR), the leading cause of blindness in patients with sickle cell disease. Here, we shift our attention upstream from the genes that promote retinal neovascularization (NV) to the transcription factors that regulate their expression. We demonstrated increased expression of HIF-1α and HIF-2α in the ischemic inner retina of PSR eyes. Although both HIFs participated in promoting VEGF expression by hypoxic retinal Müller cells, HIF-1 alone was sufficient to promote retinal NV in mice, suggesting that therapies targeting only HIF-2 would not be adequate to prevent PSR. Nonetheless, administration of a HIF-2-specific inhibitor currently in clinical trials (PT2385) inhibited NV in the oxygen-induced retinopathy (OIR) mouse model. To unravel these discordant observations, we examined the expression of HIFs in OIR mice and demonstrated rapid but transient accumulation of HIF-1α but delayed and sustained accumulation of HIF-2α; simultaneous expression of HIF-1α and HIF-2α was not observed. Staggered HIF expression was corroborated in hypoxic adult mouse retinal explants but not in human retinal organoids, suggesting that this phenomenon may be unique to mice. Using pharmacological inhibition or an in vivo nanoparticle-mediated RNAi approach, we demonstrated that inhibiting either HIF was effective for preventing NV in OIR mice. Collectively, these results explain why inhibition of either HIF-1α or HIF-2α is equally effective for preventing retinal NV in mice but suggest that therapies targeting both HIFs will be necessary to prevent NV in patients with PSR.


Asunto(s)
Anemia de Células Falciformes/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Neovascularización Retiniana/metabolismo , Vasos Retinianos/metabolismo , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Modelos Animales de Enfermedad , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Indanos/farmacología , Ratones , Neovascularización Retiniana/etiología , Neovascularización Retiniana/genética , Sulfonas/farmacología
13.
JAMA Netw Open ; 4(2): e2037880, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33616665

RESUMEN

Importance: Ten percent of the Medicare Part B budget is spent on aflibercept, used to treat a myriad of ocular neovascular diseases. A substantial portion of these costs can be attributed to a few hundred ophthalmologists, raising concerns regarding the influence of pharmaceutical companies on the choice of medication by a relatively small group of clinicians. One approach to protect patients' health care interests is to include them in deliberations on the choice of therapy for their eye disease. Objective: To examine factors associated with patients' choice between an effective and less expensive off-label drug or a more effective, but also more expensive, US Food and Drug Administration (FDA)-approved drug. Design, Setting, and Participants: This retrospective cohort analysis used data from the satellite office of a tertiary referral center from August 2, 2013, to April 9, 2018. Insured patients initiating treatment with anti-vascular endothelial growth factor were included in the analysis. Data were analyzed from March 26, 2018, to June 10, 2020. Interventions: Patients were asked to choose between bevacizumab (approximately $100 per dose), a chemotherapy that is effective, but not FDA approved, for the treatment of ocular vascular disease, or aflibercept (approximately $2000 per dose), an FDA-approved drug for ocular vascular disease that may be more effective than bevacizumab in some patients. Independent of this choice, patients were separately asked by a study coordinator to participate in an invasive clinical study for which they would not be compensated, there was a small risk for an adverse event, and they would not personally benefit from participating (a surrogate marker for altruism). Main Outcomes and Measures: Factors associated with patients' choice of medication, including age, sex, ocular disease, race, and participation in an invasive clinical study. Results: A total of 189 patients were included in the analysis (106 women [56%]; mean [SEM] age, 74.6 [0.8] years). Despite being told that it may not be as effective as aflibercept, 100 patients (53%) selected bevacizumab for their own eye care. An act of altruism (ie, participation in an invasive clinical study) when the patient was making a choice between the 2 drugs was associated with a patient's choice of bevacizumab (odds ratio [OR], 7.03; 95% CI, 2.27-21.80; P < .001); the OR for selecting bevacizumab for patients who never agreed to participate in the clinical study was 0.45 (95% CI, 0.25-0.83; P = .001). Age (OR, 1.00; 95% CI, 0.97-1.03; P = .86), race (OR, 0.70; 95% CI, 0.41-1.22; P = .21), sex (OR, 0.72; 95% CI, 0.39-1.35; P = .31), presence of diabetes (OR, 1.52; 95% CI, 0.59-3.93; P = .39), and type of eye disease (OR, 0.56; 95% CI, 0.30-1.04; P = .07) were not associated with choice of therapy. Conclusions and Relevance: These findings suggest that clinicians must consider the ethical implications of the influence of altruism when patients participate in the decision between cost-effective vs the most effective medicines for their own health care.


Asunto(s)
Altruismo , Inhibidores de la Angiogénesis/economía , Bevacizumab/economía , Conducta de Elección , Toma de Decisiones , Oftalmopatías/tratamiento farmacológico , Participación del Paciente , Proteínas Recombinantes de Fusión/economía , Negro o Afroamericano , Anciano , Inhibidores de la Angiogénesis/uso terapéutico , Asiático , Bevacizumab/uso terapéutico , Estudios de Cohortes , Análisis Costo-Beneficio , Retinopatía Diabética/tratamiento farmacológico , Costos de los Medicamentos , Femenino , Humanos , Degeneración Macular/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Neovascularización Patológica/tratamiento farmacológico , Oportunidad Relativa , Uso Fuera de lo Indicado , Receptores de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Oclusión de la Vena Retiniana/tratamiento farmacológico , Estudios Retrospectivos , Resultado del Tratamiento , Agudeza Visual , Población Blanca
14.
J Biomater Sci Polym Ed ; 32(7): 886-905, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33482702

RESUMEN

A low-shrinkage-stress (LSS), antibacterial and remineralizing nanocomposite was recently developed; however, validation of its long-term antibacterial potency in modulating human salivary-derived biofilm is an unmet need. This study aimed to evaluate the antibacterial effect of the bioactive LSS composite before and after aging in acidic solution for 90 days using a multi-species biofilm model, and to evaluate its cytotoxicity. The LSS composite consisted of urethane dimethacrylate (UDMA) and triethylene glycol divinylbenzyl ether (TEG-DVBE), 3% dimethylaminohexadecyl methacrylate (DMAHDM) and 20% nanoparticles of amorphous calcium phosphate (NACP). Biofilm colony-forming units (CFU), lactic acid production, and confocal laser scanning microscopy (3D biofilm) were evaluated before and after three months of aging. Cytotoxicity was assessed against human gingival fibroblasts (HGF). The new LSS composite presented the lowest biofilm CFU, lactic acid and biofilm biomass, compared to controls (n = 6, p < 0.05). Importantly, the new composite exhibited no significant difference in antibacterial performance before and after 90-day-aging, demonstrating long-term antibacterial activity (p > 0.1). The LSS antibacterial and remineralizing composite presented a low cell viability at original extract that has increased with further dilutions. In conclusion, this study spotlighted that the new bioactive composite not only had a low shrinkage stress, but also down-regulated the growth of oral biofilms, reduced acid production, maintained antibacterial activity after the 90-day-aging, and did not compromise the cytocompatibility.


Asunto(s)
Nanocompuestos , Nanopartículas , Antibacterianos/farmacología , Biopelículas , Fosfatos de Calcio , Humanos , Ácido Láctico , Metacrilatos
15.
J Dent ; 99: 103406, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32526346

RESUMEN

OBJECTIVES: Polymerization shrinkage stress may lead to marginal damage, microleakage and failure of composite restorations. The objectives of this study were to : (1) develop a novel nanocomposite with low-shrinkage-stress, antibacterial and remineralization properties to reduce marginal enamel demineralization under biofilms; (2) evaluate the mechanical properties of the composite and calcium (Ca) and phosphate (P) ion release; and (3) investigate the cytotoxicity of the new low-shrinkage-stress monomer in vitro. METHODS: The low-shrinkage-stress resin consisted of urethane dimethacrylate (UDMA) and triethylene glycol divinylbenzyl ether (TEG-DVBE), and 3 % dimethylaminohexadecyl methacrylate (DMAHDM) and 20 % calcium phosphate nanoparticles (NACP) were added. Mechanical properties, polymerization shrinkage stress, and degree of conversion were evaluated. The growth of Streptococcus mutans (S. mutans) on enamel slabs with different composites was assessed. Ca and P ion releases and monomer cytotoxicity were measured. RESULTS: Composite with DMAHDM and NACP had flexural strength of 84.9 ±â€¯10.3 MPa (n = 6), matching that of a commercial control composite. Adding 3 % DMAHDM did not negatively affect the composite ion release. Under S. mutans biofilm, the marginal enamel hardness was 1.2 ±â€¯0.1 GPa for the remineralizing and antibacterial group, more than 2-fold the 0.5 ±â€¯0.07 GPa for control (p < 0.05). The polymerization shrinkage stress of the new composite was 40 % lower than that of traditional composite control (p < 0.05). The new monomers had fibroblast viability similar to that of traditional monomer control (p > 0.1). CONCLUSION: A novel low-shrinkage-stress nanocomposite was developed with remineralizing and antibacterial properties. This new composite is promising to inhibit recurrent caries at the restoration margins by reducing polymerization stress and protecting enamel hardness.


Asunto(s)
Biopelículas , Nanocompuestos , Antibacterianos/farmacología , Fosfatos de Calcio/farmacología , Resinas Compuestas/farmacología , Esmalte Dental , Metacrilatos/farmacología , Streptococcus mutans
16.
J Clin Invest ; 129(11): 4593-4608, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31545295

RESUMEN

The majority of patients with diabetic macular edema (DME), the most common cause of vision loss in working-age Americans, do not respond adequately to current therapies targeting VEGFA. Here, we show that expression of angiopoietin-like 4 (ANGPTL4), a HIF-1-regulated gene product, is increased in the eyes of diabetic mice and patients with DME. We observed that ANGPTL4 and VEGF act synergistically to destabilize the retinal vascular barrier. Interestingly, while ANGPTL4 modestly enhanced tyrosine phosphorylation of VEGF receptor 2, promotion of vascular permeability by ANGPTL4 was independent of this receptor. Instead, we found that ANGPTL4 binds directly to neuropilin 1 (NRP1) and NRP2 on endothelial cells (ECs), leading to rapid activation of the RhoA/ROCK signaling pathway and breakdown of EC-EC junctions. Treatment with a soluble fragment of NRP1 (sNRP1) prevented ANGPTL4 from binding to NRP1 and blocked ANGPTL4-induced activation of RhoA as well as EC permeability in vitro and retinal vascular leakage in diabetic animals in vivo. In addition, sNRP1 reduced the stimulation of EC permeability by aqueous fluid from patients with DME. Collectively, these data identify the ANGPTL4/NRP/RhoA pathway as a therapeutic target for the treatment of DME.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/metabolismo , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Edema Macular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Diabetes Mellitus Experimental/patología , Retinopatía Diabética/patología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Edema Macular/patología , Ratones , Neuropilina-1/metabolismo , Neuropilina-2/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
17.
Invest Ophthalmol Vis Sci ; 58(11): 4514-4523, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28873177

RESUMEN

Purpose: Disappointing results from clinical studies assessing the efficacy of therapies targeting vascular endothelial growth factor (VEGF) for the treatment of pterygia suggest that other angiogenic mediators may also play a role in its development. We therefore explore the relative contribution of VEGF, hypoxia-inducible factor (HIF)-1α (the transcription factor that regulates VEGF expression in ocular neovascular disease), and a second HIF-regulated mediator, angiopoietin-like 4 (ANGPTL4), to the angiogenic phenotype of pterygia. Methods: Expression of HIF-1α, VEGF, and ANGPTL4 were examined in surgically excised pterygia, and in immortalized human (ih) and primary rabbit (pr) conjunctival epithelial cells (CjECs). Endothelial cell (EC) tubule formation assays using media conditioned by ihCjECs in the presence or absence of inducers/inhibitors of HIF-1 or RNA interference (RNAi) targeting VEGF, ANGPTL4, or both were used to assess their relative contribution to the angiogenic potential of these cells. Results: HIF-1α and VEGF expression were detected in 6/6 surgically excised pterygia and localized to CjECs. Accumulation of HIF-1α in was confirmed in ihCjECs and prCjECs, including stratified prCjECs grown on collagen vitrigel, and resulted in expression of VEGF and the promotion of EC tubule formation; the latter effect was partially blocked using RNAi targeting VEGF mRNA expression. We demonstrate expression of a second HIF-regulated angiogenic mediator, ANGPTL4, in CjECs in culture and in surgically excised pterygia. RNAi targeting ANGPTL4 inhibited EC tubule formation and was additive to RNAi targeting VEGF. Conclusions: Our results support the development of therapies targeting both ANGPTL4 and VEGF for the treatment of patients with pterygia.


Asunto(s)
Angiopoyetinas/metabolismo , Conjuntiva/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neovascularización Patológica/metabolismo , Pterigion/metabolismo , Moduladores de la Angiogénesis , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/genética , Animales , Western Blotting , Células Cultivadas , Digoxina/farmacología , Endotelio Vascular/fisiología , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Fenotipo , Interferencia de ARN , ARN Mensajero/genética , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Sci Rep ; 7(1): 10608, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28878218

RESUMEN

The development of symptoms in patients with epiretinal membranes (ERMs) often corresponds with the accumulation of interstitial fluid in the retina [i.e., the development of macular edema, (ME)]. To explore the potential value of pharmacologic therapeutic options to treat ME in patients with ERMs, we examine here the expression of vasoactive and inflammatory mediators in the vitreous of patients with idiopathic ERMs. We observed that vitreous concentrations of classic vasoactive factors (e.g., vascular endothelial growth factor) were similar in ERM patients with ME compared to controls. Using an array assessing the expression of 102 inflammatory cytokines we similarly did not observe a marked difference in cytokine expression in the vitreous of most ERM patients with ME compared to control patients. While the array data did implicate a group of inflammatory cytokines that were elevated in a subset of ERM patients who had severe ME (central subfield thickness ≥450 µm on spectral domain optical coherence tomography), expression of 3 of these inflammatory cytokines, all previously implicated in the promotion of ME in ischemic retinal disease, were not elevated by quantitative enzyme-linked immunosorbent assay. We conclude that therapies modulating vasoactive mediators or inflammatory cytokines may not affect ME in ERM patients.


Asunto(s)
Biomarcadores , Membrana Epirretinal/metabolismo , Membrana Epirretinal/patología , Mediadores de Inflamación/metabolismo , Edema Macular/metabolismo , Edema Macular/patología , Angiopoyetinas/genética , Angiopoyetinas/metabolismo , Permeabilidad Capilar , Citocinas/genética , Citocinas/metabolismo , Angiografía con Fluoresceína , Expresión Génica , Humanos , Edema Macular/etiología , Vasos Retinianos/metabolismo , Tomografía de Coherencia Óptica , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
PLoS One ; 12(8): e0183320, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28832635

RESUMEN

The recent success of therapies directly targeting the angiogenic mediator, vascular endothelial growth factor (VEGF), for the treatment of proliferative diabetic retinopathy has encouraged clinicians to extend the use of anti-VEGF therapies for the treatment of another ischemic retinal vascular disease, proliferative sickle cell retinopathy (PSR), the most common cause of irreversible blindness in patients with sickle cell disease. However, results from case reports evaluating anti-VEGF therapies for PSR have been mixed. This highlights the need to identify alternative therapeutic targets for the treatment of retinal neovascularization in sickle cell patients. In this regard, angiopoietin-like 4 (ANGPTL4) is a novel angiogenic factor regulated by the transcription factor, hypoxia-inducible factor 1, the master regulator of angiogenic mediators (including VEGF) in ischemic retinal disease. In an effort to identify alternative targets for the treatment of sickle cell retinopathy, we have explored the expression of ANGPTL4 in the eyes of patients with PSR. To this end, we examined expression and localization of ANGPTL4 by immunohistochemistry in autopsy eyes from patients with known PSR (n = 5 patients). Complementary studies were performed using enzyme-linked immunosorbent assays in aqueous (n = 8; 7 patients, 2 samples from one eye of same patient) and vitreous (n = 3 patients) samples from a second group of patients with active PSR. We detected expression of ANGPTL4 in neovascular tissue and in the ischemic inner retina in PSR, but not control, eyes. We further observed elevated expression of ANGPTL4 in the aqueous and vitreous of PSR patients compared to controls. These results suggest that ANGPTL4 could contribute to the development of retinal neovascularization in sickle cell patients and could therefore be a therapeutic target for the treatment of PSR.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Angiopoyetinas/metabolismo , Ceguera/metabolismo , Neovascularización Patológica/metabolismo , Proteína 4 Similar a la Angiopoyetina , Ceguera/etiología , Humanos , Neovascularización Patológica/etiología , Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA