1
|
Dragičević Tomičić D, Lešić N, Škrlec I, Steigmann L, Tseneva K, Čalušić Šarac M, Crnić T, Tomičić I, Perić Kačarević Ž, Čandrlić M. Effects of Vitamin D, Melatonin, and Omega-3 Fatty Acids on Periodontal Health: A Narrative Review. Dent J (Basel) 2025; 13:178. [PMID: 40277508 DOI: 10.3390/dj13040178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
Periodontitis is a chronic inflammatory disease characterized by the destruction of tooth-supporting structures, influenced by immune system dysregulation, oxidative stress, and imbalances in bone metabolism. Given its multifactorial pathogenesis, bioactive compounds such as vitamin D, melatonin, and omega-3 fatty acids have emerged as potential adjuncts to periodontal therapy due to their immunomodulatory, anti-inflammatory, and antioxidative properties. This narrative review explores the role of these three supplements in periodontal health, their potential in synergistic effects, and existing research gaps, providing a foundation for future studies on their clinical applications. Vitamin D is essential for calcium homeostasis, bone remodeling, and immune function. It modulates both innate and adaptive immune responses, enhancing antimicrobial peptide production and reducing inflammatory cytokine expression. Omega-3 fatty acids reduce the production of pro-inflammatory eicosanoids while promoting the synthesis of pro-resolving lipid mediators, contributing to bone preservation and immune balance. Melatonin, known for its antioxidant and osteogenic properties, supports bone remodeling by stimulating osteoblast proliferation and inhibiting osteoclast activity, while also regulating circadian rhythms, which may influence oral health. Although these bioactive compounds show promising effects in preclinical and clinical studies, significant knowledge gaps remain regarding optimal dosages, long-term efficacy, combined use, and standardized treatment protocols. Further clinical trials are necessary to elucidate their therapeutic value in periodontal disease management, especially those focused on their potential synergistic mechanisms. Understanding their synergistic mechanisms may open new avenues for adjunctive strategies in periodontal therapy.
Collapse
Affiliation(s)
- Dora Dragičević Tomičić
- Department of Dental Medicine, Faculty of Dental Medicine and Health Osijek, J.J. Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
| | - Nikolina Lešić
- Department of Dental Medicine, Faculty of Dental Medicine and Health Osijek, J.J. Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
| | - Ivana Škrlec
- Department of Biophysics, Biology and Chemistry, Faculty of Dental Medicine and Health Osijek, J.J. Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
| | - Larissa Steigmann
- Department of Oral Medicine, Infection and Immunity, Division of Periodontology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Kristina Tseneva
- Botiss Biomaterials AG, Ullsteinstrasse 108, 12109 Berlin, Germany
| | - Martina Čalušić Šarac
- Department of Dental Medicine, Faculty of Dental Medicine and Health Osijek, J.J. Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
- Health Center of Osijek-Baranja County, Park Kralja Petra Krešimira IV 6, 31000 Osijek, Croatia
| | - Tin Crnić
- Department of Periodontology and Operative Dentistry, University of Mainz, Augustusplatz 2, 55131 Mainz, Germany
| | - Igor Tomičić
- Department of Dental Medicine, Faculty of Dental Medicine and Health Osijek, J.J. Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
- Health Center of Osijek-Baranja County, Park Kralja Petra Krešimira IV 6, 31000 Osijek, Croatia
| | - Željka Perić Kačarević
- Botiss Biomaterials AG, Ullsteinstrasse 108, 12109 Berlin, Germany
- Department of Anatomy, Histology, Embriology, Pathology Anatomy and Pathology Histology, Faculty of Dental Medicine and Health Osijek, J.J. Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Marija Čandrlić
- Department of Integrative Dental Medicine, Faculty of Dental Medicine and Health Osijek, J.J. Strossmayer University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
2
|
Yu H, Yang S, Jiang T, Li T, Duan H, Li M. Repair mechanisms of bone system tissues based on comprehensive perspective of multi-omics. Cell Biol Toxicol 2025; 41:45. [PMID: 39966216 PMCID: PMC11836151 DOI: 10.1007/s10565-025-09995-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/28/2025] [Indexed: 02/20/2025]
Abstract
Bone disorders affect more than half of the adult population worldwide who may have a poor quality of life and physical independence worldwide. Multi-omic techniques are increasingly adopted and applied to determine the molecular mechanisms of bone tissue repair, providing perspective towards personalized medical intervention. Data from genomics, epigenomics, transcriptomics, proteomics, glycomics, and lipidomics were combined to elucidate dynamic processes in bone repair. In this narrative review, the key role of genetic and epigenetic factors in regulating injured cellular responses is highlighted, and changes in RNA and protein expression during the healing phase, as well as glucolipid metabolism adaptation, are described in detail how the repair process is affected. In a word, the integration of multi-omic techniques in this review not only benefits the comprehensive identification of new biomarkers, but also facilitates the development of personalized treatment strategies of bone disorders to revolutionize regenerative medicine.
Collapse
Affiliation(s)
- Honghao Yu
- Departments of Spine Surgery, Shengjing Hospital of China Medical University, Shengyang, China
| | - Shize Yang
- Department of Thoracic Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Tianlong Jiang
- Department of Orthopedic Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, 8 Changjiang Avenue, Tianjin, 300100, China.
| | - Hongmei Duan
- Department of Rheumatology and Immunology, First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
| | - Minglei Li
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, China.
| |
Collapse
|
3
|
Li Y, Chen W, Koo S, Liu H, Saiding Q, Xie A, Kong N, Cao Y, Abdi R, Serhan CN, Tao W. Innate immunity-modulating nanobiomaterials for controlling inflammation resolution. MATTER 2024; 7:3811-3844. [PMID: 40123651 PMCID: PMC11925551 DOI: 10.1016/j.matt.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The acute inflammatory response is an inherent protective mechanism, its unsuccessful resolution can contribute to disease pathogenesis and potentially lead to death. Innate immune cells are the first line of host defenders and play a substantial role in inflammation initiation, amplification, resolution, or subsequent disease progression. As the resolution of inflammation is an active and highly regulated process, modulating innate immune cells, including neutrophils, monocytes and macrophages, and endothelial cells, and their interactions offer opportunities to control excessive inflammation. Nanobiomaterials have shown superior therapeutic potential in inflammation-related diseases by manipulating inflammatory responses because nanobiomaterials can target and interact with innate immune cells. Versatile nanobiomaterials can be designed for targeted modulation of specific innate immune responses. Nanopro-resolving medicines have been prepared both with pro-resolving lipid mediators and peptides each demonstrated to active resolution of inflammation in animal disease models. Here, we review innovative nanobiomaterials for modulating innate immunity and alleviating inflammation. We summarise the strategies converging the design of nanobiomaterials and the nano-bio interaction in modulating innate immune profiles and propelling the advancement of nanobiomaterials for inflammatory disease treatments. We also propose the future perspectives and translational challenges of nanobiomaterials that need to be overcome in this swiftly rising field.
Collapse
Affiliation(s)
- Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- These authors contributed equally: Yongjiang Li, Wei Chen
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- These authors contributed equally: Yongjiang Li, Wei Chen
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Haijun Liu
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Angel Xie
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17177, Sweden
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Charles N. Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
4
|
Lahoud P, Faghihian H, Richert R, Jacobs R, EzEldeen M. Finite element models: A road to in-silico modeling in the age of personalized dentistry. J Dent 2024; 150:105348. [PMID: 39243802 DOI: 10.1016/j.jdent.2024.105348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024] Open
Abstract
OBJECTIVE This article reviews the applications of Finite Element Models (FEMs) in personalized dentistry, focusing on treatment planning, material selection, and CAD-CAM processes. It also discusses the challenges and future directions of using finite element analysis (FEA) in dental care. DATA This study synthesizes current literature and case studies on FEMs in personalized dentistry, analyzing research articles, clinical reports, and technical papers on the application of FEA in dental biomechanics. SOURCES Sources for this review include peer-reviewed journals, academic publications, clinical case studies, and technical papers on dental biomechanics and finite element analysis. Key databases such as PubMed, Scopus, Embase, and ArXiv were used to identify relevant studies. STUDY SELECTION Studies were selected based on their relevance to the application of FEMs in personalized dentistry. Inclusion criteria were studies that discussed the use of FEA in treatment planning, material selection, and CAD-CAM processes in dentistry. Exclusion criteria included studies that did not focus on personalized dental treatments or did not utilize FEMs as a primary tool. CONCLUSIONS FEMs are essential for personalized dentistry, offering a versatile platform for in-silico dental biomechanics modeling. They can help predict biomechanical behavior, optimize treatment outcomes, and minimize clinical complications. Despite needing further advancements, FEMs could help significantly enhance treatment precision and efficacy in personalized dental care. CLINICAL SIGNIFICANCE FEMs in personalized dentistry hold the potential to significantly improve treatment precision and efficacy, optimizing outcomes and reducing complications. Their integration underscores the need for interdisciplinary collaboration and advancements in computational techniques to enhance personalized dental care.
Collapse
Affiliation(s)
- P Lahoud
- OMFS-IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, Leuven, Belgium; Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium; Division of Periodontology and Oral Microbiology, Department of Oral Health Sciences, KU Leuven, Leuven, Belgium.
| | - H Faghihian
- Department of Odontology, Faculty of Medicine, Umeå Universitet, Umeå, Sweden.
| | - R Richert
- Hospices Civils de Lyon, PAM Odontologie, Lyon, France; Laboratoire de Mécanique Des Contacts Et Structures LaMCoS, UMR 5259 INSA Lyon, CNRS, Villeurbanne 69621, France.
| | - R Jacobs
- OMFS-IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, Leuven, Belgium; Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Dental Medicine, Karolinska Institute, Stockholm, Sweden.
| | - M EzEldeen
- OMFS-IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, Leuven, Belgium; Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Oral Health Sciences, KU Leuven and Paediatric Dentistry and Special Dental Care, University Hospitals Leuven, KU Leuven, Leuven, Belgium.
| |
Collapse
|
5
|
Rovai EDS, Polassi M, da Silveira MI, Araújo SL, Dyke TV, dos Santos NCC. Impact of Specialized Pro-Resolving Lipid Mediators on Craniofacial and Alveolar Bone Regeneration: Scoping Review. Braz Dent J 2024; 35:e246133. [PMID: 39476116 PMCID: PMC11506308 DOI: 10.1590/0103-6440202406133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/01/2024] [Indexed: 11/03/2024] Open
Abstract
Craniofacial bone defects caused by tumors, trauma, long-term tooth loss, or periodontal disease are a major challenge in the field of tissue engineering. In periodontitis and peri-implantitis, reconstructive therapy is also a major challenge for the dental surgeon. Lipoxins, resolvins, protectins, and maresins, known as specialized pro-resolving lipid mediators (SPMs), have been widely studied in the field of dental, oral, and craniofacial research for bone regeneration for their actions in restoring tissue homeostasis and promoting tissue healing and regeneration. Therefore, this study focuses on a survey of the use of SPMs for craniofacial and alveolar bone regeneration. Thus, electronic searches of five databases were performed to identify pre-clinical studies that evaluated the actions of SMPs on craniofacial and alveolar bone regeneration. Of the 523 articles retrieved from the electronic databases, 19 were included in the analysis. Resolvin (Rv) E1 was the mostly assessed SPM (n=8), followed by maresins (Ma) R1 (n=3), lipoxins (Lx) A4 (n=3), RvD1 (n=3), RvD2 (n=1), LxB4 (n=1), and maresin (M)-CTR3 (n=1). Meta-analysis showed that SPMs increased the newly formed bone by 14.85% compared to the control group (p<0.00001), decreased the area of the remaining defect by 0.35 mm2 (p<0.00001), and decreased the linear distance between the defect to the bone crest by 0.53 mm (p<0.00001). RvE1 reduced inflammatory bone resorption in periodontal defects and calvarial osteolysis and enhanced bone regeneration when RvE1 was combined with a bovine bone graft. RvD2 induced active resolution of inflammation and tissue regeneration in periapical lesions, while RvD1 controlled the inflammatory microenvironment in calvarial defects in rats, promoting bone healing and angiogenesis. MaR1 induced the proliferation and migration of mesenchymal stem cells, osteogenesis, and angiogenesis in calvarial defects, and benzo (b)-LxA4 and LxA4 promoted bone regeneration calvarial and alveolar bone defects in rats, inducing regeneration under inflammatory conditions. In summary, SPMs have emerged as pivotal contributors to the resolution of inflammation and the facilitation of bone neoformation within craniofacial and alveolar bone defects. These results are based on pre-clinical studies, in vivo and in vitro, and provide an updated review regarding the impact of SPMs in tissue engineering.
Collapse
Affiliation(s)
- Emanuel da Silva Rovai
- Department of Diagnosis and Surgery, Institute of Science and Technology, São Paulo State University, São José dos Campos, SP, Brazil
| | - Mackeler Polassi
- Dental Research Division, Guarulhos University, Guarulhos, SP, Brazil
| | - Marcela Iunes da Silveira
- Department of Diagnosis and Surgery, Institute of Science and Technology, São Paulo State University, São José dos Campos, SP, Brazil
| | - Sandy Lima Araújo
- Dental Research Division, Guarulhos University, Guarulhos, SP, Brazil
| | - Thomas Van Dyke
- The ADA Forsyth Institute, Cambridge, MA, United States
- Harvard School of Dental Medicine, Harvard University, Boston, MA, United States
| | - Nidia C. Castro dos Santos
- Dental Research Division, Guarulhos University, Guarulhos, SP, Brazil
- Albert Einstein School of Dental Medicine, Albert Einstein Israelite Hospital, São Paulo, SP, Brazil
- The ADA Forsyth Institute, Cambridge, MA, United States
| |
Collapse
|
6
|
Peh HY, Nshimiyimana R, Brüggemann TR, Duvall MG, Nijmeh J, Serhan CN, Levy BD. 15-epi-lipoxin A 5 promotes neutrophil exit from exudates for clearance by splenic macrophages. FASEB J 2024; 38:e23807. [PMID: 38989570 PMCID: PMC11344644 DOI: 10.1096/fj.202400610r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/10/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Specialized proresolving mediators (SPMs) promote local macrophage efferocytosis but excess leukocytes early in inflammation require additional leukocyte clearance mechanism for resolution. Here, neutrophil clearance mechanisms from localized acute inflammation were investigated in mouse dorsal air pouches. 15-HEPE (15-hydroxy-5Z,8Z,11Z,13E,17Z-eicosapentaenoic acid) levels were increased in the exudates. Activated human neutrophils converted 15-HEPE to lipoxin A5 (5S,6R,15S-trihydroxy-7E,9E,11Z,13E,17Z-eicosapentaenoic acid), 15-epi-lipoxin A5 (5S,6R,15R-trihydroxy-7E,9E,11Z,13E,17Z-eicosapentaenoic acid), and resolvin E4 (RvE4; 5S,15S-dihydroxy-6E,8Z,11Z,13E,17Z-eicosapentaenoic acid). Exogenous 15-epi-lipoxin A5, 15-epi-lipoxin A4 and a structural lipoxin mimetic significantly decreased exudate neutrophils and increased local tissue macrophage efferocytosis, with comparison to naproxen. 15-epi-lipoxin A5 also cleared exudate neutrophils faster than the apparent local capacity for stimulated macrophage efferocytosis, so the fate of exudate neutrophils was tracked with CD45.1 variant neutrophils. 15-epi-lipoxin A5 augmented the exit of adoptively transferred neutrophils from the pouch exudate to the spleen, and significantly increased splenic SIRPa+ and MARCO+ macrophage efferocytosis. Together, these findings demonstrate new systemic resolution mechanisms for 15-epi-lipoxin A5 and RvE4 in localized tissue inflammation, which distally engage the spleen to activate macrophage efferocytosis for the clearance of tissue exudate neutrophils.
Collapse
Affiliation(s)
- Hong Yong Peh
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Robert Nshimiyimana
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Thayse R. Brüggemann
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Melody G. Duvall
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Julie Nijmeh
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Charles N. Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Bruce D. Levy
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
7
|
Golub LM, Lee HM, Bacigalupo J, Gu Y. Host modulation therapy in periodontitis, diagnosis and treatment-status update. FRONTIERS IN DENTAL MEDICINE 2024; 5:1423401. [PMID: 39917713 PMCID: PMC11797899 DOI: 10.3389/fdmed.2024.1423401] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/01/2024] [Indexed: 02/09/2025] Open
Affiliation(s)
- Lorne M. Golub
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Hsi-Ming Lee
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, NY, United States
| | | | - Ying Gu
- Department of General Dentistry, School of Dental Medicine, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
8
|
Li Q, Wang D, Xiao C, Wang H, Dong S. Advances in Hydrogels for Periodontitis Treatment. ACS Biomater Sci Eng 2024; 10:2742-2761. [PMID: 38639082 DOI: 10.1021/acsbiomaterials.4c00220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Periodontitis is a common condition characterized by a bacterial infection and the disruption of the body's immune-inflammatory response, which causes damage to the teeth and supporting tissues and eventually results in tooth loss. Current therapy involves the systemic and local administration of antibiotics. However, the existing treatments cannot exert effective, sustained release and maintain an effective therapeutic concentration of the drug at the lesion site. Hydrogels are used to treat periodontitis due to their low cytotoxicity, exceptional water retention capability, and controlled drug release profile. Hydrogels can imitate the extracellular matrix of periodontal cells while offering suitable sites to load antibiotics. This article reviews the utilization of hydrogels for periodontitis therapy based on the pathogenesis and clinical manifestations of the disease. Additionally, the latest therapeutic strategies for smart hydrogels and the main techniques for hydrogel preparation have been discussed. The information will aid in designing and preparing future hydrogels for periodontitis treatment.
Collapse
Affiliation(s)
- Qiqi Li
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Di Wang
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Hao Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Shujun Dong
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| |
Collapse
|
9
|
Ma R, Liu Y, Xu Y, Duan D. Lipoxin A4 levels predict site-specific clinical improvements post scaling and root planing and correlate negatively with periodontal pathogens in severe periodontitis. BMC Oral Health 2024; 24:204. [PMID: 38331747 PMCID: PMC10851498 DOI: 10.1186/s12903-024-03948-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 01/27/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Serving as a stop signal of inflammation, the role of lipoxin A4 (LXA4) in periodontitis remains to be clarified. This study is aimed to examine the changes in LXA4 levels in gingival crevicular fluid (GCF) after scaling and root planing (SRP) and to determine the relationship between LXA4 levels and treatment outcomes and periodontal pathogens in severe periodontitis. METHODS A total of 74 GCF samples were collected from 21 severe periodontitis participants at the deepest affected sites. These sites were re-sampled at 1, 3, and 6 months after SRP. Besides, GCF samples were also collected from 25 periodontally healthy participants. Clinical parameters including probing depth (PD) and clinical attachment level (CAL) in periodontitis group were recorded. LXA4 levels and periodontal pathogens in the GCF were analyzed by ELISA and PCR, respectively. Correlations between GCF LXA4 levels and treatment effect and periodontal pathogens were assessed. RESULTS LXA4 levels in GCF significantly increased after SRP (p < 0.05), but remained lower than those observed in healthy individuals (p < 0.05). Sites with lower baseline LXA4 concentrations were more likely to experience greater improvements in PD at 6 months post-SRP (area under the curve [AUC] = 0.792), and the improvements were positively correlated with the increase of LXA4 at these sites post-treatment (p < 0.05). Furthermore, more elevated LXA4 levels were observed in sites that became negative for Prevotella intermedia or Tannerella forsythia after SRP. CONCLUSION Baseline LXA4 in GCF has the potential to predict the site-specific response of severe periodontal lesions to SRP. The increase of LXA4 levels after treatment was positively correlated with clinical improvements and negatively correlated with the presence of Prevotella intermedia or Tannerella forsythia.
Collapse
Affiliation(s)
- Rui Ma
- State Key Laboratory of Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, No. 14, Section 3, Renmin South Road, Chengdu, 610041, China
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yiying Liu
- State Key Laboratory of Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, No. 14, Section 3, Renmin South Road, Chengdu, 610041, China
| | - Yi Xu
- State Key Laboratory of Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, No. 14, Section 3, Renmin South Road, Chengdu, 610041, China
| | - Dingyu Duan
- State Key Laboratory of Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, No. 14, Section 3, Renmin South Road, Chengdu, 610041, China.
| |
Collapse
|
10
|
Jahangirnezhad M, Mahmoudinezhad SS, Moradi M, Moradi K, Rohani A, Tayebi L. Bone Scaffold Materials in Periodontal and Tooth-supporting Tissue Regeneration: A Review. Curr Stem Cell Res Ther 2024; 19:449-460. [PMID: 36578254 DOI: 10.2174/1574888x18666221227142055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVES Periodontium is an important tooth-supporting tissue composed of both hard (alveolar bone and cementum) and soft (gingival and periodontal ligament) sections. Due to the multi-tissue architecture of periodontium, reconstruction of each part can be influenced by others. This review focuses on the bone section of the periodontium and presents the materials used in tissue engineering scaffolds for its reconstruction. MATERIALS AND METHODS The following databases (2015 to 2021) were electronically searched: ProQuest, EMBASE, SciFinder, MRS Online Proceedings Library, Medline, and Compendex. The search was limited to English-language publications and in vivo studies. RESULTS Eighty-three articles were found in primary searching. After applying the inclusion criteria, seventeen articles were incorporated into this study. CONCLUSION In complex periodontal defects, various types of scaffolds, including multilayered ones, have been used for the functional reconstruction of different parts of periodontium. While there are some multilayered scaffolds designed to regenerate alveolar bone/periodontal ligament/cementum tissues of periodontium in a hierarchically organized construct, no scaffold could so far consider all four tissues involved in a complete periodontal defect. The progress and material considerations in the regeneration of the bony part of periodontium are presented in this work to help investigators develop tissue engineering scaffolds suitable for complete periodontal regeneration.
Collapse
Affiliation(s)
- Mahmood Jahangirnezhad
- Department of Periodontics, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sadaf Sadat Mahmoudinezhad
- Department of Periodontics, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Melika Moradi
- Department of Periodontics, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kooshan Moradi
- Department of Periodontics, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Rohani
- Department of Periodontics, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Lobat Tayebi
- School of Dentistry, Marquette University, Milwaukee, WI, 53233, USA
| |
Collapse
|
11
|
Geisinger ML. NexGen regen? Challenges and opportunities for growth factors and signaling agents in periodontal regeneration at intrabony defects. FRONTIERS IN DENTAL MEDICINE 2023; 4:1239149. [PMID: 39916925 PMCID: PMC11797756 DOI: 10.3389/fdmed.2023.1239149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/18/2023] [Indexed: 02/09/2025] Open
Abstract
Regeneration of periodontal tissues that have been destroyed by inflammatory periodontitis involves the initiation of tissue engineering and wound healing of multiple tissues involved in the function of the teeth, including the periodontal ligament, cementum, and alveolar bone. Such regeneration is termed guided tissue regeneration and the unique challenges to reconstruct these tissues involve a complex interplay of cells, signaling molecules, and scaffolds. While traditional guided tissue regeneration treatments have involved cell occlusive membranes, bone replacement graft scaffolds, and endogenous multipotent mesenchymal stem cells, the use of adjunctive materials to enhance healing outcomes has been studied and many such adjunctive factors are in common current clinical use. This report will focus on the current and emerging adjunctive growth factors and signaling molecules that can be used to optimize periodontal regeneration in periodontal intrabony defects, their mechanisms of action, the challenges associated with periodontal regeneration, and future avenues for research.
Collapse
Affiliation(s)
- Maria L. Geisinger
- Department of Periodontology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
12
|
Viglianisi G, Santonocito S, Lupi SM, Amato M, Spagnuolo G, Pesce P, Isola G. Impact of local drug delivery and natural agents as new target strategies against periodontitis: new challenges for personalized therapeutic approach. Ther Adv Chronic Dis 2023; 14:20406223231191043. [PMID: 37720593 PMCID: PMC10501082 DOI: 10.1177/20406223231191043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/07/2023] [Indexed: 09/19/2023] Open
Abstract
Periodontitis is a persistent inflammation of the soft tissue around the teeth that affects 60% of the population in the globe. The self-maintenance of the inflammatory process can cause periodontal damage from the alveolar bone resorption to tooth loss in order to contrast the effects of periodontitis, the main therapy used is scaling and root planing (SRP). At the same time, studying the physiopathology of periodontitis has shown the possibility of using a local drug delivery system as an adjunctive therapy. Using local drug delivery devices in conjunction with SRP therapy for periodontitis is a potential tool since it increases drug efficacy and minimizes negative effects by managing drug release. This review emphasized how the use of local drug delivery agents and natural agents could be promising adjuvants for the treatment of periodontitis patients affected or not by cardiovascular disease, diabetes, and other system problems. Moreover, the review evidences the current issues and new ideas that can inspire potential later study for both basic research and clinical practice for a tailored approach.
Collapse
Affiliation(s)
- Gaia Viglianisi
- Department of General Surgery and Surgical-Medical Specialities, School of Dentistry, University of Catania, Catania, Italy
| | - Simona Santonocito
- Department of General Surgery and Surgical-Medical Specialities, School of Dentistry, University of Catania, Catania, Italy
| | - Saturnino Marco Lupi
- Department of Clinical Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Mariacristina Amato
- Department of General Surgery and Surgical-Medical Specialities, School of Dentistry, University of Catania, Catania, Italy
| | - Gianrico Spagnuolo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, Naples, Italy
| | - Paolo Pesce
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialities, School of Dentistry, University of Catania, Via Santa Sofia 78, Catania 95123, Italy
| |
Collapse
|
13
|
Lee CT, Tribble GD. Roles of specialized pro-resolving mediators and omega-3 polyunsaturated fatty acids in periodontal inflammation and impact on oral microbiota. FRONTIERS IN ORAL HEALTH 2023; 4:1217088. [PMID: 37559676 PMCID: PMC10409488 DOI: 10.3389/froh.2023.1217088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/11/2023] [Indexed: 08/11/2023] Open
Abstract
Periodontitis is a chronic inflammatory disease induced by dysbiotic dental biofilms. Management of periodontitis is primarily anti-bacterial via mechanical removal of bacterial biofilm. The successful resolution requires wound healing and tissue regeneration, which are not always achieved with these traditional methods. The discovery of specialized pro-resolving mediators (SPMs), a class of lipid mediators that induce the resolution of inflammation and promote local tissue homeostasis, creates another option for the treatment of periodontitis and other diseases of chronic inflammation. In this mini-review, we discuss the host-modulatory effects of SPMs on periodontal tissues and changes in the taxonomic composition of the gut and oral microbiome in the presence of SPMs and SPM precursor lipids. Further research into the relationship between host SPM production and microbiome-SPM modification has the potential to unveil new diagnostic markers of inflammation and wound healing. Expanding this field may drive the discovery of microbial-derived bioactive therapeutics to modulate immune responses.
Collapse
Affiliation(s)
- Chun-Teh Lee
- Department of Periodontics and Dental Hygiene, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Gena D. Tribble
- Department of Periodontics and Dental Hygiene, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
14
|
Eltay EG, Van Dyke T. Resolution of inflammation in oral diseases. Pharmacol Ther 2023:108453. [PMID: 37244405 DOI: 10.1016/j.pharmthera.2023.108453] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/11/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
The resolution of inflammation is an essential endogenous process that protects host tissues from an exaggerated chronic inflammatory response. Multiple interactions between host cells and resident oral microbiome regulate the protective functions that lead to inflammation in the oral cavity. Failure of appropriate regulation of inflammation can lead to chronic inflammatory diseases that result from an imbalance between pro-inflammatory and pro-resolution mediators. Thus, failure of the host to resolve inflammation can be considered an essential pathological mechanism for progression from the late stages of acute inflammation to a chronic inflammatory response. Specialized pro-resolving mediators (SPMs), which are essential polyunsaturated fatty acid (PUFA)-derived autacoid mediators, aid in regulating the endogenous inflammation resolving process by stimulating immune cell-mediated clearance of apoptotic polymorphonuclear neutrophils, cellular debris, and microbes, restricting further neutrophil tissue infiltration, and counter-regulating pro-inflammatory cytokine production. The SPM superfamily contains four specialized lipid mediator families: lipoxins, resolvins, protectins, and maresins that can activate resolution pathways. Understanding the crosstalk between resolution signals in the tissue response to injury has therapeutic application potential for preventing, maintaining, and regenerating chronically damaged tissues. Here, we discuss the fundamental concepts of resolution as an active biochemical process, novel concepts demonstrating the role of resolution mediators in tissue regeneration in periodontal and pulpal diseases, and future directions for therapeutic applications with particular emphasis on periodontal therapy.
Collapse
Affiliation(s)
- Eiba G Eltay
- Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA, United States; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, United States
| | - Thomas Van Dyke
- Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA, United States; Center for Clinical and Translational Research, The Forsyth Institute, Cambridge, MA, United States; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, United States.
| |
Collapse
|
15
|
Wang C, Zhao Q, Chen C, Li J, Zhang J, Qu S, Tang H, Zeng H, Zhang Y. CD301b + macrophage: the new booster for activating bone regeneration in periodontitis treatment. Int J Oral Sci 2023; 15:19. [PMID: 37198150 DOI: 10.1038/s41368-023-00225-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/16/2023] [Accepted: 03/28/2023] [Indexed: 05/19/2023] Open
Abstract
Periodontal bone regeneration is a major challenge in the treatment of periodontitis. Currently the main obstacle is the difficulty of restoring the regenerative vitality of periodontal osteoblast lineages suppressed by inflammation, via conventional treatment. CD301b+ macrophages were recently identified as a subpopulation that is characteristic of a regenerative environment, but their role in periodontal bone repair has not been reported. The current study indicates that CD301b+ macrophages may be a constituent component of periodontal bone repair, and that they are devoted to bone formation in the resolving phase of periodontitis. Transcriptome sequencing suggested that CD301b+ macrophages could positively regulate osteogenesis-related processes. In vitro, CD301b+ macrophages could be induced by interleukin 4 (IL-4) unless proinflammatory cytokines such as interleukin 1β (IL-1β) and tumor necrosis factor α (TNF-α) were present. Mechanistically, CD301b+ macrophages promoted osteoblast differentiation via insulin-like growth factor 1 (IGF-1)/thymoma viral proto-oncogene 1 (Akt)/mammalian target of rapamycin (mTOR) signaling. An osteogenic inducible nano-capsule (OINC) consisting of a gold nanocage loaded with IL-4 as the "core" and mouse neutrophil membrane as the "shell" was designed. When injected into periodontal tissue, OINCs first absorbed proinflammatory cytokines in inflamed periodontal tissue, then released IL-4 controlled by far-red irradiation. These events collectively promoted CD301b+ macrophage enrichment, which further boosted periodontal bone regeneration. The current study highlights the osteoinductive role of CD301b+ macrophages, and suggests a CD301b+ macrophage-targeted induction strategy based on biomimetic nano-capsules for improved therapeutic efficacy, which may also provide a potential therapeutic target and strategy for other inflammatory bone diseases.
Collapse
Affiliation(s)
- Can Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qin Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Chen Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jiaojiao Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jing Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Shuyuan Qu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hua Tang
- Institute of Infection and Immunity, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, PR China
| | - Hao Zeng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
16
|
Zarrough AE, Hasturk H, Stephens DN, Van Dyke TE, Kantarci A. Resolvin D1 modulates periodontal ligament fibroblast function. J Periodontol 2023; 94:683-693. [PMID: 36416879 PMCID: PMC10354588 DOI: 10.1002/jper.22-0462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/05/2022] [Accepted: 11/13/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND The resolution of inflammation is an active process mediated by specialized lipid mediators called lipoxins and resolvins. Periodontal ligament fibroblasts (PDLFs) play a significant role in periodontal regeneration. The purpose of the current study was to determine the impact of resolvin D1 (RvD1) on human PDLF cell wound healing and proliferation, receptor expression (G-protein-coupled receptor 32 [GPR32] and formyl peptide receptor 2 [ALX/FPR2]), and cytokine expression and release. METHODS PDLFs were stimulated with interleukin-1β (IL-1β) (500 pg/ml) with and without RvD1 (100 nM). RvD1 receptor expression was determined by quantitative real-time polymerase chain reaction (qPCR), immunofluorescence microscopy, and fluorescence-activated cell sorting. Wound closure was measured by a scratch assay, and proliferation was determined by bromodeoxyuridine incorporation. Interleukin-6 (IL-6), interleukin-8 (IL-8), monocyte chemoattractant protein-1, cyclooxygenase-2, matrix metalloproteinases-1, -2, and -3 (MMP-1, -2, and -3), tissue inhibitors of metalloproteinases-1 and -2 (TIMP-1 and -2), prostaglandin E2, and osteoprotegerin (OPG) gene expression and production were measured using qPCR and Western blotting, multiplex immunoassay, and enzyme-linked immunosorbent assay. RESULTS PDLF expressed GPR32 and ALX/FPR2. RvD1 reversed IL-1β-induced inhibition of wound healing and proliferation of PDLF. IL-1β also induced the production of proinflammatory cytokines and MMPs. This effect was reversed by RvD1. RvD1 reversed IL-1β-induced inhibition of TIMP-1, TIMP-2, and OPG. CONCLUSION The data suggested that RvD1 has a pro-wound healing, proliferative, and anti-inflammatory impact on the PDLF that favors periodontal regeneration.
Collapse
Affiliation(s)
- Ahmed E. Zarrough
- Missouri School of Dentistry & Oral Health, A.T. Still University, St. Louis, Missouri, USA
| | - Hatice Hasturk
- Department of Clinical and Translational Research, Forsyth Institute, Cambridge, Massachusetts, USA
| | - Danielle N. Stephens
- Department of Clinical and Translational Research, Forsyth Institute, Cambridge, Massachusetts, USA
| | - Thomas E. Van Dyke
- Department of Clinical and Translational Research, Forsyth Institute, Cambridge, Massachusetts, USA
| | - Alpdogan Kantarci
- Department of Clinical and Translational Research, Forsyth Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
17
|
Tahmasebi E, Mohammadi M, Alam M, Abbasi K, Gharibian Bajestani S, Khanmohammad R, Haseli M, Yazdanian M, Esmaeili Fard Barzegar P, Tebyaniyan H. The current regenerative medicine approaches of craniofacial diseases: A narrative review. Front Cell Dev Biol 2023; 11:1112378. [PMID: 36926524 PMCID: PMC10011176 DOI: 10.3389/fcell.2023.1112378] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/08/2023] [Indexed: 03/08/2023] Open
Abstract
Craniofacial deformities (CFDs) develop following oncological resection, trauma, or congenital disorders. Trauma is one of the top five causes of death globally, with rates varying from country to country. They result in a non-healing composite tissue wound as they degenerate in soft or hard tissues. Approximately one-third of oral diseases are caused by gum disease. Due to the complexity of anatomical structures in the region and the variety of tissue-specific requirements, CFD treatments present many challenges. Many treatment methods for CFDs are available today, such as drugs, regenerative medicine (RM), surgery, and tissue engineering. Functional restoration of a tissue or an organ after trauma or other chronic diseases is the focus of this emerging field of science. The materials and methodologies used in craniofacial reconstruction have significantly improved in the last few years. A facial fracture requires bone preservation as much as possible, so tiny fragments are removed initially. It is possible to replace bone marrow stem cells with oral stem cells for CFDs due to their excellent potential for bone formation. This review article discusses regenerative approaches for different types of craniofacial diseases.
Collapse
Affiliation(s)
- Elahe Tahmasebi
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehdi Mohammadi
- School of Dentistry, Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mostafa Alam
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamyar Abbasi
- Department of Prosthodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Gharibian Bajestani
- Student Research Committee, Dentistry Research Center, Research Institute of Dental Sciences, Dental School, Shahid Behesti University of Medical Sciences, Tehran, Iran
| | - Rojin Khanmohammad
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohsen Haseli
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohsen Yazdanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Hamid Tebyaniyan
- Department of Science and Research, Islimic Azade University, Tehran, Iran
| |
Collapse
|
18
|
Perretti M, Dalli J. Resolution Pharmacology: Focus on Pro-Resolving Annexin A1 and Lipid Mediators for Therapeutic Innovation in Inflammation. Annu Rev Pharmacol Toxicol 2023; 63:449-469. [PMID: 36151051 DOI: 10.1146/annurev-pharmtox-051821-042743] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Chronic diseases that affect our society are made more complex by comorbidities and are poorly managed by the current pharmacology. While all present inflammatory etiopathogeneses, there is an unmet need for better clinical management of these diseases and their multiple symptoms. We discuss here an innovative approach based on the biology of the resolution of inflammation. Studying endogenous pro-resolving peptide and lipid mediators, how they are formed, and which target they interact with, can offer innovative options through augmenting the expression or function of pro-resolving pathways or mimicking their actions with novel targeted molecules. In all cases, resolution offers innovation for the treatment of the primary cause of a given disease and/or for the management of its comorbidities, ultimately improving patient quality of life. By implementing resolution pharmacology, we harness the whole physiology of inflammation, with the potential to bring a marked change in the management of inflammatory conditions.
Collapse
Affiliation(s)
- Mauro Perretti
- The William Harvey Research Institute, Faculty of Medicine and Dentistry, and Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom; ,
| | - Jesmond Dalli
- The William Harvey Research Institute, Faculty of Medicine and Dentistry, and Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom; ,
| |
Collapse
|
19
|
Shichiri M, Suzuki H, Isegawa Y, Tamai H. Application of regulation of reactive oxygen species and lipid peroxidation to disease treatment. J Clin Biochem Nutr 2023; 72:13-22. [PMID: 36777080 PMCID: PMC9899923 DOI: 10.3164/jcbn.22-61] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/02/2022] [Indexed: 11/05/2022] Open
Abstract
Although many diseases in which reactive oxygen species (ROS) and free radicals are involved in their pathogenesis are known, and antioxidants that effectively capture ROS have been identified and developed, there are only a few diseases for which antioxidants have been used for treatment. Here, we discuss on the following four concepts regarding the development of applications for disease treatment by regulating ROS, free radicals, and lipid oxidation with the findings of our research and previous reports. Concept 1) Utilization of antioxidants for disease treatment. In particular, the importance of the timing of starting antioxidant will be discussed. Concept 2) Therapeutic strategies using ROS and free radicals. Methods of inducing ferroptosis, which has been advocated as an iron-dependent cell death, are mentioned. Concept 3) Treatment with drugs that inhibit the synthesis of lipid mediators. In addition to the reduction of inflammatory lipid mediators by inhibiting cyclooxygenase and leukotriene synthesis, we will introduce the possibility of disease treatment with lipoxygenase inhibitors. Concept 4) Disease treatment by inducing the production of useful lipid mediators for disease control. We describe the treatment of inflammatory diseases utilizing pro-resolving mediators and propose potential compounds that activate lipoxygenase to produce these beneficial mediators.
Collapse
Affiliation(s)
- Mototada Shichiri
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan
| | - Hiroshi Suzuki
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Yuji Isegawa
- Department of Food Sciences and Nutrition, Mukogawa Women’s University, 6-46 Ikebiraki, Nishinomiya, Hyogo 663-8558, Japan
| | - Hiroshi Tamai
- Department of Pediatrics, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| |
Collapse
|
20
|
Hajishengallis G, Chavakis T. Mechanisms and Therapeutic Modulation of Neutrophil-Mediated Inflammation. J Dent Res 2022; 101:1563-1571. [PMID: 35786033 PMCID: PMC9703529 DOI: 10.1177/00220345221107602] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Neutrophils are abundant, short-lived myeloid cells that are readily recruitable to sites of inflammation, where they serve as first-line defense against infection and other types of insult to the host. In recent years, there has been increased understanding on the involvement of neutrophils in chronic inflammatory diseases, where they may act as direct effectors of destructive inflammation. However, destructive tissue inflammation is also instigated in settings of neutrophil paucity, suggesting that neutrophils also mediate critical homeostatic functions. The activity of neutrophils is regulated by a variety of local tissue factors. In addition, systemic metabolic conditions, such as hypercholesterolemia and hyperglycemia, affect the production and mobilization of neutrophils from the bone marrow. Moreover, according to the recently emerged concept of innate immune memory, the functions of neutrophils can be enhanced through the process of trained granulopoiesis. This process may have both beneficial and potentially destructive effects, depending on context, that is, protective against infections and tumors, while destructive in the context of chronic inflammatory conditions. Although we are far from a complete understanding of the mechanisms underlying the regulation and function of neutrophils, current insights enable the development of targeted therapeutic interventions that can restrain neutrophil-mediated inflammation in chronic inflammatory diseases, such as periodontitis.
Collapse
Affiliation(s)
- G. Hajishengallis
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - T. Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Sachsen, Germany
| |
Collapse
|
21
|
Lyu H, Zhou X, Qian Y, Liu X, Gopinathan G, Pandya M, Qin C, Luan X, Diekwisch TGH. Long-acting PFI-2 small molecule release and multilayer scaffold design achieve extensive new formation of complex periodontal tissues with unprecedented fidelity. Biomaterials 2022; 290:121819. [PMID: 36209579 DOI: 10.1016/j.biomaterials.2022.121819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 08/20/2022] [Accepted: 09/19/2022] [Indexed: 11/02/2022]
Abstract
The faithful engineering of complex human tissues such as the bone/soft tissue/mineralized tissue interface in periodontal tissues requires innovative molecular cues in conjunction with tailored scaffolds. To address the loss of periodontal bone and connective tissues following periodontal disease, we have generated a polydopamine and collagen coated electrospun PLGA-PCL (PP) scaffold enriched with the small molecule mediator PFI-2 (PP-PFI-pDA-COL-PFI). In vitro 3D studies using PDL progenitors revealed that the PP-PFI-pDA-COL-PFI scaffold substantially enhanced Alizarin Red staining, increased Ca/P ratios 4-fold, and stimulated cell proliferation more than 12-fold compared to PP-controls, suggestive of its potential for mineralized tissue engineering. When applied in our experimental periodontitis model, the PP-PFI-pDA-COL-PFI scaffold resulted in a substantial 34% reduction in alveolar bone defect height, a 25% root-length gain in periodontal attachment, and the formation of highly ordered regenerated acellular cementum twice as thick as in controls. Explaining the mechanism of PFI-2 mineralized tissue regeneration in periodontal tissues, PFI-2 inhibited SETD7-mediated β-Catenin protein methylation and increased β-Catenin nuclear localization. Together, dual-level PFI-2 incorporation into a degradable, dopamine/collagen coated PLGA/PCL scaffold backbone resulted in the regeneration of the tripartite periodontal complex with unprecedented fidelity, including periodontal attachment and new formation of mineralized tissues in inflamed periodontal environments.
Collapse
Affiliation(s)
- Huling Lyu
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, TX, USA; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Xuefeng Zhou
- UIC College of Dentistry, Department of Oral Biology, Chicago, IL, USA; State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yunzhu Qian
- UIC College of Dentistry, Department of Oral Biology, Chicago, IL, USA; Center for Stomatology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Xiaohua Liu
- Department of Biomedical Sciences, Texas A&M College of Dentistry, Dallas, TX, USA
| | - Gokul Gopinathan
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, TX, USA
| | - Mirali Pandya
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, TX, USA
| | - Chunlin Qin
- Department of Biomedical Sciences, Texas A&M College of Dentistry, Dallas, TX, USA
| | - Xianghong Luan
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, TX, USA; UIC College of Dentistry, Department of Oral Biology, Chicago, IL, USA
| | - Thomas G H Diekwisch
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, TX, USA; UIC College of Dentistry, Department of Oral Biology, Chicago, IL, USA.
| |
Collapse
|
22
|
Kalkar P, Cohen G, Tamari T, Schif-Zuck S, Zigdon-Giladi H, Ariel A. IFN-β mediates the anti-osteoclastic effect of bisphosphonates and dexamethasone. Front Pharmacol 2022; 13:1002550. [PMID: 36386129 PMCID: PMC9648992 DOI: 10.3389/fphar.2022.1002550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/30/2022] [Indexed: 07/06/2024] Open
Abstract
Zoledronic acid (Zol) is a potent bisphosphonate that inhibits the differentiation of monocytes into osteoclasts. It is often used in combination with dexamethasone (Dex), a glucocorticoid that promotes the resolution of inflammation, to treat malignant diseases, such as multiple myeloma. This treatment can result in bone pathologies, namely medication related osteonecrosis of the jaw, with a poor understanding of the molecular mechanism on monocyte differentiation. IFN-β is a pro-resolving cytokine well-known as an osteoclast differentiation inhibitor. Here, we explored whether Zol and/or Dex regulate macrophage osteoclastic differentiation via IFN-β. RAW 264.7 and peritoneal macrophages were treated with Zol and/or Dex for 4-24 h, and IFN-β secretion was examined by ELISA, while the IFN stimulated gene (ISG) 15 expression was evaluated by Western blotting. RANKL-induced osteoclastogenesis of RAW 264.7 cells was determined by TRAP staining following treatment with Zol+Dex or IFN-β and anti-IFN-β antibodies. We found only the combination of Zol and Dex increased IFN-β secretion by RAW 264.7 macrophages at 4 h and, correspondingly, ISG15 expression in these cells at 24 h. Moreover, Zol+Dex blocked osteoclast differentiation to a similar extent as recombinant IFN-β. Neutralizing anti-IFN-β antibodies reversed the effect of Zol+Dex on ISG15 expression and partially recovered osteoclastic differentiation induced by each drug alone or in combination. Finally, we found Zol+Dex also induced IFN-β expression in peritoneal resolution phase macrophages, suggesting these drugs might be used to enhance the resolution of acute inflammation. Altogether, our findings suggest Zol+Dex block the differentiation of osteoclasts through the expression of IFN-β. Revealing the molecular pathway behind this regulation may lead to the development of IFN-β-based therapy to inhibit osteoclastogenesis in multiple myeloma patients.
Collapse
Affiliation(s)
- Prajakta Kalkar
- Departments of Biology and Human Biology, University of Haifa, Haifa, Israel
| | - Gal Cohen
- Departments of Biology and Human Biology, University of Haifa, Haifa, Israel
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Tal Tamari
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Sagie Schif-Zuck
- Departments of Biology and Human Biology, University of Haifa, Haifa, Israel
| | - Hadar Zigdon-Giladi
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Amiram Ariel
- Departments of Biology and Human Biology, University of Haifa, Haifa, Israel
| |
Collapse
|
23
|
Rakian A, Rakian R, Shay A, Serhan C, Van Dyke T. Periodontal Stem Cells Synthesize Maresin Conjugate in Tissue Regeneration 3. J Dent Res 2022; 101:1205-1213. [PMID: 35428422 PMCID: PMC9403725 DOI: 10.1177/00220345221090879] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023] Open
Abstract
Periodontal disease is a significant public health problem worldwide. Excess unresolved chronic inflammation destroys the periodontal tissues that surround and support the teeth, and efforts to control inflammation by removal of bacterial deposits on the teeth have limited long-term impact. Likewise, procedures aimed at regeneration of the periodontal tissues have shown limited success. Recent advances in stem cell research have shown promising novel prospects for the use of periodontal ligament stem cells (PDLSCs) in tissue regeneration; however, control of inflammation remains a barrier. Human PDLSCs have been shown to release specialized proresolving lipid mediators (SPMs) that modulate the immune response and promote resolution of inflammation, tissue repair, and regeneration. Studies on stem cell biology in periodontology have also been limited by the lack of a good large animal model. Herein, we describe PDLSC biology of the Yorkshire pig (pPDLSCs). pPDLSCs were isolated and characterized. Using lipid mediator profiling, we demonstrate for the first time that pPDLSCs biosynthesize cysteinyl-containing SPMs (cys-SPMs), specifically, maresin conjugates in tissue regeneration 3 (MCTR3) and its authentication using liquid chromatography/tandem mass spectrometry. The exogenous addition of the n-3 precursor docosahexaenoic acid enhances MCTR3 biosynthesis. Using immunocytochemistry, we show that pPDLSCs express 4 of the SPM biosynthetic pathway enzymes necessary for SPM biosynthesis, including 5-lipoxygenase, 12-lipoxygenase, and 15-lipoxygenase-1. In addition, we identified and quantified the cytokine/chemokine profile of pPDLSCs using a 13-plex immunology multiplex assay and found that the pretreatment of pPDLSCs with MCTR3 in an inflammatory environment reduced the production of acute and chronic proinflammatory cytokines/chemokines. Together, these results suggest that enhancing resolution of inflammation pathways and mediators may be a possible key early event in predictable periodontal regeneration.
Collapse
Affiliation(s)
- A. Rakian
- Department of Applied Oral Science, The Forsyth Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - R. Rakian
- Department of Applied Oral Science, The Forsyth Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - A.E. Shay
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - C.N. Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - T.E. Van Dyke
- Department of Applied Oral Science, The Forsyth Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
24
|
Pamuk F, Kantarci A. Inflammation as a link between periodontal disease and obesity. Periodontol 2000 2022; 90:186-196. [PMID: 35916870 DOI: 10.1111/prd.12457] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nutrition plays a critical role in the homeostatic balance, maintenance of health, and longevity. There is a close link between inflammatory diseases and nutritional health. Obesity is a severe pathological process with grave implications on several organ systems and disease processes, including type 2 diabetes, cardiovascular disease, osteoarthritis, and rheumatoid arthritis. The impact of obesity on periodontal inflammation has not been fully understood; the association between nutritional balance and periodontal inflammation is much less explored. This review is focused on the potential mechanistic links between periodontal diseases and obesity and common inflammatory activity pathways that can be pharmacologically targeted.
Collapse
Affiliation(s)
- Ferda Pamuk
- Forsyth Institute, Cambridge, Massachusetts, USA.,Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium
| | | |
Collapse
|
25
|
Batool F, Gegout PY, Stutz C, White B, Kolodziej A, Benkirane-Jessel N, Petit C, Huck O. Lenabasum Reduces Porphyromonas gingivalis-Driven Inflammation. Inflammation 2022; 45:1752-1764. [PMID: 35274214 DOI: 10.1007/s10753-022-01658-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 01/05/2023]
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.
Collapse
Affiliation(s)
- Fareeha Batool
- Faculté de Chirurgie-Dentaire, Université de Strasbourg, 8 rue Sainte-Elisabeth, 67000, Strasbourg, France.,UMR 1260, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM (French National Institute of Health and Medical Research), Regenerative Nanomedicine, Strasbourg, France
| | - Pierre-Yves Gegout
- Faculté de Chirurgie-Dentaire, Université de Strasbourg, 8 rue Sainte-Elisabeth, 67000, Strasbourg, France
| | - Céline Stutz
- Faculté de Chirurgie-Dentaire, Université de Strasbourg, 8 rue Sainte-Elisabeth, 67000, Strasbourg, France.,UMR 1260, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM (French National Institute of Health and Medical Research), Regenerative Nanomedicine, Strasbourg, France
| | | | | | - Nadia Benkirane-Jessel
- UMR 1260, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM (French National Institute of Health and Medical Research), Regenerative Nanomedicine, Strasbourg, France
| | - Catherine Petit
- Faculté de Chirurgie-Dentaire, Université de Strasbourg, 8 rue Sainte-Elisabeth, 67000, Strasbourg, France.,UMR 1260, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM (French National Institute of Health and Medical Research), Regenerative Nanomedicine, Strasbourg, France.,Pôle de Médecine Et Chirurgie Bucco-Dentaire, Hôpitaux Universitaires de Strasbourg, 67000, Strasbourg, France
| | - Olivier Huck
- Faculté de Chirurgie-Dentaire, Université de Strasbourg, 8 rue Sainte-Elisabeth, 67000, Strasbourg, France. .,UMR 1260, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM (French National Institute of Health and Medical Research), Regenerative Nanomedicine, Strasbourg, France. .,Pôle de Médecine Et Chirurgie Bucco-Dentaire, Hôpitaux Universitaires de Strasbourg, 67000, Strasbourg, France.
| |
Collapse
|
26
|
Bezerra B, Monajemzadeh S, Silva D, Pirih FQ. Modulating the Immune Response in Periodontitis. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.879131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Periodontitis is a chronic inflammatory condition initiated by the accumulation of bacterial biofilm. It is highly prevalent and when left untreated can lead to tooth loss. The presence of bacterial biofilm is essential for the initiation of the inflammatory response but is not the sole initiator. Currently it is unknown which mechanisms drive the dysbiosis of the bacterial biofilm leading to the dysregulation of the inflammatory response. Other players in this equation include environmental, systemic, and genetic factors which can play a role in exacerbating the inflammatory response. Treatment of periodontal disease consists of removal of the bacterial biofilm with the goal of resolving the inflammatory response; however, this does not occur in every case. Understanding the way the inflammatory response does not return to a state of homeostasis has led investigators to consider both systemic and local pharmacological interventions. Nonetheless, a better understanding of the impact that genetics and environmental factors may have on the inflammatory response could be key to helping identify how inflammation can be modulated therefore stopping the destruction of the periodontium. In this article, we will explore the current evidence associating the microbial dysbiosis and the dysregulation of the immune response, potential mechanisms or pathways that may be targeted for the modulation of the inflammatory response, and discuss the advantages and drawbacks associated with local and systemic inflammatory modulation in the management of periodontal disease. This information will be valuable for those interested in understanding potential adjunct methods for managing periodontal diseases, but not limited to, dental professionals, clinical researchers and the public at large.
Collapse
|
27
|
Resolution of inflammation: Intervention strategies and future applications. Toxicol Appl Pharmacol 2022; 449:116089. [DOI: 10.1016/j.taap.2022.116089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 11/23/2022]
|
28
|
Vanzant E, Loftus T, Kamel A, Carmichael E, Rosenthal MD. Nutritional impact of omega 3 fatty acids and metabolites in acute and chronic critical illness. Curr Opin Clin Nutr Metab Care 2022; 25:75-80. [PMID: 35115447 DOI: 10.1097/mco.0000000000000818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Lipids have been utilized historically as a calorie dense means to ensure delivery of essential fatty acids (FA). Since the development of mixed lipid emulsion and investigation of immunomodulatory formulas, there has been an awakening that not all lipids are created equal. This narrative review focuses on contemporary evidence in the utilization of lipids (namely omega 3 fatty acids) in both acute and chronic critical illness. RECENT FINDINGS Though randomized control trials and meta-analyses provide little guidance regarding clinical practice for patients suffering from chronic critical illness, available literature suggests the potential to use lipid formulas to decrease the inflammatory cycle that drives catabolism. Additionally, this review will address the expanding evidence that specialized pro-resolving mediators (SPMs) may be the future of immunomodulating inflammation in acute and chronic critical illness and the persistent inflammation, immunosuppression, and catabolic syndrome (PICS). SUMMARY Although societal guidelines, expert consensus, and literature support the use of omega 3 fatty acids in the acute critically ill population, more research is needed regarding omega 3 fatty acids for chronic critical illness and PICS.
Collapse
Affiliation(s)
- Erin Vanzant
- Division of Trauma and Acute Care Surgery, Department of Surgery, College of Medicine, University of Florida
| | - Tyler Loftus
- Division of Trauma and Acute Care Surgery, Department of Surgery, College of Medicine, University of Florida
| | - Amir Kamel
- Department of Pharmacy, UF Health Shands Hospital, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Ethan Carmichael
- Division of Trauma and Acute Care Surgery, Department of Surgery, College of Medicine, University of Florida
| | - Martin D Rosenthal
- Division of Trauma and Acute Care Surgery, Department of Surgery, College of Medicine, University of Florida
| |
Collapse
|
29
|
Yadalam PK, Kalaivani V, Fageeh HI, Ibraheem W, Al-Ahmari MM, Khan SS, Ahmed ZH, Abdulkarim HH, Baeshen HA, Balaji TM, Bhandi S, Raj AT, Patil S. Future Drug Targets in Periodontal Personalised Medicine-A Narrative Review. J Pers Med 2022; 12:371. [PMID: 35330371 PMCID: PMC8955099 DOI: 10.3390/jpm12030371] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/22/2022] [Accepted: 02/26/2022] [Indexed: 02/08/2023] Open
Abstract
Periodontal disease is an infection-driven inflammatory disease characterized by the destruction of tooth-supporting tissues. The establishment of chronic inflammation will result in progressive destruction of bone and soft tissue changes. Severe periodontitis can lead to tooth loss. The disease has complex pathogenesis with an interplay between genetic, environmental, and host factors and pathogens. Effective management consists of plaque control and non-surgical interventions, along with adjuvant strategies to control inflammation and disrupt the pathogenic subgingival biofilms. Recent studies have examined novel approaches for managing periodontal diseases such as modulating microbial signaling mechanisms, tissue engineering, and molecular targeting of host inflammatory substances. Mounting evidence suggests the need to integrate omics-based approaches with traditional therapy to address the disease. This article discusses the various evolving and future drug targets, including proteomics, gene therapeutics, vaccines, and nanotechnology in personalized periodontal medicine for the effective management of periodontal diseases.
Collapse
Affiliation(s)
- Pradeep Kumar Yadalam
- Department of Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602117, India;
| | - V. Kalaivani
- Department of Periodontics, SRM Kattankulathur Dental College & Hospital, SRM Nagar, Chennai 603203, India;
| | - Hammam Ibrahim Fageeh
- Department of Preventive Dental Sciences, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia; (H.I.F.); (W.I.)
| | - Wael Ibraheem
- Department of Preventive Dental Sciences, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia; (H.I.F.); (W.I.)
| | - Manea Musa. Al-Ahmari
- Department of Periodontics and Community Medical Science, College of Dentistry, King Khalid University, Abha 61421, Saudi Arabia;
| | - Samar Saeed Khan
- Department of Maxillofacial Surgery & Diagnostic Sciences, Division of Oral Pathology, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia;
| | - Zeeshan Heera Ahmed
- Department of Restorative Dental Sciences, College of Dentistry, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Hesham H. Abdulkarim
- Advanced Periodontal and Dental Implant Care, Missouri School of Dentistry and Oral Health, A. T. Still University, St. Louis, MO 63104, USA;
| | - Hosam Ali Baeshen
- Department of Orthodontics, College of Dentistry, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | | | - Shilpa Bhandi
- Department of Restorative Dental Sciences, Division of Operative Dentistry, College of dentistry, Jazan University, Jazan 45142, Saudi Arabia;
| | - A. Thirumal Raj
- Department of Oral Pathology and Microbiology, Sri Venkateswara Dental College and Hospital, Chennai 600130, India;
| | - Shankargouda Patil
- Department of Maxillofacial Surgery & Diagnostic Sciences, Division of Oral Pathology, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia;
| |
Collapse
|
30
|
Cecconello C, Clària Ribas P, Norling LV. Resolving acute inflammation; what happens when inflammation goes haywire? How can it get back in line? DIET, INFLAMMATION, AND HEALTH 2022:113-162. [DOI: 10.1016/b978-0-12-822130-3.00018-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
31
|
Edranov SS, Matveeva NY, Kalinichenko SG. On-Bone Fixation of Free Gingival Graft Induces an Osteoinductive Effect in Human Alveolar Bone. Bull Exp Biol Med 2021; 172:276-281. [PMID: 34855081 DOI: 10.1007/s10517-021-05375-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Indexed: 12/26/2022]
Abstract
We examined alveolar bone samples in the area of on-bone fixation of a free gingival graft performed during surgery in patients aged 37-55 years with a diagnosis of secondary partial adentia of the upper and lower jaws. Six months after fixation of the graft in the alveolar bone, foci of neoosteogenesis were found in the contact zone. They were characterized by the appearance of appositional lines, cords of basophilic osteoblasts, and growing osteons. An immunohistochemical study revealed an increase in the number of CD44+, CD29+, and osteocalcin+ cells in the layer of the outer circumferential lamellae, primary osteons, and the lining of the Haversian canals. TGF-β1+ cells were located in the intertrabecular reticular tissue and wall of microvessels. The results indicate activation of mesenchymal stem cells in the area of localization of the graft and differentiating osteoblasts. The observed osteoinductive effect of free gingival graft is associated with its participation in reorganization in MSC and induction of morphogenetic molecules.
Collapse
Affiliation(s)
- S S Edranov
- Department of Histology, Embryology, and Cytology, Pacific State Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N Yu Matveeva
- Department of Histology, Embryology, and Cytology, Pacific State Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - S G Kalinichenko
- Department of Histology, Embryology, and Cytology, Pacific State Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
32
|
Vaernewyck V, Arzi B, Sanders NN, Cox E, Devriendt B. Mucosal Vaccination Against Periodontal Disease: Current Status and Opportunities. Front Immunol 2021; 12:768397. [PMID: 34925337 PMCID: PMC8675580 DOI: 10.3389/fimmu.2021.768397] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
Approximately 9 out of 10 adults have some form of periodontal disease, an infection-induced inflammatory disease of the tooth-supporting tissues. The initial form, gingivitis, often remains asymptomatic, but this can evolve into periodontitis, which is typically associated with halitosis, oral pain or discomfort, and tooth loss. Furthermore, periodontitis may contribute to systemic disorders like cardiovascular disease and type 2 diabetes mellitus. Control options remain nonspecific, time-consuming, and costly; largely relying on the removal of dental plaque and calculus by mechanical debridement. However, while dental plaque bacteria trigger periodontal disease, it is the host-specific inflammatory response that acts as main driver of tissue destruction and disease progression. Therefore, periodontal disease control should aim to alter the host's inflammatory response as well as to reduce the bacterial triggers. Vaccines may provide a potent adjunct to mechanical debridement for periodontal disease prevention and treatment. However, the immunopathogenic complexity and polymicrobial aspect of PD appear to complicate the development of periodontal vaccines. Moreover, a successful periodontal vaccine should induce protective immunity in the oral cavity, which proves difficult with traditional vaccination methods. Recent advances in mucosal vaccination may bridge the gap in periodontal vaccine development. In this review, we offer a comprehensive overview of mucosal vaccination strategies to induce protective immunity in the oral cavity for periodontal disease control. Furthermore, we highlight the need for additional research with appropriate and clinically relevant animal models. Finally, we discuss several opportunities in periodontal vaccine development such as multivalency, vaccine formulations, and delivery systems.
Collapse
Affiliation(s)
- Victor Vaernewyck
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Boaz Arzi
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA, United States
- Veterinary Institute for Regenerative Cures (VIRC) School of Veterinary Medicine, University of California, Davis, CA, United States
| | - Niek N. Sanders
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Eric Cox
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Bert Devriendt
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
33
|
Abstract
Technological innovations in cellular and molecular aspects of tissue engineering --scaffolds, stem cells and 3D printed tissues --have been dramatically increased in the last decade. However, regenerative treatment still has challenges in translation to clinic. This is partly due to failure of addressing an essential element of wound healing, inflammation. It is now well-recognized that inflammation is an active process. This paradigm shift opened up a new avenue of therapeutic approaches called "host-modulation." Host-modulation therapies capable of modulating inflammatory response at multiple levels and mimicking the natural sequence of wound healing offer a new direction and promising clinical translation.
Collapse
|
34
|
Yu N, Rakian A, Dean A, Van Dyke TE. Specialized Proresolving Mediators Facilitate the Immunomodulation of the Periodontal Ligament Stem Cells. FRONTIERS IN DENTAL MEDICINE 2021. [DOI: 10.3389/fdmed.2021.701197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recent investigations into the regulation of the inflammation in the periodontitis have revealed that chronic inflammatory diseases such as periodontitis are characterized by an imbalance in the proinflammatory and proresolution mediators and can be characterized by a failure of the resolution pathways in the late stages of the acute inflammatory response. The proresolution mediators, termed as specialized proresolving mediators (SPMs), comprise the lipoxins, resolvins, protectins, and maresins that are derived from the arachidonic acid or omega-3 polyunsaturated fatty acids. In the animal studies, treatment of the periodontitis with the topical SPMs return the inflammatory lesion to the homeostasis with the regeneration of all the components of the periodontal organ lost to the disease. In this article, the study investigates the immunomodulatory role of SPMs in the periodontal ligament stem cells (PDLSCs). Primary porcine PDLSCs (pPDLSCs) were stimulated with interleukin-1β (IL-1β) and interleukin-17 (IL-17) in vitro to simulate the periodontal inflammation in the presence or absence of SPMs. This study found that IL-1β and IL-17 synergistically activated the proinflammatory genes of pPDLSCs and altered the immune phenotype of pPDLSCs including the key signaling pathways. Addition of SPMs rescued the pPDLSCs phenotype and induced further production of the additional SPMs, which was reflected by upregulation of the requisite enzymes 12- and 15-lipoxygenase by pPDLSCs. This study interrogated the immunomodulatory actions of pPDLSCs on the monocytes/macrophages, focusing on the porcine CD14/CD16/CD163 markers by using flow cytometry. This study utilized the CD14+CD16+/CD14+CD16− ratio and CD163 on the monocytes/macrophages to differentiate between a proinflammation phenotype (lower ratio) and a resolution of the inflammation phenotype (higher ratio). This study also found that the conditioned medium from pPDLSCs treated with the cytokines and Maresin1 increased the CD14+CD16+/CD14+CD16− ratio and had the highest CD163 expression. This study concludes that in an inflammatory environment, pPDLSCs become proinflammatory and exert immunomodulatory functions. Maresin 1 resolves the inflammation by acting on pPDLSCs directly and by shifting the monocytes/macrophages phenotype to the proresolution dominance.
Collapse
|
35
|
Panigrahy D, Gilligan MM, Serhan CN, Kashfi K. Resolution of inflammation: An organizing principle in biology and medicine. Pharmacol Ther 2021; 227:107879. [PMID: 33915177 DOI: 10.1016/j.pharmthera.2021.107879] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023]
Abstract
The resolution of inflammation has emerged as a critical endogenous process that protects host tissues from prolonged or excessive inflammation that can become chronic. Failure of the resolution of inflammation is a key pathological mechanism that drives the progression of numerous inflammation-driven diseases. Essential polyunsaturated fatty acid (PUFA)-derived autacoid mediators termed 'specialized pro-resolving mediators' (SPMs) regulate endogenous resolution programs by limiting further neutrophil tissue infiltration and stimulating local immune cell (e.g., macrophage)-mediated clearance of apoptotic polymorphonuclear neutrophils, cellular debris, and microbes, as well as counter-regulating eicosanoid/cytokine production. The SPM superfamily encompasses lipoxins, resolvins, protectins, and maresins. Our understanding of the resolution phase of acute inflammation has grown exponentially in the past three decades with the discovery of novel pro-resolving lipid mediators, their pro-efferocytosis mechanisms, and their receptors. Technological advancement has further facilitated lipid mediator metabolipidomic based profiling of healthy and diseased human tissues, highlighting the extraordinary therapeutic potential of SPMs across a broad array of inflammatory diseases including cancer. As current front-line cancer therapies such as surgery, chemotherapy, and radiation may induce various unwanted side effects such as robust pro-inflammatory and pro-tumorigenic host responses, characterizing SPMs and their receptors as novel therapeutic targets may have important implications as a new direction for host-targeted cancer therapy. Here, we discuss the origins of inflammation resolution, key discoveries and the failure of resolution mechanisms in diseases with an emphasis on cancer, and future directions focused on novel therapeutic applications for this exciting and rapidly expanding field.
Collapse
Affiliation(s)
- Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Molly M Gilligan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, City University of New York, School of Medicine, New York, NY 10031, USA; Graduate Program in Biology, City University of New York Graduate Center, New York, NY 10016, USA
| |
Collapse
|
36
|
Deng J, Golub LM, Lee HM, Raja V, Johnson F, Kucine A, Lee W, Xu TM, Gu Y. A Novel Modified-Curcumin Promotes Resolvin-Like Activity and Reduces Bone Loss in Diabetes-Induced Experimental Periodontitis. J Inflamm Res 2021; 14:5337-5347. [PMID: 34703272 PMCID: PMC8528548 DOI: 10.2147/jir.s330157] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/29/2021] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Clinically, it is challenging to manage diabetic patients with periodontitis. Biochemically, both involve a wide range of inflammatory/collagenolytic conditions which exacerbate each other in a "bi-directional manner." However, standard treatments for this type of periodontitis rely on reducing the bacterial burden and less on controlling hyper-inflammation/excessive-collagenolysis. Thus, there is a crucial need for new therapeutic strategies to modulate this excessive host response and to promote enhanced resolution of inflammation. The aim of the current study is to evaluate the impact of a novel chemically-modified curcumin 2.24 (CMC2.24) on host inflammatory response in diabetic rats. METHODS Type I diabetes was induced by streptozotocin injection; periodontal breakdown then results as a complication of uncontrolled hyperglycemia. Non-diabetic rats served as controls. CMC2.24, or the vehicle-alone, was administered by oral gavage daily for 3 weeks to the diabetics. Micro-CT was used to analyze morphometric changes and quantify bone loss. MMPs were analyzed by gelatin zymography. Cell function was examined by cell migration assay, and cytokines and resolvins were measured by ELISA. RESULTS In this severe inflammatory disease model, administration of the pleiotropic CMC2.24 was found to normalize the excessive accumulation and impaired chemotactic activity of macrophages in peritoneal exudates, significantly decrease MMP-9 and pro-inflammatory cytokines to near normal levels, and markedly increase resolvin D1 (RvD1) levels in the thioglycolate-elicited peritoneal exudates (tPE). Similar effects on MMPs and RvD1 were observed in the non-elicited resident peritoneal washes (rPW). Regarding clinical relevance, CMC2.24 significantly inhibited the loss of alveolar bone height, volume and mineral density (ie, diabetes-induced periodontitis and osteoporosis). CONCLUSION In conclusion, treating hyperglycemic diabetic rats with CMC2.24 (a tri-ketonic phenylaminocarbonyl curcumin) promotes the resolution of local and systemic inflammation, reduces bone loss, in addition to suppressing collagenolytic MMPs and pro-inflammatory cytokines, suggesting a novel therapeutic strategy for treating periodontitis complicated by other chronic diseases.
Collapse
Affiliation(s)
- Jie Deng
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, People’s Republic of China
| | - Lorne M Golub
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Hsi-Ming Lee
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Veena Raja
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Francis Johnson
- Department of Chemistry and Pharmacological Sciences, School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Allan Kucine
- Department of Oral & Maxillofacial Surgery, School of Dental Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Wonsae Lee
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Tian-Min Xu
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, People’s Republic of China
| | - Ying Gu
- Department of General Dentistry, School of Dental Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| |
Collapse
|
37
|
Hasturk H, Schulte F, Martins M, Sherzai H, Floros C, Cugini M, Chiu CJ, Hardt M, Van Dyke T. Safety and Preliminary Efficacy of a Novel Host-Modulatory Therapy for Reducing Gingival Inflammation. Front Immunol 2021; 12:704163. [PMID: 34589083 PMCID: PMC8475270 DOI: 10.3389/fimmu.2021.704163] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 08/23/2021] [Indexed: 12/22/2022] Open
Abstract
Background Periodontal disease is among the sixth most common inflammatory diseases worldwide with high risk to promote complications from other inflammatory diseases including diabetes, cardiovascular disease and Alzheimer’s Disease. Failure of active resolution of inflammation pathways is implicated in pathogenesis of periodontal diseases, including gingivitis. Lipoxin A4 (LXA4), a member of the specialized pro-resolving lipid mediators (SPMs) that drive resolution of inflammation via GPC-receptor mediated pathways, offered therapeutic advantages in preclinical models of periodontitis. Methods We conducted a randomized, placebo-controlled, parallel-group Phase 1 clinical trial to determine the safety and preliminary efficacy of an LXA4 analog in patients with gingival inflammation. One hundred twenty-seven (127) individuals were randomized to daily use of an oral rinse containing a LXA4 mimetic, methyl ester-benzo-lipoxin A4 (BLXA4), placebo rinse or a no-rinse control group for 28 days. Treatment emergent adverse events (TEAEs) were assessed for safety, the primary outcome. Secondary outcomes included the change in the level of gingival inflammation and periodontal pocket depth (PD). Serum SPMs were monitored using targeted lipid mediator lipidomics to assess potential systemic impact of BLXA4. Results The frequency of TEAEs was similar in BLXA4 and placebo-treated groups with no study-related SAEs. Once-daily rinsing with BLXA4 for 28-days resulted in a greater decrease in gingival inflammation compared to placebo rinse and no-rinse control groups (mean change: 0.26 GI unit vs 0.21 and 0.17, respectively). PD reduction was also greater with BLXA4 oral rinse compared to placebo and no-rinse groups (mean reduction: 1.23 mm vs. 0.71 mm and 0.46 mm, respectively). Topical application of BLXA4 increased serum levels of SPMs. Conclusion Treatment with BLXA4 reduces local inflammation, and increases abundance of pro-resolution molecules systemically, which may dampen inflammation that can mediate progression and course of inflammatory diseases beyond periodontitis. Clinical Trial Registration ClinicalTrials.gov, identifier (NCT02342691).
Collapse
Affiliation(s)
- Hatice Hasturk
- Center for Clinical and Translational Research, The Forsyth Institute, Cambridge, MA, United States.,Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA, United States
| | - Fabian Schulte
- Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA, United States.,Center for Salivary Diagnostics, The Forsyth Institute, Cambridge, MA, United States.,Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, United States
| | - Melissa Martins
- Center for Clinical and Translational Research, The Forsyth Institute, Cambridge, MA, United States.,Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA, United States
| | - Homa Sherzai
- Center for Clinical and Translational Research, The Forsyth Institute, Cambridge, MA, United States.,Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA, United States
| | - Constantinos Floros
- Center for Clinical and Translational Research, The Forsyth Institute, Cambridge, MA, United States.,Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA, United States
| | - MaryAnn Cugini
- Center for Clinical and Translational Research, The Forsyth Institute, Cambridge, MA, United States.,Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA, United States
| | - Chung-Jung Chiu
- Epidemiology and Biostatistics, The Forsyth Institute, Cambridge, MA, United States
| | - Markus Hardt
- Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA, United States.,Center for Salivary Diagnostics, The Forsyth Institute, Cambridge, MA, United States.,Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, United States
| | - Thomas Van Dyke
- Center for Clinical and Translational Research, The Forsyth Institute, Cambridge, MA, United States.,Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA, United States.,Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, United States
| |
Collapse
|
38
|
Haas AN, Furlaneto F, Gaio EJ, Gomes SC, Palioto DB, Castilho RM, Sanz M, Messora MR. New tendencies in non-surgical periodontal therapy. Braz Oral Res 2021; 35:e095. [PMID: 34586209 DOI: 10.1590/1807-3107bor-2021.vol35.0095] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022] Open
Abstract
The aim of this review was to update the evidence of new approaches to non-surgical therapy (NSPT) in the treatment of periodontitis. Preclinical and clinical studies addressing the benefits of adjunctive antimicrobial photodynamic therapy, probiotics, prebiotics/synbiotics, statins, pro-resolving mediators, omega-6 and -3, ozone, and epigenetic therapy were scrutinized and discussed. Currently, the outcomes of these nine new approaches, when compared with subgingival debridement alone, did not demonstrate a significant added clinical benefit. However, some of these new alternative interventions may have the potential to improve the outcomes of NSPT alone. Future evidence based on randomized controlled clinical trials would help clinicians and patients in the selection of different adjunctive therapies.
Collapse
Affiliation(s)
- Alex Nogueira Haas
- Universidade Federal do Rio Grande do Sul - UFRGS, School of Dentistry, Department of Periodontology, Porto Alegre, RS, Brazil
| | - Flavia Furlaneto
- Universidade de São Paulo - USP, School of Dentistry of Ribeirão Preto, Department of Oral Surgery and Periodontology, Ribeirão Preto, SP, Brazil
| | - Eduardo José Gaio
- Universidade Federal do Rio Grande do Sul - UFRGS, School of Dentistry, Department of Periodontology, Porto Alegre, RS, Brazil
| | - Sabrina Carvalho Gomes
- Universidade Federal do Rio Grande do Sul - UFRGS, School of Dentistry, Department of Periodontology, Porto Alegre, RS, Brazil
| | - Daniela Bazan Palioto
- Universidade de São Paulo - USP, School of Dentistry of Ribeirão Preto, Department of Oral Surgery and Periodontology, Ribeirão Preto, SP, Brazil
| | - Rogerio Moraes Castilho
- Michigan University, School of Dentistry, Department of Periodontics and Oral Medicine, Ann Arbor, MI, USA
| | - Mariano Sanz
- Complutense University of Madrid, Etiology and Therapy of Periodontal and Peri-implant Diseases Research Group, Madrid, Spain
| | - Michel Reis Messora
- Universidade de São Paulo - USP, School of Dentistry of Ribeirão Preto, Department of Oral Surgery and Periodontology, Ribeirão Preto, SP, Brazil
| |
Collapse
|
39
|
Newman H, Shih YV, Varghese S. Resolution of inflammation in bone regeneration: From understandings to therapeutic applications. Biomaterials 2021; 277:121114. [PMID: 34488119 DOI: 10.1016/j.biomaterials.2021.121114] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/10/2021] [Accepted: 08/28/2021] [Indexed: 12/12/2022]
Abstract
Impaired bone healing occurs in 5-10% of cases following injury, leading to a significant economic and clinical impact. While an inflammatory response upon injury is necessary to facilitate healing, its resolution is critical for bone tissue repair as elevated acute or chronic inflammation is associated with impaired healing in patients and animal models. This process is governed by important crosstalk between immune cells through mediators that contribute to resolution of inflammation in the local healing environment. Approaches modulating the initial inflammatory phase followed by its resolution leads to a pro-regenerative environment for bone regeneration. In this review, we discuss the role of inflammation in bone repair, the negative impact of dysregulated inflammation on bone tissue regeneration, and how timely resolution of inflammation is necessary to achieve normal healing. We will discuss applications of biomaterials to treat large bone defects with a specific focus on resolution of inflammation to modulate the immune environment following bone injury, and their observed functional benefits. We conclude the review by discussing future strategies that could lead to the realization of anti-inflammatory therapeutics for bone tissue repair.
Collapse
Affiliation(s)
- Hunter Newman
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27710, USA
| | - Yuru Vernon Shih
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Shyni Varghese
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27710, USA; Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC, 27710, USA.
| |
Collapse
|
40
|
Kang Y, Sun B, Chen Y, Lou Y, Zheng M, Li Z. Dental Plaque Microbial Resistomes of Periodontal Health and Disease and Their Changes after Scaling and Root Planing Therapy. mSphere 2021; 6:e0016221. [PMID: 34287005 PMCID: PMC8386447 DOI: 10.1128/msphere.00162-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 05/23/2021] [Indexed: 11/20/2022] Open
Abstract
The human oral microbial community has been considered a reservoir of antibiotic resistance. Currently, the effects of periodontitis and the scaling and root planing (SRP) treatment on the performance of antibiotic-resistant genes (ARGs) and metal-resistant genes (MRGs) in the dental plaque microbiota are not well characterized. To explore this issue, we selected 48 healthy-state (HS), 40 periodontitis-state (PS; before treatment), and 24 resolved-state (RS; after SRP treatment) metagenomic data of dental plaque samples from the Sequence Read Archive (SRA) database. NetShift analysis identified Fretibacterium fastidiosum, Tannerella forsythia, and Campylobacter rectus as key drivers during dental plaque microbiota alteration in the progression of periodontitis. Periodontitis and SRP treatment resulted in an increase in the number of ARGs and MRGs in dental plaque and significantly altered the composition of ARG and MRG profiles. Bacitracin, beta-lactam, macrolide-lincosamide-streptogramin (MLS), tetracycline, and multidrug resistance genes were the main classes of ARGs with high relative abundance, whereas multimetal, iron, chromium, and copper resistance genes were the primary types of MRGs in dental plaque microbiota. The cooccurrence of ARGs, MRGs, and mobile genetic elements (MGEs) indicated that a coselection phenomenon exists in the resistomes of dental plaque microbiota. Overall, our data provide new insights into the standing of the distribution of ARGs and MRGs in oral microbiota of periodontitis patients, and it was possible to contribute to the understanding of the complicated correlations among microorganisms, resistomes, and MGEs. IMPORTANCE The emergence and development of resistance to antibiotics in periodontal pathogens have affected the success rate of treatment for periodontitis. The development of new antibacterial strategies is urgently needed to help control and treat periodontal disease, and dental plaque microbiome studies offer a promising new angle of attack. In this study, we investigated the dental plaque microbiota and resistomes in periodontal health and disease states and their changes after SRP therapy. This is the first analysis of the profile of the microbial community and antibiotic and metal resistance genes in dental plaque by the metagenomic approach, to the best of our knowledge. Monitoring the profile of these resistomes has huge potential to provide reference levels for proper antibiotics use and the development of new antimicrobial strategies in periodontitis therapy and thereby improve actual efficacy of the treatment regimens.
Collapse
Affiliation(s)
- Yutong Kang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bianjin Sun
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, China
| | - Yiju Chen
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongliang Lou
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Meiqin Zheng
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, China
| | - Zhenjun Li
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
41
|
Aubeux D, Peters OA, Hosseinpour S, Tessier S, Geoffroy V, Pérez F, Gaudin A. Specialized pro-resolving lipid mediators in endodontics: a narrative review. BMC Oral Health 2021; 21:276. [PMID: 34030680 PMCID: PMC8142493 DOI: 10.1186/s12903-021-01619-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 05/09/2021] [Indexed: 02/06/2023] Open
Abstract
Endodontics is the branch of dentistry concerned with the morphology, physiology, and pathology of the human dental pulp and periradicular tissues. Human dental pulp is a highly dynamic tissue equipped with a network of resident immunocompetent cells that play major roles in the defense against pathogens and during tissue injury. However, the efficiency of these mechanisms during dental pulp inflammation (pulpitis) varies due to anatomical and physiological restrictions. Uncontrolled, excessive, or unresolved inflammation can lead to pulp tissue necrosis and subsequent bone infections called apical periodontitis. In most cases, pulpitis treatment consists of total pulp removal. Although this strategy has a good success rate, this treatment has some drawbacks (lack of defense mechanisms, loss of healing capacities, incomplete formation of the root in young patients). In a sizeable number of clinical situations, the decision to perform pulp extirpation and endodontic treatment is justifiable by the lack of therapeutic tools that could otherwise limit the immune/inflammatory process. In the past few decades, many studies have demonstrated that the resolution of acute inflammation is necessary to avoid the development of chronic inflammation and to promote repair or regeneration. This active process is orchestrated by Specialized Pro-resolving lipid Mediators (SPMs), including lipoxins, resolvins, protectins and maresins. Interestingly, SPMs do not have direct anti-inflammatory effects by inhibiting or directly blocking this process but can actively reduce neutrophil infiltration into inflamed tissues, enhance efferocytosis and bacterial phagocytosis by monocytes and macrophages and simultaneously inhibit inflammatory cytokine production. Experimental clinical application of SPMs has shown promising result in a wide range of inflammatory diseases, such as renal fibrosis, cerebral ischemia, marginal periodontitis, and cancer; the potential of SPMs in endodontic therapy has recently been explored. In this review, our objective was to analyze the involvement and potential use of SPMs in endodontic therapies with an emphasis on SPM delivery systems to effectively administer SPMs into the dental pulp space.
Collapse
Affiliation(s)
- Davy Aubeux
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, 44042, Nantes, France
- Université de Nantes, UFR Odontologie, 44042, Nantes, France
| | - Ove A Peters
- School of Dentistry, The University of Queensland, Brisbane, Australia
| | | | - Solène Tessier
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, 44042, Nantes, France
- Université de Nantes, UFR Odontologie, 44042, Nantes, France
| | - Valérie Geoffroy
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, 44042, Nantes, France
- Université de Nantes, UFR Odontologie, 44042, Nantes, France
| | - Fabienne Pérez
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, 44042, Nantes, France
- Université de Nantes, UFR Odontologie, 44042, Nantes, France
- CHU Nantes, PHU4 OTONN44093, Nantes, France
| | - Alexis Gaudin
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, 44042, Nantes, France.
- Université de Nantes, UFR Odontologie, 44042, Nantes, France.
- CHU Nantes, PHU4 OTONN44093, Nantes, France.
| |
Collapse
|
42
|
Cardoso EOC, Fine N, Glogauer M, Johnson F, Goldberg M, Golub LM, Tenenbaum HC. The Advent of COVID-19; Periodontal Research Has Identified Therapeutic Targets for Severe Respiratory Disease; an Example of Parallel Biomedical Research Agendas. FRONTIERS IN DENTAL MEDICINE 2021. [DOI: 10.3389/fdmed.2021.674056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The pathophysiology of SARS-CoV-2 infection is characterized by rapid virus replication and aggressive inflammatory responses that can lead to acute respiratory distress syndrome (ARDS) only a few days after the onset of symptoms. It is suspected that a dysfunctional immune response is the main cause of SARS-CoV-2 infection-induced lung destruction and mortality due to massive infiltration of hyperfunctional neutrophils in these organs. Similarly, neutrophils are recruited constantly to the oral cavity to combat microorganisms in the dental biofilm and hyperfunctional neutrophil phenotypes cause destruction of periodontal tissues when periodontitis develops. Both disease models arise because of elevated host defenses against invading organisms, while concurrently causing host damage/disease when the immune cells become hyperfunctional. This represents a clear nexus between periodontal and medical research. As researchers begin to understand the link between oral and systemic diseases and their potential synergistic impact on general health, we argue that translational research from studies in periodontology must be recognized as an important source of information that might lead to different therapeutic options which can be effective for the management of both oral and non-oral diseases. In this article we connect concepts from periodontal research on oral inflammation while exploring host modulation therapy used for periodontitis as a potential strategy for the prevention of ARDS a deadly outcome of COVID-19. We suggest that host modulation therapy, although developed initially for management of periodontitis, and which inhibits proteases, cytokines, and the oxidative stress that underlie ARDS, will provide an effective and safe treatment for COVID-19.
Collapse
|
43
|
Zidar A, Kristl J, Kocbek P, Zupančič Š. Treatment challenges and delivery systems in immunomodulation and probiotic therapies for periodontitis. Expert Opin Drug Deliv 2021; 18:1229-1244. [PMID: 33760648 DOI: 10.1080/17425247.2021.1908260] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Periodontitis is a widespread illness that arises due to disrupted interplay between the oral microbiota and the host immune response. In some cases, conventional therapies can provide temporary remission, although this is often followed by disease relapse. Recent studies of periodontitis pathology have promoted the development of new therapeutics to improve treatment options, together with local application using advanced drug delivery systems.Areas covered: This paper provides a critical review of the status of current treatment approaches to periodontitis, with a focus on promising immunomodulation and probiotic therapies. These are based on delivery of small molecules, peptides, proteins, DNA or RNA, and probiotics. The key findings on novel treatment strategies and formulation of advanced delivery systems, such as nanoparticles and nanofibers, are highlighted.Expert opinion: Multitarget therapy based on antimicrobial, immunomodulatory, and probiotic active ingredients incorporated into advanced delivery systems for application to the periodontal pocket can improve periodontitis treatment outcomes. Translation of such adjuvant therapy from laboratory to patient is expected in the future.
Collapse
Affiliation(s)
- Anže Zidar
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Julijana Kristl
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Petra Kocbek
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Špela Zupančič
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
44
|
Dommisch H, Stolte KN, Jager J, Vogel K, Müller R, Hedtrich S, Unbehauen M, Haag R, Danker K. Characterization of an ester-based core-multishell (CMS) nanocarrier for the topical application at the oral mucosa. Clin Oral Investig 2021; 25:5795-5805. [PMID: 33821321 PMCID: PMC8443517 DOI: 10.1007/s00784-021-03884-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 03/11/2021] [Indexed: 01/09/2023]
Abstract
OBJECTIVES Topical drug administration is commonly applied to control oral inflammation. However, it requires sufficient drug adherence and a high degree of bioavailability. Here, we tested the hypothesis whether an ester-based core-multishell (CMS) nanocarrier is a suitable nontoxic drug-delivery system that penetrates efficiently to oral mucosal tissues, and thereby, increase the bioavailability of topically applied drugs. MATERIAL AND METHODS To evaluate adhesion and penetration, the fluorescence-labeled CMS 10-E-15-350 nanocarrier was applied to ex vivo porcine masticatory and lining mucosa in a Franz cell diffusion assay and to an in vitro 3D model. In gingival epithelial cells, potential cytotoxicity and proliferative effects of the nanocarrier were determined by MTT and sulphorhodamine B assays, respectively. Transepithelial electrical resistance (TEER) was measured in presence and absence of CMS 10-E-15-350 using an Endohm-12 chamber and a volt-ohm-meter. Cellular nanocarrier uptake was analyzed by laser scanning microscopy. Inflammatory responses were determined by monitoring pro-inflammatory cytokines using real-time PCR and ELISA. RESULTS CMS nanocarrier adhered to mucosal tissues within 5 min in an in vitro model and in ex vivo porcine tissues. The CMS nanocarrier exhibited no cytotoxic effects and induced no inflammatory responses. Furthermore, the physical barrier expressed by the TEER remained unaffected by the nanocarrier. CONCLUSIONS CMS 10-E-15-350 adhered to the oral mucosa and adhesion increased over time which is a prerequisite for an efficient drug release. Since TEER is unaffected, CMS nanocarrier may enter the oral mucosa transcellularly. CLINICAL RELEVANCE Nanocarrier technology is a novel and innovative approach for efficient topical drug delivery at the oral mucosa.
Collapse
Affiliation(s)
- H Dommisch
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 14197, Berlin, Germany. .,Department of Periodontology, University of Washington, Seattle, WA, USA.
| | - K N Stolte
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 14197, Berlin, Germany
| | - J Jager
- Institute for Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - K Vogel
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 14197, Berlin, Germany
| | - R Müller
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 14197, Berlin, Germany
| | - S Hedtrich
- Pharmacology and Toxicology, Institute of Pharmacy, Freie Universität Berlin, 14195, Berlin, Germany.,Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, V6T1Z3, Canada
| | - M Unbehauen
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - R Haag
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - K Danker
- Institute for Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| |
Collapse
|
45
|
Galli M, Yao Y, Giannobile WV, Wang HL. Current and future trends in periodontal tissue engineering and bone regeneration. PLASTIC AND AESTHETIC RESEARCH 2021; 8. [PMID: 35765666 PMCID: PMC9236184 DOI: 10.20517/2347-9264.2020.176] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Periodontal tissue engineering involves a multi-disciplinary approach towards the regeneration of periodontal ligament, cementum and alveolar bone surrounding teeth, whereas bone regeneration specifically applies to ridge reconstruction in preparation for future implant placement, sinus floor augmentation and regeneration of peri-implant osseous defects. Successful periodontal regeneration is based on verifiable cementogenesis on the root surface, oblique insertion of periodontal ligament fibers and formation of new and vital supporting bone. Ultimately, regenerated periodontal and peri-implant support must be able to interface with surrounding host tissues in an integrated manner, withstand biomechanical forces resulting from mastication, and restore normal function and structure. Current regenerative approaches utilized in everyday clinical practice are mainly guided tissue/bone regeneration-based. Although these approaches have shown positive outcomes for small and medium-sized defects, predictability of clinical outcomes is heavily dependent on the defect morphology and clinical case selection. In many cases, it is still challenging to achieve predictable regenerative outcomes utilizing current approaches. Periodontal tissue engineering and bone regeneration (PTEBR) aims to improve the state of patient care by promoting reconstitution of damaged and lost tissues through the use of growth factors and signaling molecules, scaffolds, cells and gene therapy. The present narrative review discusses key advancements in PTEBR including current and future trends in preclinical and clinical research, as well as the potential for clinical translatability.
Collapse
Affiliation(s)
- Matthew Galli
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Yao Yao
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - William V Giannobile
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA.,Biointerfaces Institute, North Campus Research Complex, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA.,Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Hom-Lay Wang
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| |
Collapse
|
46
|
The Role of Epigenetic Functionalization of Implants and Biomaterials in Osseointegration and Bone Regeneration-A Review. Molecules 2020; 25:molecules25245879. [PMID: 33322654 PMCID: PMC7763898 DOI: 10.3390/molecules25245879] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
The contribution of epigenetic mechanisms as a potential treatment model has been observed in cancer and autoimmune/inflammatory diseases. This review aims to put forward the epigenetic mechanisms as a promising strategy in implant surface functionalization and modification of biomaterials, to promote better osseointegration and bone regeneration, and could be applicable for alveolar bone regeneration and osseointegration in the future. Materials and Methods: Electronic and manual searches of the literature in PubMed, MEDLINE, and EMBASE were conducted, using a specific search strategy limited to publications in the last 5 years to identify preclinical studies in order to address the following focused questions: (i) Which, if any, are the epigenetic mechanisms used to functionalize implant surfaces to achieve better osseointegration? (ii) Which, if any, are the epigenetic mechanisms used to functionalize biomaterials to achieve better tissue regeneration? Findings from several studies have emphasized the role of miRNAs in functionalizing implants surfaces and biomaterials to promote osseointegration and bone regeneration, respectively. However, there are scarce data on the role of DNA methylation and histone modifications for these specific applications, despite being commonly applied in cancer research. Studies over the past few years have demonstrated that biomaterials are immunomodulatory rather than inert materials. In this context, epigenetics can act as next generation of advanced treatment tools for future regenerative techniques. Yet, there is a need to evaluate the efficacy/cost effectiveness of these techniques in comparison to current standards of care.
Collapse
|
47
|
Elangovan S, Gajendrareddy P, Ravindran S, Salem AK. Emerging local delivery strategies to enhance bone regeneration. ACTA ACUST UNITED AC 2020; 15:062001. [PMID: 32647095 PMCID: PMC10148649 DOI: 10.1088/1748-605x/aba446] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In orthopedics and dentistry there is an increasing need for novel biomaterials and clinical strategies to achieve predictable bone regeneration. These novel molecular strategies have the potential to eliminate the limitations of currently available approaches. Specifically, they have the potential to reduce or eliminate the need to harvest autogenous bone, and the overall complexity of the clinical procedures. In this review, emerging tissue engineering strategies that have been, or are currently being, developed based on the current understanding of bone biology, development and wound healing will be discussed. In particular, protein/peptide based approaches, DNA/RNA therapeutics, cell therapy, and the use of exosomes will be briefly covered. The review ends with a summary of the current status of these approaches, their clinical translational potentials and their challenges.
Collapse
Affiliation(s)
- Satheesh Elangovan
- Department of Periodontics, The University of Iowa College of Dentistry, Iowa City, IA 52242, United States of America
| | | | | | | |
Collapse
|
48
|
D'Ângelo MQ, Queiroz-Junior CM, Maltos KLDM, Ferreira AJ, Pacheco CMDF, Soares RV. The blockade of kappa opioid receptors exacerbates alveolar bone resorption in rats. Arch Oral Biol 2020; 120:104923. [PMID: 33091661 DOI: 10.1016/j.archoralbio.2020.104923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Bone resorption associated to chronic diseases, such as arthritis and periodontitis, results from exacerbated immuno-inflammatory host response that leads to tissue breakdown. The significance of opioid pathways as endogenous modulators of inflammatory events has already been described. Thus, the aim of this work is to determine whether some of the main three opioid receptors are endogenously activated to prevent bone loss during experimentally-induced alveolar bone resorption. DESIGN This study used an experimental model of alveolar bone resorption induced by ligature in rats. A silk thread was placed around the 2nd maxillary molar of male Wistar rats. In the 3rd, 4th and 5th day after ligation the rats received a local injection of different concentrations of opioid antagonists Cyprodime, Naltrindole, or Nor-binaltorphimine, which specifically block mü, delta and kappa opioid receptors, respectively. In the 7th experimental day, rats were euthanized and their maxillae collected for evaluation of alveolar bone and fiber attachment loss, morphometric counting of osteoclasts and osteoblasts, as well as the levels of cytokines IL-1β, IFN-γ, and IL-6 by ELISA. RESULTS Selective antagonism of kappa opioid receptors, but not mü and delta, exacerbated alveolar bone resorption induced by ligature in rats. The increased bone loss associated with higher number of osteoclasts surrounding alveolar bone, although osteoblasts' counting remained unchanged. The concentrations of IL-1β and IL-6 in periodontal tissues were also significantly higher in the rats treated with the kappa antagonist. CONCLUSION Inhibiting kappa opioid receptors exacerbates alveolar bone resorption.
Collapse
Affiliation(s)
- Marcelo Queiroz D'Ângelo
- Graduate Program in Dentistry, Pontifícia Universidade Católica de Minas Gerais (PUCMINAS), Pontifícia Universidade Católica de Minas Gerais (PUCMINAS), Rua Dom José Gaspar 500, Coração Eucarístico, Belo Horizonte, MG, Brazil
| | - Celso Martins Queiroz-Junior
- Translational Biology Laboratory, Morphology Department, Institute of Biological Sciences, Universidade Federal de Minas Gerais (ICB-UFMG), Avenida Antônio Carlos 6627, Belo Horizonte, MG, Brazil
| | - Kátia Lucy de Melo Maltos
- School of Dentistry, Universidade Federal de Minas Gerais (FOUFMG), Avenida Antônio Carlos 6627, Belo Horizonte, MG, Brazil
| | - Anderson José Ferreira
- Translational Biology Laboratory, Morphology Department, Institute of Biological Sciences, Universidade Federal de Minas Gerais (ICB-UFMG), Avenida Antônio Carlos 6627, Belo Horizonte, MG, Brazil
| | | | - Rodrigo Villamarim Soares
- Graduate Program in Dentistry, Pontifícia Universidade Católica de Minas Gerais (PUCMINAS), Pontifícia Universidade Católica de Minas Gerais (PUCMINAS), Rua Dom José Gaspar 500, Coração Eucarístico, Belo Horizonte, MG, Brazil
| |
Collapse
|
49
|
Wang B, Booij-Vrieling HE, Bronkhorst EM, Shao J, Kouwer PHJ, Jansen JA, Walboomers XF, Yang F. Antimicrobial and anti-inflammatory thermo-reversible hydrogel for periodontal delivery. Acta Biomater 2020; 116:259-267. [PMID: 32937208 DOI: 10.1016/j.actbio.2020.09.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 01/06/2023]
Abstract
In periodontal treatment, topical adjunctive therapy with antimicrobials or anti-inflammatory agents is frequently applied. However, currently available drug carrier biomaterials often exhibit poor perfusion into small crevices, such as the deep and irregular periodontal pockets, due to relatively high viscosity. Moreover, high polymer concentrations of the polymer can potentially be cytotoxic upon confined local administration. This study aimed to formulate an antimicrobial and anti-inflammatory treatment option, by incorporating doxycycline (DOX) and/or lipoxin A4 (LXA4) into 0.5 wt% thermo-reversible polyisocyanopeptide (PIC). PIC can form hydrogels upon low polymer concentration, and we hypothesized that the thermo-reversible nature of the material would allow for application into the periodontal pocket. The formulations were characterized in vitro and finally tested in dogs with naturally occurring periodontitis, which were not euthanized afterward. Results showed that PIC/DOX/LXA4 hydrogel could be easily prepared and injected into periodontal pockets. The PIC hydrogel facilitated the release of DOX or LXA4 for around 4 days in vitro. When applied in dogs, the hydrogel exerted no local or systemic adverse effects. Gels loaded with LXA4 and/or DOX reduced the subgingival bacterial load and pro-inflammatory interleukin-8 level. In addition, PIC-DOX and PIC-DOX+LXA4 improved gingival clinical attachment by 0.6 mm compared with conventional periodontal treatment alone (i.e. mechanical debridement). In conclusion, the thermo-reversible PIC hydrogel is a safe and effective vehicle for periodontal drug delivery.
Collapse
Affiliation(s)
- Bing Wang
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Dentistry - Biomaterials, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; School of Stomatology, Shandong University, Jinan, Shandong, China
| | - Henriëtte E Booij-Vrieling
- Department of Clinical Sciences of Companion Animals, General Surgery, Faculty of Veterinary Medicine, Utrecht University, the Netherlands
| | - Ewald M Bronkhorst
- Radboud University Medical Center, Department of Dentistry, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - Jinlong Shao
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Dentistry - Biomaterials, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; School of Stomatology, Shandong University, Jinan, Shandong, China
| | - Paul H J Kouwer
- Institute for Molecules and Materials, Radboud University, Nijmegen, the Netherlands
| | - John A Jansen
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Dentistry - Biomaterials, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - X Frank Walboomers
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Dentistry - Biomaterials, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - Fang Yang
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Dentistry - Biomaterials, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands.
| |
Collapse
|
50
|
Hajishengallis G, Chavakis T, Lambris JD. Current understanding of periodontal disease pathogenesis and targets for host-modulation therapy. Periodontol 2000 2020; 84:14-34. [PMID: 32844416 DOI: 10.1111/prd.12331] [Citation(s) in RCA: 238] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
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.
Collapse
Affiliation(s)
- George Hajishengallis
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Faculty of Medicine, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|