Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 509
Filter
1.
Cancer Immunol Immunother ; 72(1): 55-71, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35708739

ABSTRACT

Tumour acidosis contributes to cancer progression by inhibiting anti-tumour immunity. However, the effect of acidosis on anti-tumour T cell phenotypes in oesophageal adenocarcinoma (OAC) is unknown. Therefore, this study investigated the effect of acidosis on anti-tumour T cell profiles and if immune checkpoint blockade (ICB) could enhance anti-tumour T cell immunity under acidosis. Acidic conditions substantially altered immune checkpoint expression profiles of OAC patient-derived T cells, upregulating TIM-3, LAG-3 and CTLA-4. Severe acidosis (pH 5.5) significantly decreased the percentage of central memory CD4+ T cells, an effect that was attenuated by ICB treatment. ICB increased T cell production of IFN-γ under moderate acidosis (pH 6.6) but not severe acidosis (pH 5.5) and decreased IL-10 production by T cells under severe acidic conditions only. A link between lactate and metastasis was also depicted; patients with nodal metastasis had higher serum lactate levels (p = 0.07) which also positively correlated with circulating levels of pro-angiogenic factor Tie-2. Our findings establish that acidosis-induced upregulation of immune checkpoints on T cells may potentially contribute to immune evasion and disease progression in OAC. However, acidic conditions curtailed ICB efficacy, supporting a rationale for utilizing systemic oral buffers to neutralize tumour acidity to improve ICB efficacy. Study schematic-PBMCs were isolated from OAC patients (A) and expanded ex vivo for 7 days using anti-CD3/28 +IL-2 T cell activation protocol (B) and further cultured for 48 h under increasing acidic conditions in the absence or presence of immune checkpoint blockade (nivolumab, ipilimumab or dual nivolumab + ipilimumab) (C). Immunophenotyping was then carried out to assess immune checkpoint expression profiles and anti-tumour T cell phenotypes (D). Serum lactate was assessed in OAC patients (E-F) and levels were correlated with patient demographics (G) and the levels of circulating immune/pro-angiogenic cytokines that were determined by multiplex ELISA (H). Key Findings-severe acidic conditions upregulated multiple immune checkpoints on T cells (I). Efficacy of ICB was curtailed under severe acidic conditions (J). Circulating lactate levels positively correlated with circulating levels of pro-angiogenic factor tie-2 and higher serum lactate levels were found in patients who had nodal metastasis (K).


Subject(s)
Adenocarcinoma , T-Lymphocytes , Humans , T-Lymphocytes/metabolism , Ipilimumab/therapeutic use , Nivolumab/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Angiogenesis Inducing Agents/therapeutic use , Adenocarcinoma/pathology
2.
Allergol. immunopatol ; 50(4): 57-63, jul. 2022. graf
Article in English | IBECS | ID: ibc-208894

ABSTRACT

The increased proliferation and extracellular matrix (ECM) production of airway smooth muscle cells (ASMCs) are crucial factors in asthma progression. JNJ0966, one of the metal-loproteinase-9 (MMP-9)-specific inhibitors, has been demonstrated to be involved in the pro-gression and development of diversified diseases. Nevertheless, the function of JNJ0966 in ASMCs remains unclear. This study aimed at investigating the effects of JNJ0966 on asthma progression. In our study, the platelet-derived growth factor BB (PDGF-BB) was first utilized to stimulate the cell model for asthma. Results demonstrated that the cell viability of ASMCs was increased by PDGF-BB (0, 10, 20, and 30 ng/mL) in a dose-dependent manner. Further investigation revealed that JNJ0966 inhibited the cell activity and migration ability of PDGF-BB-induced ASMCs. In addition, JNJ0966 relieved ECM deposition in PDGF-BB-induced ASMCs. Finally, through rescue assays, the results showed that overexpression of MMP-9 reversed the inhibitory effects of JNJ0966 on cell viability and ECM deposition in ASMCs. In conclusion, our findings suggested that JNJ0966 inhibited PDGF-BB-induced ASMC proliferation and ECM pro-duction by modulating MMP-9. These findings might provide novel insight for the treatment of asthm (AU)


Subject(s)
Humans , Disease Progression , Asthma/metabolism , Asthma/drug therapy , Becaplermin/therapeutic use , Angiogenesis Inducing Agents/therapeutic use , Matrix Metalloproteinase 9/metabolism , Cell Proliferation , Myocytes, Smooth Muscle/physiology
3.
Curr Treat Options Oncol ; 23(6): 864-886, 2022 06.
Article in English | MEDLINE | ID: mdl-35412196

ABSTRACT

OPINION STATEMENT: Medulloblastoma (MB) is the most frequent pediatric brain tumor. Despite conventional therapy, MB patients have high mortality and morbidity rates mainly due to the incomplete understanding of the molecular and cellular processes involved in development of this cancer. Similar to other solid tumors, MB demonstrated high endothelial cell proliferation and angiogenic activity, wherein new blood vessels arise from the pre-existing vasculature, a process named angiogenesis. MB angiogenesis is considered a hallmark for MB development, progression, and metastasis emphasizing its potential target for antitumor therapy. However, angiogenesis is tightly regulated by a set of angiogenic factors making it a complex process to be targeted. Although agents targeting these factors and their receptors are early in development, the potential for their targeting may translate into improvement in the clinical care for MB patients. In this review, we focus on the most potent angiogenic factors and their corresponding receptors, highlighting their basic properties and expression in MB. We describe their contribution to MB tumorigenesis and angiogenesis and the potential therapeutic targeting of these factors.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Angiogenesis Inducing Agents/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/etiology , Child , Humans , Medulloblastoma/drug therapy , Medulloblastoma/etiology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism
4.
Bioengineered ; 13(3): 7340-7350, 2022 03.
Article in English | MEDLINE | ID: mdl-35259055

ABSTRACT

The objective of the research was to investigate action mechanism of oxidative stress and cerebral injuries after subarachnoid hemorrhage (SAH) by Ghrelin and angiogenic factor G-patch and FHA domain 1 (Aggf1) and offer new research ideas to SAH clinical treatment and SAH-induced early cerebral injuries. SAH rat models were prepared by prechiasmatic anterior cistern injection. Specific Ghrelin and Aggf1 small interfering ribonucleic acid (siRNA) were designed and injected into silence Ghrelin or Aggf1 in rat left lateral ventricles. Rats were divided randomly into sham-operated (sham), SAH model, negative control siRNA, Ghrelin silence (Ghrelin(-/-)), and Aggf1 silence groups. Changes of rat neurological impairment, encephaledema, cerebral tissue phosphorylated protein kinase (p-Akt), and content changes of caspase-3 protein and oxidative stress indexes were observed, including glutathione (GSH) and oxidized glutathione (GSSG). Results showed scores of neurological impairment and water content in SAH model group were reduced compared with sham group, while p-Akt protein and GSH contents were enhanced. However, caspase-3 protein and GSSG contents were declined, showing statistically meaningful difference (P < 0.05). Compared with SAH model group, scores of neurological impairment, cerebral tissue water content, and caspase-3 protein and GSSG contents in silence Ghrelin and Aggf1 groups were increased, while p-Akt protein and GSH contents were decreased, demonstrating statistically meaningful difference (P < 0.05). To conclude, silence Ghrelin and Aggf1 aggravated early cerebral injuries after SAH, revealing that Ghrelin and Aggf1 could protect brains to some degree.


Subject(s)
Subarachnoid Hemorrhage , Angiogenesis Inducing Agents/therapeutic use , Animals , Apoptosis , Caspase 3/genetics , Caspase 3/metabolism , Disease Models, Animal , Ghrelin/genetics , Ghrelin/pharmacology , Ghrelin/therapeutic use , Glutathione Disulfide , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/complications , Water
5.
Sci Adv ; 8(9): eabm1896, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35235351

ABSTRACT

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.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Retinal Neovascularization , Angiogenesis Inducing Agents/therapeutic use , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/etiology , Diabetic Retinopathy/metabolism , Humans , Mice , Neovascularization, Pathologic , Plasminogen Activator Inhibitor 1/genetics , Retinal Neovascularization/etiology , Vascular Endothelial Growth Factor A/metabolism
6.
Oncogene ; 41(2): 159-172, 2022 01.
Article in English | MEDLINE | ID: mdl-34716430

ABSTRACT

Pseudogenes are generally considered "junk" DNA or "genomic fossils" generated during the evolution process that lack biological activity. However, accumulating reports indicate that pseudogenes have biological functions critical for cancer development. Experiments from the current study showed marked overexpression of the cytidine monophospho-N-acetylneuraminic acid hydroxylase pseudogene (CMAHP) in gastric cancer, which was associated with poor overall survival. However, the mechanisms underlying the activity of CMAHP in tumor development are largely unknown. Gene Set Enrichment Analysis (GSEA) revealed that CMAHP-correlated genes are significantly involved in epithelial-mesenchymal transition (EMT) and angiogenesis. Functional studies further confirmed that CMAHP mediates metastasis and angiogenesis in vitro and in vivo. Furthermore, CMAHP promoted cancer cell migration, invasion, and metastasis through Snail overexpression, which decreased ubiquitination mediated by NF-κB signaling. Angiogenesis is known to be induced by granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulation. CMAHP increased GM-CSF transactivation via promoting direct binding of c-Jun to the -1981/-1975 region of the GM-CSF promoter. Notably, CMAHP interacts with Histone H1.4 promoting histone acetylation to enhance c-Jun and RelA (p65) expression. Our collective findings provide novel evidence that CMAHP contributes to tumor progression and modulates metastasis and angiogenesis in gastric cancer.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Stomach Neoplasms/genetics , Ubiquitination/drug effects , Angiogenesis Inducing Agents/pharmacology , Cell Line, Tumor , Humans , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Analysis
7.
Angiology ; 73(4): 299-311, 2022 04.
Article in English | MEDLINE | ID: mdl-34541892

ABSTRACT

Peripheral arterial disease can involve tissue loss in up to 50% of patients with diabetic foot syndrome (DFS). Consequently, revascularization of narrowed or occluded arteries is one of the most common forms of comprehensive treatment. However, technically successful angioplasty does not always result in the healing of ulcers. The pathomechanism of this phenomenon is still not fully understood, but inadequate angiogenesis in tissue repair may play an essential role. Changes in pro- and anti-angiogenic factors among patients with DFS are not always clear and conclusive. In particular, some studies underline the role of decreased concentration of pro-angiogenic factors and higher levels of anti-angiogenic mediators. Nevertheless, there are still controversial issues, including the paradox of impaired wound healing despite high concentrations of some pro-angiogenic factors, dynamics of their expression during the healing process, and their mutual relationships. Exploring this process among diabetic patients may provide new insight into well-known methods of treatment and show their real benefits and chances for improving outcomes.


Subject(s)
Diabetes Mellitus , Diabetic Foot , Peripheral Arterial Disease , Angiogenesis Inducing Agents/therapeutic use , Humans , Peripheral Arterial Disease/therapy , Vascular Surgical Procedures , Wound Healing
8.
Cardiovasc Res ; 118(1): 53-64, 2022 01 07.
Article in English | MEDLINE | ID: mdl-33620071

ABSTRACT

It is well established that the vasculature plays a crucial role in maintaining oxygen and nutrients supply to the heart. Increasing evidence further suggests that the microcirculation has additional roles in supporting a healthy microenvironment. Heart failure is well known to be associated with changes and functional impairment of the microvasculature. The specific ablation of protective signals in endothelial cells in experimental models is sufficient to induce heart failure. Therefore, restoring a healthy endothelium and microcirculation may be a valuable therapeutic strategy to treat heart failure. This review article will summarize the current understanding of the vascular contribution to heart failure with reduced or preserved ejection fraction. Novel therapeutic approaches including next generation pro-angiogenic therapies and non-coding RNA therapeutics, as well as the targeting of metabolites or metabolic signalling, vascular inflammation and senescence will be discussed.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Coronary Vessels/drug effects , Genetic Therapy , Heart Failure, Diastolic/therapy , Heart Failure, Systolic/therapy , Microvessels/drug effects , Neovascularization, Physiologic/drug effects , Vaccines/therapeutic use , Angiogenesis Inducing Agents/adverse effects , Animals , Coronary Circulation/drug effects , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Genetic Therapy/adverse effects , Heart Failure, Diastolic/genetics , Heart Failure, Diastolic/metabolism , Heart Failure, Diastolic/physiopathology , Heart Failure, Systolic/genetics , Heart Failure, Systolic/metabolism , Heart Failure, Systolic/physiopathology , Humans , Microcirculation/drug effects , Microvessels/metabolism , Microvessels/physiopathology , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Recovery of Function , Vaccines/adverse effects , Ventricular Function, Left/drug effects
9.
Leuk Lymphoma ; 62(13): 3160-3169, 2021 12.
Article in English | MEDLINE | ID: mdl-34806520

ABSTRACT

Angiogenesis is in a constant balance between pro and anti-angiogenic factors. Neoangiogenesis, implicated in metastatic spreading is characterized in solid cancers, but fairly new in chronic lymphocytic leukemia (CLL). We hypothesize that secretion of angiogenic factors could be correlated to the pathogenesis of CLL, and therefore predict the outcome of patients. We investigated concentrations of 22 cytokines and chemokines in 137 non-del 17p B-CLL patients, treated with a fludarabine-cyclophosphamide-rituximab (FCR)-based regimen. We constructed a biomarker index defining different risk groups based on lymphocyte count, the intensity of CD20 antigen on CD19+ cells, Ang-2, and PDGF-BB plasma concentrations at diagnosis. Four groups were defined, exhibiting specific molecular signatures and correlated with progression-free survival of patients. Our results suggest that we can determine at diagnosis of non-del 17p B-CLL patients, those with a very high probability of progression-free survival, independently of IGVH mutational status and residual disease at the end of treatment.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Angiogenesis Inducing Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cyclophosphamide/therapeutic use , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Neoplasm, Residual/etiology , Rituximab/therapeutic use , Vidarabine/therapeutic use
10.
Eur J Pharmacol ; 913: 174642, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34822791

ABSTRACT

It is widely accepted that the stria vascularis (SV) in cochlea plays a critical role in the generation of endocochlear potential (EP) and the secretion of the endolymph. 17ß-estradiol (E2) is the most potent and abundant endogenous estrogen during the premenopausal period, thus, considered as the reference estrogen. This study aimd to investigate the protective effect of E2 by promoting the expression of vascular endothelial growth factor (VEGF) and thus promoting the vascular regeneration of the SV in elderly mice. After being treated with E2 either in vivo or in vitro, the hearing threshold changes of C57BL/6J elder mice continuously reduced, endothelial cell morphology improved, the number of endothelial cells (ECs) tubular nodes increased significantly, the ability of tubular formation enhanced significantly and the expression of VEGF increased. In vitro, cell model in conjunction with in vivo ovariectomized model was established to demonstrate for the first time that E2 promotes angiogenesis by promoting the secretion of VEGF through the phosphatidylinositol 3-kinase (PI3K)/AKT pathway (PI3K/AKT). In conclusion, E2 demonstrated potent angiogenesis properties with significant protection against Age-Related Hearing Loss (ARHL), which provides a new idea for the improvement of ARHL.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Estradiol/pharmacology , Hearing Loss/prevention & control , Neovascularization, Physiologic/drug effects , Stria Vascularis/drug effects , Aging/physiology , Angiogenesis Inducing Agents/therapeutic use , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Estradiol/therapeutic use , Female , Hearing Loss/physiopathology , Humans , Mice , Organ Culture Techniques , Regeneration/drug effects , Signal Transduction/drug effects , Stria Vascularis/physiology , Vascular Endothelial Growth Factor A/agonists , Vascular Endothelial Growth Factor A/metabolism
11.
Oxid Med Cell Longev ; 2021: 7624389, 2021.
Article in English | MEDLINE | ID: mdl-34691360

ABSTRACT

The development of psoriasis is associated with the consequences of oxidative stress and inflammation leading to metabolic changes locally, in the skin cells, and systemically, in the blood. Therefore, the aim of this study was to analyze the effect of psoriasis vulgaris (PsV) and psoriatic arthritis (PsA) on the basal plasma/keratinocyte levels of matrix metalloproteinases (MMPs), tissue inhibitors of matrix metalloproteinases (TIMPs), and angiogenesis factors, as well as to evaluate the effect of CBD on these parameters in keratinocytes isolated from psoriatic/healthy individuals with and without in vitro irradiation by UVB. A quantitative chemiluminescent method of detection based on an ELISA protocol and zymography technique was used during analysis. It was shown that activity levels of MMP-9 and TIMP-2 in PsA plasma were higher than in PsV. Changes in the proteolytic activity were accompanied by an increase in markers of angiogenesis (angiopoietin-2, HGF, VEGF, TNFα, PDGF, FGF), where in the specific case of angiopoietin-2 and TNFα, the overexpression in PsV was significantly stronger than in PsA. CBD application to keratinocytes partially restored levels of MMP-1/2/3/7 and TIMP-1/2 (in an effect which was particularly enhanced by UVB irradiation), as well as levels of the examined angiogenic factors except TNFα (levels of which were increased in psoriatic keratinocytes and decreased in healthy keratinocytes). Presented results indicate that CBD may be suggested as an antiangiogenic factor that reduces the proinflammatory action of UVB in psoriatic keratinocytes and partially has a protective effect for healthy keratinocytes.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Cannabidiol/therapeutic use , Keratinocytes/drug effects , Matrix Metalloproteinases/drug effects , Psoriasis/drug therapy , Adult , Angiogenesis Inducing Agents/pharmacology , Cannabidiol/pharmacology , Case-Control Studies , Female , Humans , Male
12.
Arch. Soc. Esp. Oftalmol ; 96(9): 455-461, sept. 2021. ilus, tab
Article in Spanish | IBECS | ID: ibc-218027

ABSTRACT

Introducción y objetivos La degeneración macular asociada con la edad (DMAE) es la causa primaria de ceguera en los países desarrollados, especialmente en adultos mayores. Actualmente, la inyección intravítrea del factor de crecimiento endotelial vascular (VEGF) es el tratamiento estándar para la forma neovascular de la DMAE. Existen pocos estudios que informen sobre la creación de un agujero macular (AM) después de un tratamiento anti-VEGF, y la patogénesis exacta de AM permanece en debate. El presente estudio tiene por objetivo analizar las características clínicas de los ojos que desarrollan AM después de recibir terapia anti-VEGF para la DMAE neovascular. Materiales y métodos Los pacientes fueron tratados con agentes anti-VEGF intravítreos durante al menos un año, permaneciendo estables por, al menos, seis meses. Se evaluaron la mejor agudeza visual corregida (MAVC) y los hallazgos de tomografía de coherencia óptica. Resultado Se incluyeron en el estudio 19 ojos de 18 pacientes. La edad media de los mismos fue de 77,7 años en la primera visita. Ocho eran de sexo femenino. El número medio de inyecciones antes de la formación de un AM fue de cuatro. El AM se desarrolló después de un seguimiento medio de 5,1 meses desde la última inyección. Dieciséis ojos (84,2%) exhibieron membrana coroidal neovascular sin tracción vitreomacular anormal. Once ojos (57,8%) mostraron desprendimiento del epitelio pigmentario (DEP) de la retina, dos (10,5%) tuvieron membrana epirretinal (MER) y uno (5,2%) presentó desgarro del epitelio pigmentario de la retina (PER). La media de la MAVC fue de 1,07 ± 0,48 LogMAR (0,3 a 1,8) y 1,16 ± 0,38 logMAR (0,4 a 1,8), respectivamente (AU)


Introduction and objectives Age-related macular degeneration (AMD) is the primary cause of blindness in developed countries, particularly in older adults. Anti-vascular endothelial growth factor (anti-VEGF) intravitreal injection is the current standard treatment for neovascular form of AMD. Studies reporting macular hole (MH) formation following anti-VEGF treatment are limited, and the exact pathogenesis is still under discussion. With the present study, we aim to analyse the clinical features of eyes developing MH after anti-VEGF therapy for neovascular AMD. Materials and methods Patients were treated with intravitreal anti-VEGF agents for at least one year and stable for at least six months. Best-corrected visual acuity (BCVA) and optical coherence tomography findings were evaluated. Results Nineteen eyes of 18 patients were included in this study. Patients had an average age of 77.7 years at first visit and eight were female. The average number of injections before the MH formation was four. MH developed after a mean follow-up of 5.1 months after the last injection. Sixteen eyes had (84.2%) had choroidal neovascular membrane without any abnormal vitreomacular traction. Eleven eyes (57.8%) had retinal pigment epithelium detachment (PED), two (10.5%) had an epiretinal membrane (ERM), and one (5.2%) had retinal pigment epithelium (RPE) tear. The mean first and last BCVA was 1.07 ± 0.48 LogMAR (0.3-1.8) and 1.16 ± 0.38 logMAR (0.4-1.8), respectively. Conclusions A macular hole can be observed in AMD patients receiving anti-VEGF therapy. Increased fibrovascular scar tissue due to subretinal fluid resolution, neovascular membrane contraction, and the presence of PED, RPE tear, and ERM may contribute to MH formation (AU)


Subject(s)
Humans , Male , Female , Aged , Aged, 80 and over , Angiogenesis Inducing Agents/adverse effects , Angiogenesis Inducing Agents/therapeutic use , Retinal Perforations/chemically induced , Macular Degeneration/drug therapy , Retrospective Studies , Fluorescein Angiography , Retinal Pigments , Vascular Endothelial Growth Factor A , Visual Acuity
13.
Circ Res ; 128(12): 1944-1957, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34110899

ABSTRACT

The prevalence of peripheral arterial disease (PAD) in the United States exceeds 10 million people, and PAD is a significant cause of morbidity and mortality across the globe. PAD is typically caused by atherosclerotic obstructions in the large arteries to the leg(s). The most common clinical consequences of PAD include pain on walking (claudication), impaired functional capacity, pain at rest, and loss of tissue integrity in the distal limbs that may lead to lower extremity amputation. Patients with PAD also have higher than expected rates of myocardial infarction, stroke, and cardiovascular death. Despite advances in surgical and endovascular procedures, revascularization procedures may be suboptimal in relieving symptoms, and some patients with PAD cannot be treated because of comorbid conditions. In some cases, relieving obstructive disease in the large conduit arteries does not assure complete limb salvage because of severe microvascular disease. Despite several decades of investigational efforts, medical therapies to improve perfusion to the distal limb are of limited benefit. Whereas recent studies of anticoagulant (eg, rivaroxaban) and intensive lipid lowering (such as PCSK9 [proprotein convertase subtilisin/kexin type 9] inhibitors) have reduced major cardiovascular and limb events in PAD populations, chronic ischemia of the limb remains largely resistant to medical therapy. Experimental approaches to improve limb outcomes have included the administration of angiogenic cytokines (either as recombinant protein or as gene therapy) as well as cell therapy. Although early angiogenesis and cell therapy studies were promising, these studies lacked sufficient control groups and larger randomized clinical trials have yet to achieve significant benefit. This review will focus on what has been learned to advance medical revascularization for PAD and how that information might lead to novel approaches for therapeutic angiogenesis and arteriogenesis for PAD.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Peripheral Arterial Disease/therapy , Adult Stem Cells/transplantation , Amputation, Surgical , Angiogenesis Modulating Agents/therapeutic use , Animals , Anticoagulants/therapeutic use , Atherosclerosis/complications , Endothelium, Vascular/metabolism , Endovascular Procedures , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Intermittent Claudication/etiology , Limb Salvage , Lower Extremity/blood supply , Lower Extremity/surgery , Mice , Microcirculation , Myocardial Infarction/epidemiology , Neovascularization, Physiologic/physiology , Peripheral Arterial Disease/epidemiology , Prevalence , Proprotein Convertase 9 , RNA, Untranslated/therapeutic use , Stroke/epidemiology
14.
Pharm Biol ; 59(1): 584-593, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34010584

ABSTRACT

CONTEXT: Cerebral ischaemia/reperfusion (I/R) injury has a high disability and fatality worldwide. Myrtenol has protective effects on myocardial I/R injury through antioxidant and anti-apoptotic effects. OBJECTIVE: This study investigated the effect of myrtenol on cerebral ischaemia/reperfusion (I/R) injury and the underlying mechanism. MATERIALS AND METHODS: Cerebral I/R injury was induced in adult Sprague-Dawley rats by middle cerebral artery occlusion (MCAO) for 90 min. MCAO rats were treated with or without myrtenol (10, 30, or 50 mg/kg/day) or/and U0126 (10 µL) intraperitoneally for 7 days. RESULTS: In the present study, myrtenol had no toxicity at concentrations up to 1.3 g/kg. Myrtenol treatment improved neurological function of MCAO rats, with significantly (p < 0.05) improved neurological deficits (4.31 ± 1.29 vs. 0.00) and reduced brain edoema (78.95 ± 2.27% vs. 85.48 ± 1.24%). Myrtenol extenuated brain tissue injury and neuronal apoptosis, with increased Bcl-2 expression (0.48-fold) and decreased Bax expression (2.02-fold) and caspase-3 activity (1.36-fold). Myrtenol promoted angiogenesis in the brain tissues of MCAO rats, which was reflected by increased VEGF (0.86-fold) and FGF2 (0.51-fold). Myrtenol promoted the phosphorylation of MEK1/2 (0.80-fold) and ERK1/2 (0.97-fold) in MCAO rats. U0126, the inhibitor of ERK1/2 pathway, reversed the protective effects of myrtenol on brain tissue damage and angiogenesis in MCAO rats. DISCUSSION AND CONCLUSIONS: Myrtenol reduced brain damage and angiogenesis through activating the ERK1/2 signalling pathway, which may provide a novel alternative strategy for preventing cerebral I/R injury. Further in vitro work detailing its mechanism-of-action for improving ischaemic cerebral infarction is needed.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Bicyclic Monoterpenes/therapeutic use , Cerebral Infarction/drug therapy , MAP Kinase Signaling System/drug effects , Reperfusion Injury/drug therapy , Angiogenesis Inducing Agents/pharmacology , Animals , Bicyclic Monoterpenes/pharmacology , Cerebral Infarction/metabolism , Cerebral Infarction/pathology , Dose-Response Relationship, Drug , MAP Kinase Signaling System/physiology , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
15.
Hum Exp Toxicol ; 40(11): 1867-1879, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33896237

ABSTRACT

Some traditional Chinese decoctions, such as Zhuyu Annao, exert favorable therapeutic effects on acute cerebral hemorrhage, hemorrhagic stroke, and other neurological diseases, but the underlying mechanism remains unclear. This study aimed to determine whether Zhuyu Annao decoction (ZYAND) protects the injured brain by promoting angiogenesis following intracerebral hemorrhage (ICH) and elucidate its specific mechanism. The effect of ZYAND on the nervous system of mice after ICH was explored through behavioral experiments, such as the Morris water maze and Rotarod tests, and its effects on oxidative stress were explored by detecting several oxidative stress markers, including malondialdehyde, nitric oxide, glutathione peroxidase, and superoxide dismutase. Real-time quantitative RT-PCR and WB were used to detect the effects of ZYAND on the levels of prolyl hydroxylase domain 3 (PHD3), hypoxia-inducible factor-1α (HIF-1α), and vascular endothelial growth factor (VEGF) in the brain tissues of mice. The effect of ZYAND on the NF-κB signaling pathway was detected using a luciferase reporter gene. A human umbilical cord vascular endothelial cell angiogenesis experiment was performed to determine whether ZYAND promotes angiogenesis. The Morris water maze test and other behavioral experiments verified that ZYAND improved the neurobehavior of mice after ICH. ZYAND activated the PHD3/HIF-1α signaling pathway, inhibiting the oxidative damage caused by ICH. In angiogenesis experiments, it was found that ZYAND promoted VEGF-induced angiogenesis by upregulating the expression of HIF-1α, and NF-κB signaling regulated the expression of HIF-1α by inhibiting PHD3. ZYAND exerts a reparative effect on brain tissue damaged after ICH through the NF-κB/ PHD3/HIF-1α/VEGF signaling axis.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Cerebral Hemorrhage/drug therapy , Enzyme Inhibitors/metabolism , Medicine, Chinese Traditional/methods , Plant Extracts/therapeutic use , Procollagen-Proline Dioxygenase/drug effects , Procollagen-Proline Dioxygenase/metabolism , Animals , China , Disease Models, Animal , Humans , Mice
16.
J Ethnopharmacol ; 273: 113824, 2021 Jun 12.
Article in English | MEDLINE | ID: mdl-33581257

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Buxuhuayu decoction (BXHYD) has been frequently used to treat patients with diabetic ulcers (DUs), without notable adverse reactions. However, the related molecular mechanism remains unelucidated. AIM OF THE STUDY: This study assessed the potential mechanism of BXHYD against DUs by using network pharmacology and animal experiments. MATERIALS AND METHODS: First, high-performance liquid chromatography (HPLC) was used for quality control of BXHYD. Further, the hub compounds and targets were screened from the Active Compound-Targets (ACT) network and the protein and protein interaction (PPI) network. Enrichment analysis was performed using DAVID, and molecular docking technology was used to identify active compounds that may play a key role in pub targets. Finally, a DUs animal model was established and used to elucidate the effect of BXHYD on the PI3K/Akt/eNOS signalling pathway. RESULTS: (1) Calycosin-7-glucoside, amygdalin, and tanshinone iiA were detected in the freeze-dried powder of BXHYD. (2) Twelve hub compounds and eight hub targets were screened using the ACT and PPI networks. Through molecular docking, it was found that the four hub targets (TP53, IL6, VEGFA, and AKT1) binds luteolin and quercetin more tightly. (3) BXHYD is most likely to promote angiogenesis and wound healing by activating the PI3K/Akt/eNOS signalling pathway. CONCLUSIONS: This research revealed that BXHYD might activate the PI3K/Akt/eNOS signalling pathway to promote DUs healing. These findings support the clinical use of BXHYD and provide the foundation for its subsequent studies.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Neovascularization, Physiologic/drug effects , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ulcer/drug therapy , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/therapeutic use , Angiogenesis Inducing Agents/toxicity , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/toxicity , Male , Medicine, Chinese Traditional , Molecular Docking Simulation , Protein Interaction Maps , Rats, Sprague-Dawley , Signal Transduction/drug effects , Streptozocin , Ulcer/etiology , Ulcer/pathology , Wound Healing/drug effects
17.
Biofactors ; 47(3): 270-291, 2021 May.
Article in English | MEDLINE | ID: mdl-33606893

ABSTRACT

Finding effective treatments for cardiac diseases is among the hottest subjects in medicine; cell-based therapies have brought great promises for managing a broad range of life-threatening heart complications such as myocardial infarction. After clarifying the critical role of angiogenesis in tissue repair and regeneration, various stem/progenitor cell were utilized to accelerate the healing of injured cardiac tissue. Embryonic, fetal, adult, and induced pluripotent stem cells have shown the appropriate proangiogenic potential for tissue repair strategies. The capability of stem cells for differentiating into endothelial lineages was initially introduced as the primary mechanism involved in improving angiogenesis and accelerated heart tissue repair. However, recent studies have demonstrated the leading role of paracrine factors secreted by stem cells in advancing neo-vessel formation. Genetically modified stem cells are also being applied for promoting angiogenesis regarding their ability to considerably overexpress and secrete angiogenic bioactive molecules. Yet, conducting further research seems necessary to precisely identify molecular mechanisms behind the proangiogenic potential of stem cells, including the signaling pathways and regulatory molecules such as microRNAs. In conclusion, stem cells' pivotal roles in promoting angiogenesis and consequent improved cardiac healing and remodeling processes should not be ignored, especially in the case of stem cell-derived extracellular vesicles.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Exosomes/metabolism , Heart Diseases/therapy , Stem Cell Transplantation/methods , Animals , Disease Models, Animal , Heart Diseases/metabolism , Rats , Zebrafish
18.
J Biomed Mater Res A ; 109(4): 453-478, 2021 04.
Article in English | MEDLINE | ID: mdl-32985051

ABSTRACT

Skin injuries and in particular, chronic wounds, are one of the major prevalent medical problems, worldwide. Due to the pivotal role of angiogenesis in tissue regeneration, impaired angiogenesis can cause several complications during the wound healing process and skin regeneration. Therefore, induction or promotion of angiogenesis can be considered as a promising approach to accelerate wound healing. This article presents a comprehensive overview of current and emerging angiogenesis induction methods applied in several studies for skin regeneration, which are classified into the cell, growth factor, scaffold, and biological/chemical compound-based strategies. In addition, the advantages and disadvantages of these angiogenic strategies along with related research examples are discussed in order to demonstrate their potential in the treatment of wounds.


Subject(s)
Neovascularization, Physiologic , Skin/blood supply , Tissue Engineering/methods , Wound Healing , Angiogenesis Inducing Agents/therapeutic use , Animals , Biocompatible Materials/therapeutic use , Humans , Neovascularization, Physiologic/drug effects , Regeneration/drug effects , Skin/drug effects , Skin Physiological Phenomena/drug effects , Wound Healing/drug effects
19.
Cardiovasc Res ; 117(5): 1257-1273, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33063086

ABSTRACT

Acute myocardial infarction (MI) inflicts massive injury to the coronary microcirculation leading to vascular disintegration and capillary rarefication in the infarct region. Tissue repair after MI involves a robust angiogenic response that commences in the infarct border zone and extends into the necrotic infarct core. Technological advances in several areas have provided novel mechanistic understanding of postinfarction angiogenesis and how it may be targeted to improve heart function after MI. Cell lineage tracing studies indicate that new capillary structures arise by sprouting angiogenesis from pre-existing endothelial cells (ECs) in the infarct border zone with no meaningful contribution from non-EC sources. Single-cell RNA sequencing shows that ECs in infarcted hearts may be grouped into clusters with distinct gene expression signatures, likely reflecting functionally distinct cell populations. EC-specific multicolour lineage tracing reveals that EC subsets clonally expand after MI. Expanding EC clones may arise from tissue-resident ECs with stem cell characteristics that have been identified in multiple organs including the heart. Tissue repair after MI involves interactions among multiple cell types which occur, to a large extent, through secreted proteins and their cognate receptors. While we are only beginning to understand the full complexity of this intercellular communication, macrophage and fibroblast populations have emerged as major drivers of the angiogenic response after MI. Animal data support the view that the endogenous angiogenic response after MI can be boosted to reduce scarring and adverse left ventricular remodelling. The improved mechanistic understanding of infarct angiogenesis therefore creates multiple therapeutic opportunities. During preclinical development, all proangiogenic strategies should be tested in animal models that replicate both cardiovascular risk factor(s) and the pharmacotherapy typically prescribed to patients with acute MI. Considering that the majority of patients nowadays do well after MI, clinical translation will require careful selection of patients in need of proangiogenic therapies.


Subject(s)
Endothelial Progenitor Cells/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Neovascularization, Physiologic , Regeneration , Angiogenesis Inducing Agents/therapeutic use , Angiogenic Proteins/metabolism , Animals , Autocrine Communication , Cell Lineage , Cell Movement , Cell Proliferation , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Humans , Inflammation Mediators/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Neovascularization, Physiologic/drug effects , Paracrine Communication , Phenotype , Recovery of Function , Signal Transduction
20.
Sci Rep ; 10(1): 16656, 2020 10 07.
Article in English | MEDLINE | ID: mdl-33028854

ABSTRACT

Nerve injury-induced protein 1 (Ninjurin 1, Ninj1) is a cell adhesion molecule responsible for cell-to-cell interactions between immune cells and endothelial cells. In our previous paper, we have shown that Ninj1 plays an important role in the infiltration of neutrophils in the postischemic brain and that the dodecamer peptide harboring the Ninj1 N-terminal adhesion motif (N-NAM, Pro26-Asn37) inhibits infiltration of neutrophils in the postischemic brain and confers robust neuroprotective and anti-inflammatory effects. In the present study, we examinedt the pro-angiogenic effect of N-NAM using human umbilical vein endothelial cells (HUVECs) and rat MCAO (middle cerebral artery occlusion) model of stroke. We found that N-NAM promotes proliferation, migration, and tube formation of HUVECs and demonstrate that the suppression of endogenous Ninj1 is responsible for the N-NAM-mediated pro-angiogenic effects. Importantly, a pull-down assay revealed a direct binding between exogenously delivered N-NAM and endogenous Ninj1 and it is N-terminal adhesion motif dependent. In addition, N-NAM activated the Ang1-Tie2 and AKT signaling pathways in HUVECs, and blocking those signaling pathways with specific inhibitors suppressed N-NAM-induced tube formation, indicating critical roles of those signaling pathways in N-NAM-induced angiogenesis. Moreover, in a rat MCAO model, intranasal administration of N-NAM beginning 4 days post-MCAO (1.5 µg daily for 3 days) augmented angiogenesis in the penumbra of the ipsilateral hemisphere of the brain and significantly enhanced total vessel lengths, vessel densities, and pro-angiogenic marker expression. These results demonstrate that the 12-amino acid Ninj1 peptide, which contains the N-terminal adhesion motif of Ninj1, confers pro-angiogenic effects and suggest that those effects might contribute to its neuroprotective effects in the postischemic brain.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Cell Adhesion Molecules, Neuronal/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Ischemic Stroke/drug therapy , Neovascularization, Physiologic/drug effects , Nerve Growth Factors/pharmacology , Neuroprotective Agents/pharmacology , Angiogenesis Inducing Agents/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemic Stroke/metabolism , Nerve Growth Factors/metabolism , Neuroprotective Agents/therapeutic use , Neutrophils/drug effects , Neutrophils/metabolism , Rats , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...