Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(14): 3690-3711.e19, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38838669

RESUMEN

Clonal hematopoiesis of indeterminate potential (CHIP) arises from aging-associated acquired mutations in hematopoietic progenitors, which display clonal expansion and produce phenotypically altered leukocytes. We associated CHIP-DNMT3A mutations with a higher prevalence of periodontitis and gingival inflammation among 4,946 community-dwelling adults. To model DNMT3A-driven CHIP, we used mice with the heterozygous loss-of-function mutation R878H, equivalent to the human hotspot mutation R882H. Partial transplantation with Dnmt3aR878H/+ bone marrow (BM) cells resulted in clonal expansion of mutant cells into both myeloid and lymphoid lineages and an elevated abundance of osteoclast precursors in the BM and osteoclastogenic macrophages in the periphery. DNMT3A-driven clonal hematopoiesis in recipient mice promoted naturally occurring periodontitis and aggravated experimentally induced periodontitis and arthritis, associated with enhanced osteoclastogenesis, IL-17-dependent inflammation and neutrophil responses, and impaired regulatory T cell immunosuppressive activity. DNMT3A-driven clonal hematopoiesis and, subsequently, periodontitis were suppressed by rapamycin treatment. DNMT3A-driven CHIP represents a treatable state of maladaptive hematopoiesis promoting inflammatory bone loss.


Asunto(s)
Hematopoyesis Clonal , ADN (Citosina-5-)-Metiltransferasas , ADN Metiltransferasa 3A , Periodontitis , Animales , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Ratones , Hematopoyesis Clonal/genética , Humanos , Periodontitis/genética , Periodontitis/patología , Mutación , Masculino , Femenino , Inflamación/genética , Inflamación/patología , Osteoclastos/metabolismo , Ratones Endogámicos C57BL , Adulto , Interleucina-17/metabolismo , Interleucina-17/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Hematopoyesis/genética , Osteogénesis/genética , Células Madre Hematopoyéticas/metabolismo , Resorción Ósea/genética , Resorción Ósea/patología , Persona de Mediana Edad
2.
Cell ; 185(10): 1709-1727.e18, 2022 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-35483374

RESUMEN

Bone marrow (BM)-mediated trained innate immunity (TII) is a state of heightened immune responsiveness of hematopoietic stem and progenitor cells (HSPC) and their myeloid progeny. We show here that maladaptive BM-mediated TII underlies inflammatory comorbidities, as exemplified by the periodontitis-arthritis axis. Experimental-periodontitis-related systemic inflammation in mice induced epigenetic rewiring of HSPC and led to sustained enhancement of production of myeloid cells with increased inflammatory preparedness. The periodontitis-induced trained phenotype was transmissible by BM transplantation to naive recipients, which exhibited increased inflammatory responsiveness and disease severity when subjected to inflammatory arthritis. IL-1 signaling in HSPC was essential for their maladaptive training by periodontitis. Therefore, maladaptive innate immune training of myelopoiesis underlies inflammatory comorbidities and may be pharmacologically targeted to treat them via a holistic approach.


Asunto(s)
Artritis , Periodontitis , Animales , Células Madre Hematopoyéticas , Inmunidad Innata , Ratones , Mielopoyesis
3.
Cell ; 184(15): 4090-4104.e15, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34129837

RESUMEN

The oral mucosa remains an understudied barrier tissue. This is a site of rich exposure to antigens and commensals, and a tissue susceptible to one of the most prevalent human inflammatory diseases, periodontitis. To aid in understanding tissue-specific pathophysiology, we compile a single-cell transcriptome atlas of human oral mucosa in healthy individuals and patients with periodontitis. We uncover the complex cellular landscape of oral mucosal tissues and identify epithelial and stromal cell populations with inflammatory signatures that promote antimicrobial defenses and neutrophil recruitment. Our findings link exaggerated stromal cell responsiveness with enhanced neutrophil and leukocyte infiltration in periodontitis. Our work provides a resource characterizing the role of tissue stroma in regulating mucosal tissue homeostasis and disease pathogenesis.


Asunto(s)
Inmunidad Mucosa , Mucosa Bucal/citología , Mucosa Bucal/inmunología , Neutrófilos/citología , Adulto , Células Epiteliales/citología , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Encía/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Microbiota , Células Mieloides/citología , Periodontitis/genética , Periodontitis/inmunología , Periodontitis/patología , Análisis de la Célula Individual , Células del Estroma/citología , Linfocitos T/citología
4.
Nat Immunol ; 20(1): 40-49, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30455459

RESUMEN

Resolution of inflammation is essential for tissue homeostasis and represents a promising approach to inflammatory disorders. Here we found that developmental endothelial locus-1 (DEL-1), a secreted protein that inhibits leukocyte-endothelial adhesion and inflammation initiation, also functions as a non-redundant downstream effector in inflammation clearance. In human and mouse periodontitis, waning of inflammation was correlated with DEL-1 upregulation, whereas resolution of experimental periodontitis failed in DEL-1 deficiency. This concept was mechanistically substantiated in acute monosodium-urate-crystal-induced inflammation, where the pro-resolution function of DEL-1 was attributed to effective apoptotic neutrophil clearance (efferocytosis). DEL-1-mediated efferocytosis induced liver X receptor-dependent macrophage reprogramming to a pro-resolving phenotype and was required for optimal production of at least certain specific pro-resolving mediators. Experiments in transgenic mice with cell-specific overexpression of DEL-1 linked its anti-leukocyte-recruitment action to endothelial cell-derived DEL-1 and its efferocytic/pro-resolving action to macrophage-derived DEL-1. Thus, the compartmentalized expression of DEL-1 facilitates distinct homeostatic functions in an appropriate context that can be harnessed therapeutically.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamación/inmunología , Macrófagos/fisiología , Neutrófilos/inmunología , Periodontitis/inmunología , Adulto , Animales , Proteínas de Unión al Calcio , Proteínas Portadoras/genética , Moléculas de Adhesión Celular , Reprogramación Celular , Citocinas/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamación/inducido químicamente , Péptidos y Proteínas de Señalización Intercelular , Células K562 , Receptores X del Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis
5.
Immunol Rev ; 314(1): 93-110, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36271881

RESUMEN

Neutrophils are of key importance in periodontal health and disease. In their absence or when they are functionally defective, as occurs in certain congenital disorders, affected individuals develop severe forms of periodontitis in early age. These observations imply that the presence of immune-competent neutrophils is essential to homeostasis. However, the presence of supernumerary or hyper-responsive neutrophils, either because of systemic priming or innate immune training, leads to imbalanced host-microbe interactions in the periodontium that culminate in dysbiosis and inflammatory tissue breakdown. These disease-provoking imbalanced interactions are further exacerbated by periodontal pathogens capable of subverting neutrophil responses to their microbial community's benefit and the host's detriment. This review attempts a synthesis of these findings for an integrated view of the neutrophils' ambivalent role in periodontal disease and, moreover, discusses how some of these concepts underpin the development of novel therapeutic approaches to treat periodontal disease.


Asunto(s)
Neutrófilos , Periodontitis , Humanos , Inflamación , Periodoncio , Homeostasis
6.
Semin Immunol ; 59: 101608, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35691883

RESUMEN

Periodontitis is an inflammatory disease caused by biofilm accumulation and dysbiosis in subgingival areas surrounding the teeth. If not properly treated, this oral disease may result in tooth loss and consequently poor esthetics, deteriorated masticatory function and compromised quality of life. Epidemiological and clinical intervention studies indicate that periodontitis can potentially aggravate systemic diseases, such as, cardiovascular disease, type 2 diabetes mellitus, rheumatoid arthritis, and Alzheimer disease. Therefore, improvements in the treatment of periodontal disease may benefit not only oral health but also systemic health. The complement system is an ancient host defense system that plays pivotal roles in immunosurveillance and tissue homeostasis. However, complement has unwanted consequences if not controlled appropriately or excessively activated. Complement overactivation has been observed in patients with periodontitis and in animal models of periodontitis and drives periodontal inflammation and tissue destruction. This review places emphasis on a promising periodontal host-modulation therapy targeting the complement system, namely the complement C3-targeting drug, AMY-101. AMY-101 has shown safety and efficacy in reducing gingival inflammation in a recent Phase 2a clinical study. We also discuss the potential of AMY-101 to treat peri-implant inflammatory conditions, where complement also seems to be involved and there is an urgent unmet need for effective treatment.


Asunto(s)
Diabetes Mellitus Tipo 2 , Periodontitis , Animales , Humanos , Complemento C3 , Calidad de Vida , Periodontitis/terapia , Inflamación
7.
J Immunol ; 211(3): 453-461, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37306457

RESUMEN

A minimized version of complement factor H (FH), designated mini-FH, was previously engineered combining the N-terminal regulatory domains (short consensus repeat [SCR]1-4) and C-terminal host-surface recognition domains (SCR19-20) of the parent molecule. Mini-FH conferred enhanced protection, as compared with FH, in an ex vivo model of paroxysmal nocturnal hemoglobinuria driven by alternative pathway dysregulation. In the current study, we tested whether and how mini-FH could block another complement-mediated disease, namely periodontitis. In a mouse model of ligature-induced periodontitis (LIP), mini-FH inhibited periodontal inflammation and bone loss in wild-type mice. Although LIP-subjected C3-deficient mice are protected relative to wild-type littermates and exhibit only modest bone loss, mini-FH strikingly inhibited bone loss even in C3-deficient mice. However, mini-FH failed to inhibit ligature-induced bone loss in mice doubly deficient in C3 and CD11b. These findings indicate that mini-FH can inhibit experimental periodontitis even in a manner that is independent of its complement regulatory activity and is mediated by complement receptor 3 (CD11b/CD18). Consistent with this notion, a complement receptor 3-interacting recombinant FH segment that lacks complement regulatory activity (specifically encompassing SCRs 19 and 20; FH19-20) was also able to suppress bone loss in LIP-subjected C3-deficient mice. In conclusion, mini-FH appears to be a promising candidate therapeutic for periodontitis by virtue of its ability to suppress bone loss via mechanisms that both include and go beyond its complement regulatory activity.


Asunto(s)
Factor H de Complemento , Periodontitis , Ratones , Animales , Factor H de Complemento/metabolismo , Vía Alternativa del Complemento , Proteínas del Sistema Complemento , Receptores de Complemento
8.
Nat Immunol ; 13(5): 465-73, 2012 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-22447028

RESUMEN

Aging is linked to greater susceptibility to chronic inflammatory diseases, several of which, including periodontitis, involve neutrophil-mediated tissue injury. Here we found that aging-associated periodontitis was accompanied by lower expression of Del-1, an endogenous inhibitor of neutrophil adhesion dependent on the integrin LFA-1, and by reciprocal higher expression of interleukin 17 (IL-17). Consistent with that, IL-17 inhibited gingival endothelial cell expression of Del-1, thereby promoting LFA-1-dependent recruitment of neutrophils. Young Del-1-deficient mice developed spontaneous periodontitis that featured excessive neutrophil infiltration and IL-17 expression; disease was prevented in mice doubly deficient in Del-1 and LFA-1 or in Del-1 and the IL-17 receptor. Locally administered Del-1 inhibited IL-17 production, neutrophil accumulation and bone loss. Therefore, Del-1 suppressed LFA-1-dependent recruitment of neutrophils and IL-17-triggered inflammatory pathology and may thus be a promising therapeutic agent for inflammatory diseases.


Asunto(s)
Pérdida de Hueso Alveolar/inmunología , Proteínas Portadoras/metabolismo , Interleucina-17/antagonistas & inhibidores , Interleucina-17/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Periodontitis/metabolismo , Envejecimiento/inmunología , Animales , Proteínas de Unión al Calcio , Proteínas Portadoras/inmunología , Proteínas Portadoras/farmacología , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Femenino , Integrinas/antagonistas & inhibidores , Integrinas/inmunología , Integrinas/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Interleucina-17/inmunología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Atrofia Periodontal/inmunología , Atrofia Periodontal/metabolismo , Periodontitis/inmunología , Periodontitis/terapia , Receptores de Interleucina-17/deficiencia , Receptores de Interleucina-17/metabolismo
9.
Trends Immunol ; 42(10): 856-864, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34483038

RESUMEN

Complement plays a key role in immunosurveillance and homeostasis. When dysregulated or overactivated, complement can become a pathological effector, as seen in several inflammatory disorders, including periodontal disease. Recently, clinical correlative studies and preclinical mechanistic investigations have collectively demonstrated that complement is hyperactivated during periodontitis and that targeting its central component (C3) provides therapeutic benefit in nonhuman primates (NHPs). The preclinical efficacy of a C3-targeted drug candidate combined with excellent safety and pharmacokinetic profiles supported its use in a recent Phase IIa clinical study in which C3 inhibition resolved gingival inflammation in patients with periodontal disease. We posit that C3-targeted intervention might represent a novel and transformative host-modulation therapy meriting further investigation in Phase III clinical trials for the treatment of periodontitis.


Asunto(s)
Complemento C3 , Periodontitis , Animales , Protocolos de Ensayos Clínicos como Asunto , Ensayos Clínicos Fase III como Asunto , Humanos , Periodontitis/tratamiento farmacológico
10.
J Immunol ; 209(7): 1370-1378, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36028293

RESUMEN

In both mice and humans, complement and Th17 cells have been implicated in periodontitis, an oral microbiota-driven inflammatory disease associated with systemic disorders. A recent clinical trial showed that a complement C3 inhibitor (AMY-101) causes sustainable resolution of periodontal inflammation, the main effector of tissue destruction in this oral disease. Although both complement and Th17 are required for periodontitis, it is uncertain how these immune components cooperate in disease development. In this study, we dissected the complement-Th17 relationship in the setting of ligature-induced periodontitis (LIP), a model that previously established that microbial dysbiosis drives Th17 cell expansion and periodontal bone loss. Complement was readily activated in the periodontal tissue of LIP-subjected mice but not when the mice were placed on broad-spectrum antibiotics. Microbiota-induced complement activation generated critical cytokines, IL-6 and IL-23, which are required for Th17 cell expansion. These cytokines as well as Th17 accumulation and IL-17 expression were significantly suppressed in LIP-subjected C3-deficient mice relative to wild-type controls. As IL-23 has been extensively studied in periodontitis, we focused on IL-6 and showed that LIP-induced IL-17 and bone loss required intact IL-6 receptor signaling in the periodontium. LIP-induced IL-6 was predominantly produced by gingival epithelial cells that upregulated C3a receptor upon LIP challenge. Experiments in human gingival epithelial cells showed that C3a upregulated IL-6 production in cooperation with microbial stimuli that upregulated C3a receptor expression in ERK1/2- and JNK-dependent manner. In conclusion, complement links the periodontal microbiota challenge to Th17 cell accumulation and thus integrates complement- and Th17-driven immunopathology in periodontitis.


Asunto(s)
Pérdida de Hueso Alveolar , Periodontitis , Animales , Antibacterianos , Complemento C3 , Humanos , Interleucina-17 , Interleucina-23 , Interleucina-6/metabolismo , Ratones , Receptores de Interleucina-6 , Células Th17
11.
Periodontol 2000 ; 89(1): 9-18, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35244969

RESUMEN

Periodontitis, a microbiome-driven inflammatory disease of the tooth-attachment apparatus, is epidemiologically linked with other disorders, including cardio-metabolic, cognitive neurodegenerative and autoimmune diseases, respiratory infections, and certain cancers. These associations may, in part, be causal, as suggested by interventional studies showing that local treatment of periodontitis reduces systemic inflammation and surrogate markers of comorbid diseases. The potential cause-and-effect connection between periodontitis and comorbidities is corroborated by studies in preclinical models of disease, which additionally provided mechanistic insights into these associations. This overview discusses recent advances in our understanding of the periodontitis-systemic disease connection, which may potentially lead to innovative therapeutic options to reduce the risk of periodontitis-linked comorbidities.


Asunto(s)
Microbiota , Enfermedades Periodontales , Periodontitis , Humanos , Inflamación , Enfermedades Periodontales/complicaciones , Enfermedades Periodontales/epidemiología , Enfermedades Periodontales/terapia , Periodontitis/complicaciones , Periodontitis/epidemiología , Periodontitis/terapia
12.
Periodontol 2000 ; 89(1): 215-230, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35244943

RESUMEN

Periodontitis is bidirectionally associated with systemic inflammatory disorders. The prevalence and severity of this oral disease and linked comorbidities increases with aging. Here, we review two newly emerged concepts, trained innate immunity (TII) and clonal hematopoiesis of indeterminate potential (CHIP), which together support a potential hypothesis on how periodontitis affects and is affected by comorbidities and why the susceptibility to periodontitis and comorbidities increases with aging. Given that chronic diseases are largely triggered by the action of inflammatory immune cells, modulation of their bone marrow precursors, the hematopoietic stem and progenitor cells (HSPCs), may affect multiple disorders that emerge as comorbidities. Such alterations in HSPCs can be mediated by TII and/or CHIP, two non-mutually exclusive processes sharing a bias for enhanced myelopoiesis and production of innate immune cells with heightened proinflammatory potential. TII is a state of elevated immune responsiveness based on innate immune (epigenetic) memory. Systemic inflammation can initiate TII in the bone marrow via sustained rewiring of HSPCs, which thereby display a skewing toward the myeloid lineage, resulting in generation of hyper-reactive or "trained" myeloid cells. CHIP arises from aging-related somatic mutations in HSPCs, which confer a survival and proliferation advantage to the mutant HSPCs and give rise to an outsized fraction of hyper-inflammatory mutant myeloid cells in the circulation and tissues. This review discusses emerging evidence that supports the notion that TII and CHIP may underlie a causal and age-related association between periodontitis and comorbidities. A holistic mechanistic understanding of the periodontitis-systemic disease connection may offer novel diagnostic and therapeutic targets for treating inflammatory comorbidities.


Asunto(s)
Hematopoyesis Clonal , Periodontitis , Células Madre Hematopoyéticas , Humanos , Inmunidad Innata , Inflamación , Periodontitis/complicaciones
13.
FASEB J ; 34(10): 13726-13740, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32812255

RESUMEN

The hypoxia-inducible factor 1α (HIF-1α) is critically involved in tissue regeneration. Hence, the pharmacological prevention of HIF-1α degradation by prolyl hydroxylase (PHD) under normoxic conditions is emerging as a promising option in regenerative medicine. Using a mouse model of ligature-induced periodontitis and resolution, we tested the ability of an injectable hydrogel-formulated PHD inhibitor, 1,4-dihydrophenonthrolin-4-one-3-carboxylic acid (1,4-DPCA/hydrogel), to promote regeneration of alveolar bone lost owing to experimental periodontitis. Mice injected subcutaneously with 1,4-DPCA/hydrogel at the onset of periodontitis resolution displayed significantly increased gingival HIF-1α protein levels and bone regeneration, as compared to mice treated with vehicle control. The 1,4-DPCA/hydrogel-induced increase in bone regeneration was associated with elevated expression of osteogenic genes, decreased expression of pro-inflammatory cytokine genes, and increased abundance of FOXP3+ T regulatory (Treg) cells in the periodontal tissue. The enhancing effect of 1,4-DPCA/hydrogel on Treg cell accumulation and bone regeneration was reversed by AMD3100, an antagonist of the chemokine receptor CXCR4 that mediates Treg cell recruitment. In conclusion, the administration of 1,4-DPCA/hydrogel at the onset of periodontitis resolution promotes CXCR4-dependent accumulation of Treg cells and alveolar bone regeneration, suggesting a novel approach for regaining bone lost due to periodontitis.


Asunto(s)
Regeneración Ósea , Inhibidores Enzimáticos/uso terapéutico , Hidrogeles/uso terapéutico , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Periodontitis/tratamiento farmacológico , Linfocitos T Reguladores/inmunología , Animales , Línea Celular , Células Cultivadas , Citocinas/metabolismo , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Femenino , Factores de Transcripción Forkhead/metabolismo , Encía/metabolismo , Humanos , Hidrogeles/administración & dosificación , Hidrogeles/química , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Osteogénesis , Linfocitos T Reguladores/fisiología
14.
Periodontol 2000 ; 86(1): 210-230, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33690950

RESUMEN

In health, indigenous polymicrobial communities at mucosal surfaces maintain an ecological balance via both inter-microbial and host-microbial interactions that promote their own and the host's fitness, while preventing invasion by exogenous pathogens. However, genetic and acquired destabilizing factors (including immune deficiencies, immunoregulatory defects, smoking, diet, obesity, diabetes and other systemic diseases, and aging) may disrupt this homeostatic balance, leading to selective outgrowth of species with the potential for destructive inflammation. This process, known as dysbiosis, underlies the development of periodontitis in susceptible hosts. The pathogenic process is not linear but involves a positive-feedback loop between dysbiosis and the host inflammatory response. The dysbiotic community is essentially a quasi-organismal entity, where constituent organisms communicate via sophisticated physical and chemical signals and display functional specialization (eg, accessory pathogens, keystone pathogens, pathobionts), which enables polymicrobial synergy and dictates the community's pathogenic potential or nososymbiocity. In this review, we discuss early and recent studies in support of the polymicrobial synergy and dysbiosis model of periodontal disease pathogenesis. According to this concept, disease is not caused by individual "causative pathogens" but rather by reciprocally reinforced interactions between physically and metabolically integrated polymicrobial communities and a dysregulated host inflammatory response.


Asunto(s)
Enfermedades Periodontales , Periodontitis , Disbiosis , Interacciones Huésped-Patógeno , Humanos , Inflamación
15.
Proteomics ; 20(3-4): e1900253, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31881116

RESUMEN

Understanding the progression of periodontal tissue destruction is at the forefront of periodontal research. The authors aimed to capture the dynamics of gingival tissue proteome during the initiation and progression of experimental (ligature-induced) periodontitis in mice. Pressure cycling technology (PCT), a recently developed platform that uses ultra-high pressure to disrupt tissues, is utilized to achieve efficient and reproducible protein extraction from ultra-small amounts of gingival tissues in combination with liquid chromatography-tandem mass spectrometry (MS). The MS data are processed using Progenesis QI and the regulated proteins are subjected to METACORE, STRING, and WebGestalt for functional enrichment analysis. A total of 1614 proteins with ≥2 peptides are quantified with an estimated protein false discovery rate of 0.06%. Unsupervised clustering analysis shows that the gingival tissue protein abundance is mainly dependent on the periodontitis progression stage. Gene ontology enrichment analysis reveals an overrepresentation in innate immune regulation (e.g., neutrophil-mediated immunity and antimicrobial peptides), signal transduction (e.g., integrin signaling), and homeostasis processes (e.g., platelet activation and aggregation). In conclusion, a PCT-assisted label-free quantitative proteomics workflow that allowed cataloging the deepest gingival tissue proteome on a rapid timescale and provided novel mechanistic insights into host perturbation during periodontitis progression is applied.


Asunto(s)
Encía/metabolismo , Periodontitis/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos , Tecnología Odontológica/métodos , Animales , Cromatografía Liquida/métodos , Modelos Animales de Enfermedad , Ontología de Genes , Líquido del Surco Gingival/metabolismo , Humanos , Ligadura/efectos adversos , Ratones Endogámicos C57BL , Periodontitis/etiología , Periodontitis/genética , Presión , Mapas de Interacción de Proteínas , Proteoma/genética
16.
N Engl J Med ; 376(12): 1141-1146, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28328326

RESUMEN

A patient with leukocyte adhesion deficiency type 1 (LAD1) had severe periodontitis and an intractable, deep, nonhealing sacral wound. We had previously found a dominant interleukin-23-interleukin-17 signature at inflamed sites in humans with LAD1 and in mouse models of the disorder. Blockade of this pathway in mouse models has resulted in resolution of the immunopathologic condition. We treated our patient with ustekinumab, an antibody that binds the p40 subunit of interleukin-23 and interleukin-12 and thereby blocks the activity of these cytokines, inhibiting interleukin-23-dependent production of interleukin-17. After 1 year of therapy, our patient had resolution of his inflammatory lesions without serious infections or adverse reactions. Inhibition of interleukin-23 and interleukin-17 may have a role in the management of LAD1. (Funded by the National Institute of Allergy and Infectious Diseases and others.).


Asunto(s)
Interleucina-12/antagonistas & inhibidores , Interleucina-23/antagonistas & inhibidores , Síndrome de Deficiencia de Adhesión del Leucocito/tratamiento farmacológico , Ustekinumab/uso terapéutico , Encía/patología , Humanos , Inyecciones Subcutáneas , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Síndrome de Deficiencia de Adhesión del Leucocito/complicaciones , Masculino , Enfermedades Periodontales/tratamiento farmacológico , Enfermedades Periodontales/etiología , Enfermedades Periodontales/patología , ARN Mensajero/metabolismo , Úlcera Cutánea/tratamiento farmacológico , Úlcera Cutánea/etiología , Úlcera Cutánea/patología , Ustekinumab/efectos adversos , Adulto Joven
17.
Periodontol 2000 ; 82(1): 78-92, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31850633

RESUMEN

Neutrophils have been historically associated with antimicrobial functions in acute infections but are now appreciated as functionally versatile cells with critical roles in chronic inflammation. Recent advances in neutrophil biology have contributed to a better understanding of periodontal disease pathogenesis and, reciprocally, the study of periodontitis has led to important insights into neutrophil regulation and function. Here, the contributions by our group to this field through interdisciplinary collaboration are discussed. The study of leukocyte adhesion deficiency-associated periodontitis has revealed that the connection of neutrophils with destructive inflammation may involve mechanisms beyond the typical bystander injury dogma. In this regard, neutrophils are required for important immunomodulatory functions and their absence from the periodontium leads to dysregulated overproduction of interleukin-17, which drives inflammatory bone loss. We have also discovered that both the production of neutrophils in the bone marrow and their recruitment to peripheral tissues, including the periodontium, are homeostatically regulated by a secreted protein designated developmental endothelial locus-1. However, developmental endothelial locus-1 expression, and hence developmental endothelial locus-1-dependent homeostasis, declines considerably with aging and contributes to an increased susceptibility to periodontitis in old age. Moreover, our work has mechanistically supported the concept that periodontitis is a dysbiotic disease and we have shown that neutrophils become targets of immune subversion by periodontal bacteria in a manner that promotes dysbiosis. The mechanism involves microbial exploitation of key neutrophil receptors (complement C5a receptor-1 and toll-like receptor-2), leading to crosstalk signaling that uncouples neutrophil-mediated killing (which is impaired) from neutrophil-induced inflammation (which is enhanced). These studies have collectively established new mechanisms governing the protective and destructive functions of neutrophils in periodontitis and offered targeted host-modulation approaches for the treatment of periodontal diseases.


Asunto(s)
Trastornos Leucocíticos , Periodontitis , Humanos , Inflamación , Neutrófilos , Periodoncio
18.
Periodontol 2000 ; 84(1): 14-34, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32844416

RESUMEN

Recent advances indicate that periodontitis is driven by reciprocally reinforced interactions between a dysbiotic microbiome and dysregulated inflammation. Inflammation is not only a consequence of dysbiosis but, via mediating tissue dysfunction and damage, fuels further growth of selectively dysbiotic communities of bacteria (inflammophiles), thereby generating a self-sustained feed-forward loop that perpetuates the disease. These considerations provide a strong rationale for developing adjunctive host-modulation therapies for the treatment of periodontitis. Such host-modulation approaches aim to inhibit harmful inflammation and promote its resolution or to interfere directly with downstream effectors of connective tissue and bone destruction. This paper reviews diverse strategies targeted to modulate the host periodontal response and discusses their mechanisms of action, perceived safety, and potential for clinical application.


Asunto(s)
Microbiota , Periodontitis/terapia , Disbiosis , Humanos , Inflamación
19.
Semin Immunol ; 28(2): 146-58, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26936034

RESUMEN

Although historically viewed as merely anti-microbial effectors in acute infection or injury, neutrophils are now appreciated to be functionally versatile with critical roles also in chronic inflammation. Periodontitis, a chronic inflammatory disease that destroys the tooth-supporting gums and bone, is particularly affected by alterations in neutrophil numbers or function, as revealed by observations in monogenic disorders and relevant mouse models. Besides being a significant debilitating disease and health burden in its own right, periodontitis is thus an attractive model to dissect uncharted neutrophil-associated (patho)physiological pathways. Here, we summarize recent evidence that neutrophils can contribute to inflammatory bone loss not only through the typical bystander injury dogma but intriguingly also through their absence from the affected tissue, where they normally perform important immunomodulatory functions. Moreover, we discuss recent advances in the interactions of neutrophils with the vascular endothelium and - upon extravasation - with bacteria, and how the dysregulation of these interactions leads to inflammatory tissue damage. Overall, neutrophils have both protective and destructive roles in periodontitis, as they are involved in both the maintenance of periodontal tissue homeostasis and the induction of inflammatory bone loss. This highlights the importance of developing approaches that promote or sustain a fine balance between homeostatic immunity and inflammatory pathology.


Asunto(s)
Resorción Ósea/etiología , Resorción Ósea/metabolismo , Inflamación/complicaciones , Neutrófilos/inmunología , Neutrófilos/metabolismo , Pérdida de Hueso Alveolar/etiología , Pérdida de Hueso Alveolar/metabolismo , Pérdida de Hueso Alveolar/patología , Animales , Resorción Ósea/patología , Adhesión Celular/inmunología , Comunicación Celular/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunomodulación , Inflamación/etiología , Inflamación/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Activación Neutrófila/inmunología , Infiltración Neutrófila/inmunología , Neutrófilos/patología , Periodontitis/etiología , Periodontitis/metabolismo , Periodontitis/patología
20.
Semin Immunol ; 28(3): 285-91, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27021500

RESUMEN

Periodontitis is a dysbiotic inflammatory disease leading to the destruction of the tooth-supporting tissues. Current therapies are not always effective and this prevalent oral disease continues to be a significant health and economic burden. Early clinical studies have associated periodontitis with elevated complement activity. Consistently, subsequent genetic and pharmacological studies in rodents have implicated the central complement component C3 and downstream signaling pathways in periodontal host-microbe interactions that promote dysbiosis and inflammatory bone loss. This review discusses these mechanistic advances and moreover focuses on the compstatin family of C3 inhibitors as a novel approach to treat periodontitis. In this regard, local application of the current lead analog Cp40 was recently shown to block both inducible and naturally occurring periodontitis in non-human primates. These promising results from non-human primate studies and the parallel development of Cp40 for clinical use highlight the feasibility for developing an adjunctive, C3-targeted therapy for human periodontitis.


Asunto(s)
Inactivadores del Complemento/uso terapéutico , Disbiosis/terapia , Boca/inmunología , Periodontitis/terapia , Piridonas/uso terapéutico , Animales , Complemento C3/metabolismo , Complemento C5/metabolismo , Evaluación Preclínica de Medicamentos , Disbiosis/inmunología , Humanos , Boca/microbiología , Periodontitis/inmunología , Primates , Receptor de Anafilatoxina C5a/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA