Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Clin Case Rep ; 11(11): e8224, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38028083

ABSTRACT

Key Clinical Message: Tuberculous myocarditis is a rare presentation of tuberculosis, posing diagnostic challenges in endemic countries. Clinicians should consider this entity in patients with unexplained heart failure, conduction abnormalities, or sudden cardiac events in tuberculosis-endemic regions. Abstract: Tuberculous myocarditis is an uncommon manifestation of tuberculosis, often presenting as a diagnostic challenge, particularly in tuberculosis-endemic regions. We report a case of a 58-year-old male with a history of chronic cough and fever, who presented with progressive dyspnea, generalized body swelling, and New York Heart Association (NYHA) Class IV heart failure. Clinical examination revealed signs of cardiac decompensation and congestive heart failure. Emergency echocardiography demonstrated biventricular dysfunction, and imaging showed clots in both atria and the left ventricle. The patient responded well to initial treatment with anticoagulants, antibiotics, diuretics, and inotropic support. Subsequent investigations, including computed tomography pulmonary angiogram (CTPA) and high-resolution computed tomography (HRCT), confirmed active pulmonary tuberculosis. Anti-tuberculous treatment (ATT) was initiated, and the patient showed remarkable improvement. The diagnosis of tuberculous myocarditis was based on clinical, radiological, and laboratory evidence, as cardiac biopsy was not performed due to resource limitations. Tuberculous myocarditis is an underreported condition, and clinicians should be vigilant about its occurrence, especially in tuberculosis-endemic regions. Early recognition and prompt initiation of ATT can lead to favorable outcomes. This case highlights the importance of considering tuberculous myocarditis in patients with unexplained heart failure or cardiac abnormalities in areas with a high burden of tuberculosis.

2.
Front Cardiovasc Med ; 10: 1156474, 2023.
Article in English | MEDLINE | ID: mdl-37273881

ABSTRACT

Cardiac Sarcoidosis (CS) is a deadly consequence of systemic sarcoidosis that inflames all three layers of the heart, especially the myocardium-clinical signs of CS range from asymptomatic disease to abrupt cardiac death. CS generally remains undiagnosed secondary to a lack of definitive diagnostic criteria, a high percentage of false negative results on endomyocardial biopsy, and ill-defining clinical manifestations of the disease. Consequently, there is a lack of evidence-based recommendations for CS, and the present diagnostic and therapeutic management depend on expert opinion. The aetiology, risk factors, clinical symptoms, diagnosis, and therapy of CS will be covered in this review. A particular emphasis will be placed on enhanced cardiovascular imaging and early identification of CS. We review the emerging evidence regarding the use of Electrocardiograms (ECGs), Magnetic Resonance Imaging (MRI), and Positron Emission Tomography (PET) imaging of the heart to identify and quantify the extent of myocardial inflammation, as well as to guide the use of immunotherapy and other treatment regimens, such as ablation therapy, device therapy, and heart transplantation, to improve patient outcomes.

3.
J Periodontol ; 93(11): 1712-1724, 2022 11.
Article in English | MEDLINE | ID: mdl-35536914

ABSTRACT

BACKGROUND: Porphyromonas gingivalis exacerbates tissue hypoxia and worsens periodontal inflammation. This study investigated the effect of a therapeutic oxygen carrier (M101), derived from Arenicola marina, on hypoxia and associated inflammation in the context of periodontitis. METHODS: The effect of M101 on GLUT-1, GLUT-3, HIF-1α, and MMP-9 expression, hypoxia, and antioxidant status in oral epithelial cells (EC) exposed to CoCl2 (1000 µM), P. gingivalis (MOI 100), and CoCl2 + P. gingivalis was evaluated through hypoxia detection fluorescence assay, antioxidant concentration colorimetric assay, and RTqPCR. Evaluation of M101 on EC proliferation was evaluated in an in vitro wound assay. In experimental periodontitis, periodontal wound healing and osteoclastic activity were compared among natural wound healing, placebo, and gels containing M101 (1  and 2 g/L) groups through histomorphometry and TRAP (tartrate-resistant acid phosphatase activity assay) assay respectively. The expression of HIF-1α, MMP-9, and NFκB in periodontal tissues was also evaluated through immunofluorescence studies. RESULTS: M101 downregulated GLUT-1, GLUT-3, HIF-1α, and MMP-9 levels in EC exposed to CoCl2 , P. gingivalis, and CoCl2 + P. gingivalis (p < 0.05). Fluorescence and colorimetric analyses confirmed hypoxia reduction and antioxidant capacity improvement in such EC upon M101 treatment. Moreover, M101 improved significantly the in vitro wound closure. In vivo, the attachment level was significantly improved, and osteoclastic activity was reduced in mice treated with M101 gels compared to placebo and natural wound healing groups (p < 0.05). HIF-1α, MMP-9, and NFκB expression in periodontal tissues was reduced in M101 gels treated mice compared to the controls. CONCLUSION: M101 showed promise in resolving hypoxia and associated inflammation-mediated tissue degradation. Its potential in the clinical management of periodontitis must be further investigated.


Subject(s)
Periodontitis , Porphyromonas gingivalis , Animals , Mice , Porphyromonas gingivalis/metabolism , Matrix Metalloproteinase 9/metabolism , Oxygen/metabolism , Oxygen/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Antioxidants/metabolism , Hypoxia/metabolism , Periodontitis/drug therapy , Periodontitis/metabolism , Inflammation , Wound Healing , NF-kappa B/metabolism
4.
J Vis Exp ; (182)2022 04 13.
Article in English | MEDLINE | ID: mdl-35499333

ABSTRACT

Microvesicles (MVs) are submicron fragments released from the plasma membrane of activated cells that act as proinflammatory and procoagulant cellular effectors. In rats, spleen MVs (SMVs) are surrogate markers of pathophysiological conditions. Previous in vitro studies demonstrated that Porphyromonas gingivalis (P. gingivalis), a major periodontal pathogen, enables the endothelial shedding and apoptosis while lipopolysaccharide (LPS) favors the shedding of splenocyte-derived microvesicles (SMVs). In vivo studies showed the feasibility of pharmacological control of SMV shedding. The present protocol establishes a standardized procedure for isolating splenic SMVs from the P. gingivalis acute murine infection model. P. gingivalis infection was induced in young C57BL/6 mice by intraperitoneal injection (three injections of 5 x 107 bacteria/week). After two weeks, the spleens were collected, weighed, and the splenocytes were counted. SMVs were isolated and quantified by protein, RNA, and prothrombinase assays. Cell viability was assessed by either propidium iodide or trypan blue exclusion dyes. Following splenocyte extraction, neutrophil counts were obtained by flow cytometry after 24 h of splenocyte culture. In P. gingivalis-injected mice, a 2.5-fold increase in spleen weight and a 2.3-fold rise in the splenocyte count were observed, while the neutrophils count was enhanced by 40-folds. The cell viability of splenocytes from P. gingivalis-injected mice ranged from 75%-96% and was decreased by 50% after 24 h of culture without any significant difference compared to unexposed controls. However, splenocytes from injected mice shed higher amounts of MVs by prothrombinase assay or protein measurements. The data demonstrate that the procoagulant SMVs are reliable tools to assess an early spleen response to intraperitoneal P. gingivalis infection.


Subject(s)
Bacterial Infections , Spleen , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Rats , Thromboplastin
5.
Inflammation ; 45(4): 1752-1764, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35274214

ABSTRACT

The aim of this study was to evaluate the potential anti-inflammatory and anti-resorptive effects of lenabasum in the context of Porphyromonas gingivalis (Pg)-induced inflammation. Lenabasum or ajulemic acid (1',1'-dimethylheptyl-THC-11-oic-acid), a synthetic analog of THC-11-oic acid, has already demonstrated anti-inflammatory properties for the treatment of several inflammatory diseases. In vitro, the cytocompatibility of lenabasum was evaluated in human oral epithelial cells (EC), oral fibroblasts and osteoblasts by metabolic activity assay. The effect of lenabasum (5 µM) treatment of Pg-LPS- and P. gingivalis-infected EC on the pro- and anti-inflammatory markers was studied through RTqPCR. In vivo, lenabasum was injected subcutaneously in a P. gingivalis-induced calvarial abscess mouse model to assess its pro-healing effect. Concentrations of lenabasum up to 5 µM were cytocompatible in all cell types. Treatment of Pg-LPS and Pg-infected EC with lenabasum (5 µM; 6 h) reduced the gene expression of TNF-α, COX-2, NF-κB, and RANKL, whereas it increased the expression of IL-10 and resolvin E1 receptor respectively (p < 0.05). In vivo, the Pg-elicited inflammatory lesions' clinical size was significantly reduced by lenabasum injection (30 µM) vs untreated controls (45%) (p < 0.05). Histomorphometric analysis exhibited improved quantity and quality of bone (with reduced lacunae) and significantly reduced calvarial soft tissue inflammatory score in mice treated with lenabasum (p < 0.05). Tartrate-resistant acid phosphatase activity assay (TRAP) also demonstrated decreased osteoclastic activity in the treatment group compared to that in the controls. Lenabasum showed promising anti-inflammatory and pro-resolutive properties in the management of Pg-elicited inflammation, and thus, its potential as adjuvant periodontal treatment should be further investigated.


Subject(s)
Anti-Inflammatory Agents , Porphyromonas gingivalis , Animals , Mice , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Lipopolysaccharides/pharmacology
6.
Dent Clin North Am ; 66(1): 131-155, 2022 01.
Article in English | MEDLINE | ID: mdl-34794551

ABSTRACT

Current periodontal treatments aim to control bacterial infection and decrease inflammation. To optimize contemporary conventional treatments that present limitations owing to an inability to reach the lesion site, new methods are based on nanomedicine. Nanomedecine allows delivery of host-modulatory drugs or antibacterial molecules at the lesion site in an optimal concentration with decreased toxicity and risk of systemic side effects. Chitosan and polylactic-co-glycolic acid-loaded nanoparticles, carbon quantum dots, and mesoporous silicates open new perspectives in periodontitis management. The potential therapeutic impact of the main nanocarriers is discussed.


Subject(s)
Chitosan , Nanoparticles , Periodontitis , Anti-Bacterial Agents/therapeutic use , Humans , Nanomedicine , Periodontitis/therapy
7.
J Tissue Eng ; 12: 20417314211041428, 2021.
Article in English | MEDLINE | ID: mdl-34721831

ABSTRACT

Control of inflammation is indispensable for optimal oral wound healing and tissue regeneration. Several biomaterials have been used to enhance the regenerative outcomes; however, the biomaterial implantation can ensure an immune-inflammatory response. The interface between the cells and the biomaterial surface plays a critical role in determining the success of soft and hard tissue regeneration. The initial inflammatory response upon biomaterial implantation helps in tissue repair and regeneration, however, persistant inflammation impairs the wound healing response. The cells interact with the biomaterials through extracellular matrix proteins leading to protein adsorption followed by recruitment, attachment, migration, and proliferation of several immune-inflammatory cells. Physical nanotopography of biomaterials, such as surface proteins, roughness, and porosity, is crucial for driving cellular attachment and migration. Similarly, modification of scaffold surface chemistry by adapting hydrophilicity, surface charge, surface coatings, can down-regulate the initiation of pro-inflammatory cascades. Besides, functionalization of scaffold surfaces with active biological molecules can down-regulate pro-inflammatory and pro-resorptive mediators' release as well as actively up-regulate anti-inflammatory markers. This review encompasses various strategies for the optimization of physical, chemical, and biological properties of biomaterial and the underlying mechanisms to modulate the immune-inflammatory response, thereby, promoting the tissue integration and subsequent soft and hard tissue regeneration potential of the administered biomaterial.

8.
Biomed Mater ; 16(6)2021 10 04.
Article in English | MEDLINE | ID: mdl-34500442

ABSTRACT

Several chemical compounds are considered to be promising as adjuvants in the treatment of periodontitis. Antimicrobials, anti-inflammatory drugs or, more recently, pro-regenerative or antioxidant molecules have shown a very interesting potential to improve the outcomes of mechanical biofilm removal and promote the healing of the damaged tissues. However, their clinical effect is often limited by the challenge of achieving effective and prolonged drug delivery within the periodontal lesion, while limiting the risk of toxicity.In-situforming implants (ISFI) are 'implantable' drug-delivery systems that have gained considerable attention over the last few decades due to their multiple biomedical applications. They are liquids that, when injected at the site to be treated, form a semi-solid or solid dosage form that provides safe and locally controlled drug release. This review discusses current data and future prospects for the use of ISFI in periodontal treatment.


Subject(s)
Anti-Infective Agents , Drug Delivery Systems , Drug Implants , Periodontitis/drug therapy , Animals , Humans , Mice
9.
Mar Drugs ; 19(7)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34210070

ABSTRACT

Oxygen (O2) is indispensable for aerobic respiration and cellular metabolism. In case of injury, reactive oxygen species are produced, causing oxidative stress, which triggers cell damaging chemical mediators leading to ischemic reperfusion injuries (IRI). Sufficient tissue oxygenation is necessary for optimal wound healing. In this context, several hemoglobin-based oxygen carriers have been developed and tested, especially as graft preservatives for transplant procedures. However, most of the commercially available O2 carriers increase oxidative stress and show some adverse effects. Interestingly, the hemoglobin derived from the marine lugworm Arenicola marina (M101) has been presented as an efficient therapeutic O2 carrier with potential anti-inflammatory, anti-bacterial, and antioxidant properties. Furthermore, it has demonstrated promise as a supplement to conventional organ preservatives by reducing IRI. This review summarizes the properties and various applications of M101. M101 is an innovative oxygen carrier with several beneficial therapeutic properties, and further research must be carried out to determine its efficacy in the management of different pathologies.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hemoglobins/pharmacology , Oxidative Stress/drug effects , Polychaeta , Animals , Aquatic Organisms
10.
Int J Pharm ; 605: 120810, 2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34144138

ABSTRACT

Periodontitis is an inflammatory disease associated with anaerobic bacteria leading to the destruction of tooth-supporting tissues. Porphyromonas gingivalis is a keystone anaerobic pathogen involved in the development of severe lesions. Periodontal treatment aims to suppress subgingival biofilms and to restore tissue homeostasis. However, hypoxia impairs wound healing and promotes bacterial growth within periodontal pocket. This study aimed to evaluate the potential of local oxygen delivery through the local application of a hydrogel containing Arenicola marina's hemoglobin (M101). To this end, a hydrogel (xanthan (2%), hyaluronic acid (1%)) containing M101 (1-2 g/L) (Xn(2%)-HA(1%)-M101) was prepared and characterized. Rheological tests revealed the occurrence of high deformation without the loss of elastic properties. Dialysis experiment revealed that incorporation of M101 within the gel did not modify its oxygen transportation properties. Samples of release media of the gels (1 g/L (10%) and 2 g/L (10%) M101) decreased significantly the growth of P. gingivalis after 24 h validating its antibacterial effect. Metabolic activity measurement confirmed the cytocompatibility of Xn(2%)-HA(1%)-M101. This study suggests the therapeutic interest of Xn(2%)-HA(1%)-M101 gel to optimize treatment of periodontitis with a non-invasive approach.


Subject(s)
Hyaluronic Acid , Periodontitis , Humans , Hydrogels , Oxygen , Periodontitis/drug therapy , Renal Dialysis
11.
Oral Dis ; 27(2): 151-167, 2021 Mar.
Article in English | MEDLINE | ID: mdl-31886584

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune multisystem disease with numerous clinical manifestations. There is no consensus about the ideal oral management for this group of patients to date. This review aimed to describe the broad spectrum of orofacial and clinical manifestations and their therapeutic approaches. Studies concerning orofacial manifestations of SLE and dental treatment modalities were selected by a literature search (1978-2019) using Google Scholar, PubMed/MEDLINE electronic databases. The initial search strategy provided a total of 129 articles, and of these, 30 were included for qualitative synthesis. The reviewed studies revealed that SLE patients are more at risk of compromised oral and dental health exhibiting increased risk of periodontal diseases and temporomandibular joint disorders. The use of systemic drugs especially immunosuppressive and anticoagulants in SLE patients may also influence their oral management. Results emphasize the need to carry out, at an early stage of the disease, an appropriate oral management of these patients to improve oral health-related quality of life and to prevent the need of more invasive therapeutics. A multidisciplinary approach is needed for dental and medical management of such patients.


Subject(s)
Autoimmune Diseases , Lupus Erythematosus, Systemic , Temporomandibular Joint Disorders , Dental Care , Humans , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/drug therapy , Quality of Life
12.
Adv Exp Med Biol ; 1341: 67-87, 2021.
Article in English | MEDLINE | ID: mdl-33159304

ABSTRACT

Periodontitis is an infectious inflammatory disease characterized by clinical attachment loss and tooth supporting tissue destruction. As exosomes demonstrated pro-regenerative ability, their use in periodontal treatment has been suggested. The aim of this systematic review is to gather and summarize the most recent data regarding exosomes to determine their potential impact in bone and periodontal regeneration. Electronic databases (Pubmed, Web of Science) were searched up to February 2020. Studies assessing the impact of exosomes administration in experimental bone and periodontal defects have been identified according to PRISMA guidelines. Among the 183 identified articles, 16 met the inclusion criteria and were included in this systematic review. Experimental bone defects were mainly surgically induced with a dental bur or distraction tools. All studies considered bone healing after exosomes administration as the primary outcome. Results showed that mesenchymal stem cells derived exosomes administration promoted bone healing and neovascularization. Nevertheless, a dose-effect relationship was observed. Exosomes administration appears to promote significantly the bone healing and periodontal regeneration. However, only a limited number of studies have been carried out so far and the optimized protocols in this context need to be evaluated.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Periodontitis , Bone Regeneration , Bone and Bones , Guided Tissue Regeneration, Periodontal , Humans , Periodontitis/therapy
13.
Arch Oral Biol ; 120: 104932, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33113458

ABSTRACT

OBJECTIVES: The purpose of this systematic review was to determine the potential interest of parathyroid hormone (PTH) as an adjunct to periodontal treatment based on studies performed in rodents. MATERIALS & METHODS: Electronic databases (MEDLINE, Web of Science) were searched up to December 2019. Studies assessing the impact of PTH administration in experimental periodontitis in rodents have been identified. RESULTS: Amongst the 247 identified articles, 10 met the inclusion criteria and were included in this systematic review. Experimental periodontitis was mainly induced by ligature placement or surgically with a dental bur. All studies considered bone healing after PTH administration at different frequencies as primary outcome. Results showed that an intermittent administration of PTH promoted bone healing and neovascularization. Nevertheless, a decrease of soft tissue inflammation was also observed. CONCLUSION: Intermittent administration of PTH appears to enhance significantly periodontal healing and to promote alveolar bone regeneration. However, due to the risk of side effects, the development of scaffolds allowing its local and time-controlled delivery is of importance.


Subject(s)
Bone Regeneration , Disease Models, Animal , Parathyroid Hormone/therapeutic use , Periodontitis/therapy , Wound Healing , Alveolar Bone Loss/prevention & control , Animals
14.
Sci Rep ; 10(1): 14745, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32901057

ABSTRACT

The control of inflammation and infection is crucial for periodontal wound healing and regeneration. M101, an oxygen carrier derived from Arenicola marina, was tested for its anti-inflammatory and anti-infectious potential based on its anti-oxidative and tissue oxygenation properties. In vitro, no cytotoxicity was observed in oral epithelial cells (EC) treated with M101. M101 (1 g/L) reduced significantly the gene expression of pro-inflammatory markers such as TNF-α, NF-κΒ and RANKL in P. gingivalis-LPS stimulated and P. gingivalis-infected EC. The proteome array revealed significant down-regulation of pro-inflammatory cytokines (IL-1ß and IL-8) and chemokine ligands (RANTES and IP-10), and upregulation of pro-healing mediators (PDGF-BB, TGF-ß1, IL-10, IL-2, IL-4, IL-11 and IL-15) and, extracellular and immune modulators (TIMP-2, M-CSF and ICAM-1). M101 significantly increased the gene expression of Resolvin-E1 receptor. Furthermore, M101 treatment reduced P. gingivalis biofilm growth over glass surface, observed with live/dead analysis and by decreased P. gingivalis 16 s rRNA expression (51.7%) (p < 0.05). In mice, M101 reduced the clinical abscess size (50.2%) in P. gingivalis-induced calvarial lesion concomitant with a decreased inflammatory score evaluated through histomorphometric analysis, thus, improving soft tissue and bone healing response. Therefore, M101 may be a novel therapeutic agent that could be beneficial in the management of P. gingivalis associated diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bacteroidaceae Infections/complications , Brain Abscess/drug therapy , Inflammation/drug therapy , Oxygen/pharmacology , Polychaeta/chemistry , Skull/drug effects , Animals , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/microbiology , Brain Abscess/microbiology , Brain Abscess/pathology , Gingiva/chemistry , Gingiva/microbiology , Humans , Inflammation/microbiology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Porphyromonas gingivalis/drug effects , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/isolation & purification , Signal Transduction , Skull/microbiology , Skull/pathology
15.
Int J Pharm ; 586: 119534, 2020 Aug 30.
Article in English | MEDLINE | ID: mdl-32531451

ABSTRACT

Statins have been proposed as potential adjuvant to periodontal treatment due to their pleiotropic properties. A new thermosensitive chitosan hydrogel loaded with statins (atorvastatin and lovastatin) nanoemulsions was synthesized to allow a spatially controlled local administration of active compounds at lesion site. Spontaneous nano-emulsification method was used to synthesize statins loaded nanoemulsions. In vitro, atorvastatin and lovastatin loaded nanoemulsions were cytocompatible and were able to be uptake by oral epithelial cells. Treatment of Porphyromonas gingivalis infected oral epithelial cells and gingival fibroblasts with atorvastatin and lovastatin loaded nanoemulsions decreased significantly pro-inflammatory markers expression (TNF-α and IL-1ß) and pro-osteoclastic RANKL. Nevertheless, such treatment induced the expression of Bone sialoprotein 2 (BSP2) in osteoblast emphasizing the pro-healing properties of atorvastatin and lovastatin nanoemulsions. In vivo, in a calvarial bone defect model (2 mm), treatment with the hydrogel loaded with atorvastatin and lovastatin nanoemulsions induced a significant increase of the neobone formation in comparison with systemic administration of statins. This study demonstrates the potential of this statins loaded hydrogel to improve bone regeneration and to decrease soft tissue inflammation. Its use in the specific context of periodontitis management could be considered in the future with a reduced risk of side effects.


Subject(s)
Atorvastatin/pharmacology , Bone Regeneration/drug effects , Chitosan/chemistry , Lovastatin/pharmacology , Animals , Atorvastatin/administration & dosage , Cells, Cultured , Epithelial Cells/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Gingiva/cytology , Gingiva/drug effects , Humans , Hydrogels , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/administration & dosage , Male , Mice , Mice, Inbred C57BL , Porphyromonas gingivalis/drug effects
16.
Sci Rep ; 10(1): 1778, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32019950

ABSTRACT

A link between periodontitis and atherothrombosis has been highlighted. The aim of this study was to determine the influence of Porphyromonas gingivalis on endothelial microvesicles (EMVPg) shedding and their contribution to endothelial inflammation. Endothelial cells (EC) were infected with P. gingivalis (MOI = 100) for 24 h. EMVPg were isolated and their concentration was evaluated by prothrombinase assay. EMVPg were significantly increased in comparison with EMVCtrl shedded by unstimulated cells. While EMVCtrl from untreated EC had no effect, whereas, the proportion of apoptotic EC was increased by 30 nM EMVPg and viability was decreased down to 25%, a value elicited by P. gingivalis alone. Moreover, high concentration of EMVPg (30 nM) induced a pro-inflammatory and pro-oxidative cell response including up-regulation of TNF-α, IL-6 and IL-8 as well as an altered expression of iNOS and eNOS at both mRNA and protein level. An increase of VCAM-1 and ICAM-1 mRNA expression (4.5 folds and 3 folds respectively (p < 0.05 vs untreated) was also observed after EMVPg (30 nM) stimulation whereas P. gingivalis infection was less effective, suggesting a specific triggering by EMVPg. Kinasome analysis demonstrated the specific effect induced by EMVPg on main pro-inflammatory pathways including JNK/AKT and STAT. EMVPg are effective pro-inflammatory effectors that may have detrimental effect on vascular homeostasis and should be considered as potential autocrine and paracrine effectors involved in the link between periodontitis and atherothrombosis.


Subject(s)
Bacteroidaceae Infections/metabolism , Cell-Derived Microparticles/microbiology , Endothelial Cells/microbiology , Oxidative Stress/physiology , Porphyromonas gingivalis , Cell-Derived Microparticles/metabolism , Cytokines/metabolism , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/microbiology , Humans , Inflammation/metabolism , Inflammation/microbiology , Intercellular Adhesion Molecule-1/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
17.
Sci Rep ; 9(1): 19257, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31848404

ABSTRACT

Periodontitis is a prevalent chronic inflammatory disease due to the host response (IL-1ß, IL-6, TNF-α and IL-17A) to oral bacteria such as Porphyromonas gingivalis. The newer members of the IL-1 family, IL-36s (IL-36α/IL-36ß/IL-36γ/IL-36Ra/IL-38) are known to be involved in host defense against P. gingivalis in oral epithelial cells (OECs) and are considered as key inflammatory mediators in chronic diseases. The aim of this study was to investigate the potential role of IL-36s in periodontitis. We showed here that IL-36γ mRNA gingival expression is higher in periodontitis patients, whereas IL-36ß and IL-36Ra mRNA expression are lower compared to healthy controls. Interestingly, the elevated IL-36γ expression in patients is positively correlated with the RANKL/OPG ratio, an index of bone resorption. In vitro, IL-36γ expression was induced through TLR2 activation in primary OECs infected with P. gingivalis but not in gingival fibroblasts, the most widespread cell type in gingival connective tissue. In OECs, recombinant IL-36γ enhanced the expression of inflammatory cytokines (IL-1ß, IL-6, TNF-α and IL-36γ), of TLR2 and importantly, the RANKL/OPG ratio. These findings suggest that IL-36γ could be a pivotal inflammatory player in periodontitis by perpetuating gingival inflammation and its associated alveolar bone resorption and could be a relevant therapeutic target.


Subject(s)
Alveolar Bone Loss , Bacteroidaceae Infections , Interleukin-1/metabolism , Periodontitis , Porphyromonas gingivalis/metabolism , Alveolar Bone Loss/metabolism , Alveolar Bone Loss/microbiology , Alveolar Bone Loss/pathology , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/pathology , Cell Line , Female , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Male , Periodontitis/metabolism , Periodontitis/microbiology , Periodontitis/pathology
18.
Int J Pharm ; 569: 118564, 2019 Oct 05.
Article in English | MEDLINE | ID: mdl-31352049

ABSTRACT

Control of infection and inflammation is crucial for the success of periodontal treatment. In this study, in-situ forming implants (ISFI) loaded with chlorhexidine dihydrochloride (CHX) and ibuprofen (IBU) were developed and tested to optimize periodontal treatment outcomes. Release profiles were promising. Exposure to 1.5% and 5.3% CHX-IBU loaded ISFI's release media decreased significantly the P. gingivalis growth up to 20-fold and 35-fold, respectively, after 48 h (p < 0.05). The metabolic activity assay of gingival epithelial cells (EC) demonstrated 1.5% CHX-IBU-loaded ISFI to be non-toxic, therefore, it was selected for further experimentation. Furthermore, significant down-regulation of TNF-α release (34% at 6 h and 43% at 24 h, p < 0.05) in P. gingivalis lipopolysaccharide (Pg-LPS) stimulated EC exposed to 1.5% CHX-IBU ISFI release medium was demonstrated by ELISA. In vivo, 1.5% CHX-IBU ISFI was injected into the periodontal pocket in an experimental periodontitis mouse model and the reduction in inflammation and improvement in periodontal wound healing was evaluated through inflammatory cell scoring and histomorphometry at 7- and 15-days post-treatment. The results indicate that CHX-IBU loaded ISFI could be efficient as adjuvant to periodontal therapy for the control of infection and inflammation. Moreover, other (e.g., pro-regenerative) drugs could be incorporated into ISFI to further improve periodontal treatment outcomes.


Subject(s)
Anti-Infective Agents, Local/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Chlorhexidine/administration & dosage , Ibuprofen/administration & dosage , Periodontitis/drug therapy , Animals , Anti-Infective Agents, Local/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Line , Chlorhexidine/chemistry , Drug Implants , Drug Liberation , Epithelial Cells/drug effects , Gingiva/cytology , Humans , Ibuprofen/chemistry , Lipopolysaccharides , Male , Mice, Inbred C57BL , Proof of Concept Study , Wound Healing/drug effects
19.
Mediators Inflamm ; 2019: 6367402, 2019.
Article in English | MEDLINE | ID: mdl-30936777

ABSTRACT

The pleiotropic effects of statins have been evaluated to assess their potential benefit in the treatment of various inflammatory and immune-mediated diseases including periodontitis. Herein, the adjunctive use of statins in periodontal therapy in vitro, in vivo, and in clinical trials was reviewed. Statins act through several pathways to modulate inflammation, immune response, bone metabolism, and bacterial clearance. They control periodontal inflammation through inhibition of proinflammatory cytokines and promotion of anti-inflammatory and/or proresolution molecule release, mainly, through the ERK, MAPK, PI3-Akt, and NF-κB pathways. Moreover, they are able to modulate the host response activated by bacterial challenge, to prevent inflammation-mediated bone resorption and to promote bone formation. Furthermore, they reduce bacterial growth, disrupt bacterial membrane stability, and increase bacterial clearance, thus averting the exacerbation of infection. Local statin delivery as adjunct to both nonsurgical and surgical periodontal therapies results in better periodontal treatment outcomes compared to systemic delivery. Moreover, combination of statin therapy with other regenerative agents improves periodontal healing response. Therefore, statins could be proposed as a potential adjuvant to periodontal therapy. However, optimization of the combination of their dose, type, and carrier could be instrumental in achieving the best treatment response.


Subject(s)
Inflammation/drug therapy , Periodontal Diseases/drug therapy , Animals , Cytokines/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Inflammation/metabolism , Periodontal Diseases/metabolism , Periodontitis/drug therapy , Periodontitis/metabolism
20.
Sci Rep ; 8(1): 14914, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30297793

ABSTRACT

Porphyromonas gingivalis-induced inflammatory effects are mostly investigated in monolayer cultured cells. The aim of this study was to develop a 3D spheroid model of gingiva to take into account epithelio-fibroblastic interactions. Human gingival epithelial cells (ECs) and human oral fibroblasts (FBs) were cultured by hanging drop method to generate 3D microtissue (MT) whose structure was analyzed on histological sections and the cell-to-cell interactions were observed by scanning and transmission electron microscopy (SEM and TEM). MTs were infected by P. gingivalis and the impact on cell death (Apaf-1, caspase-3), inflammatory markers (TNF-α, IL-6, IL-8) and extracellular matrix components (Col-IV, E-cadherin, integrin ß1) was evaluated by immunohistochemistry and RT-qPCR. Results were compared to those observed in situ in experimental periodontitis and in human gingival biopsies. MTs exhibited a well-defined spatial organization where ECs were organized in an external cellular multilayer, while, FBs constituted the core. The infection of MT demonstrated the ability of P. gingivalis to bypass the epithelial barrier in order to reach the fibroblastic core and induce disorganization of the spheroid structure. An increased cell death was observed in fibroblastic core. The development of such 3D model may be useful to define the role of EC-FB interactions on periodontal host-immune response and to assess the efficacy of new therapeutics.


Subject(s)
Epithelial Cells/pathology , Fibroblasts/microbiology , Fibroblasts/pathology , Inflammation/microbiology , Inflammation/pathology , Models, Biological , Porphyromonas gingivalis/physiology , Spheroids, Cellular/pathology , Adult , Apoptosis/genetics , Epithelial Cells/ultrastructure , Female , Fibroblasts/ultrastructure , Gene Expression Regulation , Gingiva/pathology , Humans , Male , Middle Aged , Periodontitis/microbiology , Periodontitis/pathology , Porphyromonas gingivalis/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...