1
|
Kumar N, Bhagwat P, Singh S, Pillai S. A review on the diversity of antimicrobial peptides and genome mining strategies for their prediction. Biochimie 2024; 227:99-115. [PMID: 38944107 DOI: 10.1016/j.biochi.2024.06.013] [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: 04/11/2024] [Revised: 06/08/2024] [Accepted: 06/27/2024] [Indexed: 07/01/2024]
Abstract
Antibiotic resistance has become one of the most serious threats to human health in recent years. In response to the increasing microbial resistance to the antibiotics currently available, it is imperative to develop new antibiotics or explore new approaches to combat antibiotic resistance. Antimicrobial peptides (AMPs) have shown considerable promise in this regard, as the microbes develop low or no resistance against them. The discovery and development of AMPs still confront numerous obstacles such as finding a target, developing assays, and identifying hits and leads, which are time-consuming processes, making it difficult to reach the market. However, with the advent of genome mining, new antibiotics could be discovered efficiently using tools such as BAGEL, antiSMASH, RODEO, etc., providing hope for better treatment of diseases in the future. Computational methods used in genome mining automatically detect and annotate biosynthetic gene clusters in genomic data, making it a useful tool in natural product discovery. This review aims to shed light on the history, diversity, and mechanisms of action of AMPs and the data on new AMPs identified by traditional as well as genome mining strategies. It further substantiates the various phases of clinical trials for some AMPs, as well as an overview of genome mining databases and tools built expressly for AMP discovery. In light of the recent advancements, it is evident that targeted genome mining stands as a beacon of hope, offering immense potential to expedite the discovery of novel antimicrobials.
Collapse
Affiliation(s)
- Naveen Kumar
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, P O Box 1334, Durban, 4000, South Africa.
| | - Prashant Bhagwat
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, P O Box 1334, Durban, 4000, South Africa.
| | - Suren Singh
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, P O Box 1334, Durban, 4000, South Africa.
| | - Santhosh Pillai
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, P O Box 1334, Durban, 4000, South Africa.
| |
Collapse
|
2
|
Ebersole JL, Kirakodu SS, Nguyen LM, Gonzalez OA. Transcriptomic features of programmed and inflammatory cell death in gingival tissues. Oral Dis 2024; 30:5274-5293. [PMID: 38623775 DOI: 10.1111/odi.14939] [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: 11/20/2023] [Revised: 03/04/2024] [Accepted: 03/09/2024] [Indexed: 04/17/2024]
Abstract
The local gingival tissue environment with homeostasis and tissue-destructive events of periodontitis demonstrates major changes in histological features and biology of the oral/sulcular epithelium, fibroblasts, vascular cells, inflammatory cell infiltration, and alveolar bone. OBJECTIVE This study used an experimental periodontitis model to detail the gingival transcriptome related to cell death processes of pyroptosis, necroptosis, ferroptosis, and cuproptosis. MATERIALS AND METHODS Healthy Macaca mulatta primates stratified by age, ≤3 years (young), 7-12 years (adolescent), 12-15 years (adult), and 17-23 years (aged), provided gingival tissue biopsies for microarray analysis focused on 257 genes representative of the four cell death processes and bacterial plaque samples for 16S rRNA gene analysis. RESULTS Age differences in the profiles of gene expression in healthy tissues were noted for cuproptosis, ferroptosis, necroptosis, and pyroptosis. Major differences were then observed with disease initiation, progression, and resolution also related to the age of the animals. Distinct bacterial families/consortia of species were significantly related to the gene expression differences for the cell death pathways. CONCLUSIONS These results emphasized age-associated differences in the gingival tissue molecular response to changes in the quality and quantity of bacteria accumulating with the disease process reflected in regulated cell death pathways that are both physiological and pathophysiological.
Collapse
Affiliation(s)
- Jeffrey L Ebersole
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, Nevada, USA
| | - Sreenatha S Kirakodu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, Kentucky, USA
| | - Linh M Nguyen
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, Nevada, USA
| | - Octavio A Gonzalez
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, Kentucky, USA
- Division of Periodontology, College of Dentistry, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
3
|
Kaur H, Gogoi B, Sharma I, Das DK, Azad MA, Pramanik DD, Pramanik A. Hydrogels as a Potential Biomaterial for Multimodal Therapeutic Applications. Mol Pharm 2024; 21:4827-4848. [PMID: 39290162 PMCID: PMC11462506 DOI: 10.1021/acs.molpharmaceut.4c00595] [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: 05/30/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Hydrogels, composed of hydrophilic polymer networks, have emerged as versatile materials in biomedical applications due to their high water content, biocompatibility, and tunable properties. They mimic natural tissue environments, enhancing cell viability and function. Hydrogels' tunable physical properties allow for tailored antibacterial biomaterial, wound dressings, cancer treatment, and tissue engineering scaffolds. Their ability to respond to physiological stimuli enables the controlled release of therapeutics, while their porous structure supports nutrient diffusion and waste removal, fostering tissue regeneration and repair. In wound healing, hydrogels provide a moist environment, promote cell migration, and deliver bioactive agents and antibiotics, enhancing the healing process. For cancer therapy, they offer localized drug delivery systems that target tumors, minimizing systemic toxicity and improving therapeutic efficacy. Ocular therapy benefits from hydrogels' capacity to form contact lenses and drug delivery systems that maintain prolonged contact with the eye surface, improving treatment outcomes for various eye diseases. In mucosal delivery, hydrogels facilitate the administration of therapeutics across mucosal barriers, ensuring sustained release and the improved bioavailability of drugs. Tissue regeneration sees hydrogels as scaffolds that mimic the extracellular matrix, supporting cell growth and differentiation for repairing damaged tissues. Similarly, in bone regeneration, hydrogels loaded with growth factors and stem cells promote osteogenesis and accelerate bone healing. This article highlights some of the recent advances in the use of hydrogels for various biomedical applications, driven by their ability to be engineered for specific therapeutic needs and their interactive properties with biological tissues.
Collapse
Affiliation(s)
- Harpreet Kaur
- Amity
Institute of Biotechnology, Amity University, Noida 201301, India
| | - Bishmita Gogoi
- Amity
Institute of Biotechnology, Amity University, Noida 201301, India
| | - Ira Sharma
- Amity
Institute of Biotechnology, Amity University, Noida 201301, India
| | - Deepak Kumar Das
- Department
of Chemistry and Nanoscience, GLA University, Mathura, Uttar Pradesh 281 406, India
| | - Mohd Ashif Azad
- Amity
Institute of Biotechnology, Amity University, Noida 201301, India
| | | | - Arindam Pramanik
- Amity
Institute of Biotechnology, Amity University, Noida 201301, India
- School
of Medicine, University of Leeds, Leeds LS97TF, United Kingdom
| |
Collapse
|
4
|
Atalay N, Balci N, Gürsoy M, Gürsoy UK. Systemic Factors Affecting Human Beta-Defensins in Oral Cavity. Pathogens 2024; 13:654. [PMID: 39204254 PMCID: PMC11357671 DOI: 10.3390/pathogens13080654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 09/03/2024] Open
Abstract
Human beta-defensins are host defense peptides with broad antimicrobial and inflammatory functions. In the oral cavity, these peptides are produced mainly by the keratinocytes of the epithelium; however, fibroblasts, monocytes, and macrophages also contribute to oral human beta-defensin expressions. The resident and immune cells of the oral cavity come into contact with various microbe-associated molecular patterns continuously and simultaneously. The overall antimicrobial cellular response is highly influenced by local and environmental factors. Recent studies have produced evidence showing that not only systemic chronic diseases but also systemic factors like hyperglycemia, pregnancy, the long-term use of certain vitamins, and aging can modulate oral cellular antimicrobial responses against microbial challenges. Therefore, the aim of this narrative review is to discuss the role of systemic factors on oral human beta-defensin expressions.
Collapse
Affiliation(s)
- Nur Atalay
- Department of Periodontology, Institute of Dentistry, University of Turku, Lemminkäisenkatu 2, 20520 Turku, Finland
| | - Nur Balci
- Department of Periodontology, Faculty of Dentistry, Medipol University, Goztepe Mahallesi, Ataturk Caddesi 40, Beykoz, 34815 Istanbul, Turkey
| | - Mervi Gürsoy
- Department of Periodontology, Institute of Dentistry, University of Turku, Lemminkäisenkatu 2, 20520 Turku, Finland
- Welfare Division, Oral Health Care, 20540 Turku, Finland
| | - Ulvi Kahraman Gürsoy
- Department of Periodontology, Institute of Dentistry, University of Turku, Lemminkäisenkatu 2, 20520 Turku, Finland
| |
Collapse
|
5
|
Li N, Wang J, Feng G, Liu Y, Shi Y, Wang Y, Chen L. Advances in biomaterials for oral-maxillofacial bone regeneration: spotlight on periodontal and alveolar bone strategies. Regen Biomater 2024; 11:rbae078. [PMID: 39055303 PMCID: PMC11272181 DOI: 10.1093/rb/rbae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/05/2024] [Accepted: 06/16/2024] [Indexed: 07/27/2024] Open
Abstract
The intricate nature of oral-maxillofacial structure and function, coupled with the dynamic oral bacterial environment, presents formidable obstacles in addressing the repair and regeneration of oral-maxillofacial bone defects. Numerous characteristics should be noticed in oral-maxillofacial bone repair, such as irregular morphology of bone defects, homeostasis between hosts and microorganisms in the oral cavity and complex periodontal structures that facilitate epithelial ingrowth. Therefore, oral-maxillofacial bone repair necessitates restoration materials that adhere to stringent and specific demands. This review starts with exploring these particular requirements by introducing the particular characteristics of oral-maxillofacial bones and then summarizes the classifications of current bone repair materials in respect of composition and structure. Additionally, we discuss the modifications in current bone repair materials including improving mechanical properties, optimizing surface topography and pore structure and adding bioactive components such as elements, compounds, cells and their derivatives. Ultimately, we organize a range of potential optimization strategies and future perspectives for enhancing oral-maxillofacial bone repair materials, including physical environment manipulation, oral microbial homeostasis modulation, osteo-immune regulation, smart stimuli-responsive strategies and multifaceted approach for poly-pathic treatment, in the hope of providing some insights for researchers in this field. In summary, this review analyzes the complex demands of oral-maxillofacial bone repair, especially for periodontal and alveolar bone, concludes multifaceted strategies for corresponding biomaterials and aims to inspire future research in the pursuit of more effective treatment outcomes.
Collapse
Affiliation(s)
- Nayun Li
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinyu Wang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guangxia Feng
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuqing Liu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yunsong Shi
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yifan Wang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
6
|
Mouchrek MMM, Franco MM, da Silva LA, Martins KAC, da Conceição SIO, de Azevedo Dos Santos APS, Rodrigues VP, Ribeiro CCC, Benatti BB. Cytokine levels in the gingival crevicular fluid and their association with periodontal status of down syndrome patients: a cross-sectional study. Clin Oral Investig 2024; 28:391. [PMID: 38907162 DOI: 10.1007/s00784-024-05789-x] [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: 12/12/2023] [Accepted: 06/13/2024] [Indexed: 06/23/2024]
Abstract
OBJECTIVE To evaluate cytokine levels of interleukin (IL)-1β, IL-4, IL-6, IL-17a, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ in the gingival crevicular fluid (GCF) of periodontal sites in individuals with Down syndrome (DS) and analyze their relationship with clinical periodontal parameters. MATERIALS AND METHODS A cross-sectional study was conducted with 49 DS patients and 32 individuals without DS (non-DS group). Periodontal probing depth (PPD), clinical attachment level (CAL), bleeding on probing (BoP), and visible plaque index (VPI) were evaluated. The periodontal sites were classified as shallow, moderate, and deep. GCF was collected in all shallow sites and, when present, in moderate and deep sites for the analysis of cytokine levels. The cytokines, IL-1β, IL-4, IL-6, IL-17a, TNF-α, and IFN-γ, were quantified using the Luminex® automatic analyzer system. RESULTS The DS group presented greater severity of periodontitis compared to the non-DS group (P = 0.005). The DS group showed a significant direct correlation of IL-1β and an inverse correlation of IFN-γ and IL-14 with all periodontal variables. In the analysis stratified by periodontal pocket depth, we observed a higher level of IFN-γ, IL-17a, IL-1β, and IL-6 in the shallow sites, and IL-17a, IL-1β, and IL-6 in deep pockets of DS group individuals. Multivariate models showed that higher levels of IL-1β, IL-4, IL-6, and IL-17a were associated with Down syndrome even after adjusting for periodontal status, sex, and age. CONCLUSION The findings suggest that people with DS have greater periodontal impairment and higher levels of cytokines in GCF, even in sites having clinical periodontal parameters similar to those of individuals without DS. These data reiterate the concept of an altered and less effective immune response in the population with DS in the face of a periodontal microbial challenge. CLINICAL RELEVANCE Elevated periodontal inflammation burden can be observed with higher cytokine levels in the gingival crevicular fluid of people with Down syndrome, especially IL-1, IL-4, IL-6, and IL-17, regardless of the stage of periodontitis.
Collapse
Affiliation(s)
- Monique Maria Melo Mouchrek
- Dentistry Graduate Program, School of Dentistry, Federal University of Maranhão, Avenida dos Portugueses, 1966, São Luís, 65080-805, Brazil
| | - Mayra Moura Franco
- Dentistry Graduate Program, School of Dentistry, Federal University of Maranhão, Avenida dos Portugueses, 1966, São Luís, 65080-805, Brazil
| | - Leslie Alves da Silva
- Dentistry Graduate Program, School of Dentistry, Federal University of Maranhão, Avenida dos Portugueses, 1966, São Luís, 65080-805, Brazil
| | - Kerry Alynne Campos Martins
- Dentistry Graduate Program, School of Dentistry, Federal University of Maranhão, Avenida dos Portugueses, 1966, São Luís, 65080-805, Brazil
| | | | | | - Vandílson Pinheiro Rodrigues
- Dentistry Graduate Program, School of Dentistry, Federal University of Maranhão, Avenida dos Portugueses, 1966, São Luís, 65080-805, Brazil
| | - Cecilia Cláudia Costa Ribeiro
- Dentistry Graduate Program, School of Dentistry, Federal University of Maranhão, Avenida dos Portugueses, 1966, São Luís, 65080-805, Brazil
| | - Bruno Braga Benatti
- Dentistry Graduate Program, School of Dentistry, Federal University of Maranhão, Avenida dos Portugueses, 1966, São Luís, 65080-805, Brazil.
| |
Collapse
|
7
|
Chen M, Hu Z, Shi J, Xie Z. Human β-defensins and their synthetic analogs: Natural defenders and prospective new drugs of oral health. Life Sci 2024; 346:122591. [PMID: 38548013 DOI: 10.1016/j.lfs.2024.122591] [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: 01/12/2024] [Revised: 03/08/2024] [Accepted: 03/24/2024] [Indexed: 04/14/2024]
Abstract
As a family of cationic host defense peptides, human β-defensins (HBDs) are ubiquitous in the oral cavity and are mainly synthesized primarily by epithelial cells, serving as the primary barrier and aiming to prevent microbial invasion, inflammation, and disease while maintaining physiological homeostasis. In recent decades, there has been great interest in their biological functions, structure-activity relationships, mechanisms of action, and therapeutic potential in oral diseases. Meanwhile, researchers are dedicated to improving the properties of HBDs for clinical application. In this review, we first describe the classification, structural characteristics, functions, and mechanisms of HBDs. Next, we cover the role of HBDs and their synthetic analogs in oral diseases, including dental caries and pulp infections, periodontitis, peri-implantitis, fungal/viral infections and oral mucosal diseases, and oral squamous cell carcinoma. Finally, we discuss the limitations and challenges of clinical translation of HBDs and their synthetic analogs, including, but not limited to, stability, bioavailability, antimicrobial activity, resistance, and toxicity. Above all, this review summarizes the biological functions, mechanisms of action, and therapeutic potential of both natural HBDs and their synthetic analogs in oral diseases, as well as the challenges associated with clinical translation, thus providing substantial insights into the laboratory development and clinical application of HBDs in oral diseases.
Collapse
Affiliation(s)
- Mumian Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Zihe Hu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Jue Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Zhijian Xie
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
8
|
Gürsel M, Bayat T, Dündar N, Yabacı Tak A, Karaduman B. Investigating the diagnostic potential of IL-1β, IL-10, and IL-36γ in gingival crevicular fluid in patients with different periodontal conditions. Biomarkers 2024; 29:118-126. [PMID: 38344835 DOI: 10.1080/1354750x.2024.2318256] [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: 09/05/2023] [Accepted: 02/06/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND The study aimed to analyze cytokine levels, including interleukin (IL)-1β, IL-10, and IL-36γ, to investigate the link between pro- and anti-inflammatory responses in periodontal conditions and assess their potential as diagnostic biomarkers for distinguishing between different types of periodontal conditions. METHODS 80 systemically healthy non-smokers (25 periodontally healthy, 25 with gingivitis, 30 with periodontitis) were included. Clinical periodontal parameters were recorded, and gingival crevicular fluid (GCF) samples were obtained. Receiver operating characteristic (ROC) curve analysis was applied to determine the diagnostic value of cytokines. RESULTS IL-36γ had the highest sensitivity for diagnosing periodontitis, although its specificity for identifying those without periodontitis was relatively low. The combination of IL-1β and IL-36γ was the most effective in differentiating periodontitis from periodontal health. IL-10 was found to be an acceptable discriminator for distinguishing gingivitis from healthy conditions. However, its sensitivity and specificity for identifying gingivitis were lower. The combination of the three cytokines showed the highest ability to distinguish between periodontitis and gingivitis. CONCLUSION The levels of IL-1β, IL-10, and IL-36γ in GCF may provide insights into periodontal health and disease status. Further studies are needed to validate these results and explore the potential of these cytokines in periodontal disease management.
Collapse
Affiliation(s)
- Mihtikar Gürsel
- Department of Periodontology,, Bezmialem Vakif University, Istanbul, Turkey
| | - Tuba Bayat
- Department of Periodontology, Sakarya University, Sakarya, Turkey
| | | | - Ayşegül Yabacı Tak
- Department of Biostatistics and Medical Informatics, Bezmialem Vakif University, Istanbul, Turkey
| | - Burcu Karaduman
- Department of Periodontology, Biruni University, Istanbul, Turkey
| |
Collapse
|
9
|
Ma Y, Qian Y, Chen Y, Ruan X, Peng X, Sun Y, Zhang J, Luo J, Zhou S, Deng C. Resveratrol modulates the inflammatory response in hPDLSCs via the NRF2/HO-1 and NF-κB pathways and promotes osteogenic differentiation. J Periodontal Res 2024; 59:162-173. [PMID: 37905727 DOI: 10.1111/jre.13200] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/29/2023] [Accepted: 10/16/2023] [Indexed: 11/02/2023]
Abstract
OBJECTIVE The purpose of this study was to investigate resveratrol's specific role as an anti-inflammatory and osteogenic differentiation of hPDLSCs in periodontitis and to reveal the mechanisms involved. BACKGROUND Numerous studies have shown that inhibiting the inflammatory response of periodontal tissues and promoting the regeneration of alveolar bone are crucial treatments for periodontitis. Resveratrol has been found to have certain anti-inflammatory property. However, the anti-inflammatory mechanism and osteogenic effect of resveratrol in periodontitis are poorly understood. MATERIALS AND METHODS We constructed an in vitro periodontitis model by LPS stimulation of hPDLSCs and performed WB, RT-qPCR, and immunofluorescence to analyze inflammatory factors and related pathways. In addition, we explored the osteogenic ability of resveratrol in in vitro models. RESULTS In vitro, resveratrol ameliorated the inflammatory response associated with activation of the NF-κB pathway through activation of the NRF2/HO-1 pathway, characterized by inhibition of p65/p50 nuclear translocation and the proinflammatory cytokines interleukin-1β levels. Resveratrol also has a positive effect on osteogenic differentiation. CONCLUSIONS Observations suggest that resveratrol modulates the inflammatory response in hPDLSCs via the NRF2/HO-1 and NF-κB pathways and promotes osteogenic differentiation.
Collapse
Affiliation(s)
- Yifan Ma
- School of Stomatology, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for dental materials and application, Wannan Medical College, Wuhu, China
| | - Yi Qian
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
| | - Yuteng Chen
- School of Stomatology, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for dental materials and application, Wannan Medical College, Wuhu, China
| | - Xiaoxu Ruan
- School of Stomatology, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for dental materials and application, Wannan Medical College, Wuhu, China
| | - Xiaoya Peng
- School of Stomatology, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for dental materials and application, Wannan Medical College, Wuhu, China
| | - Yi Sun
- School of Stomatology, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for dental materials and application, Wannan Medical College, Wuhu, China
| | - Jue Zhang
- School of Stomatology, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for dental materials and application, Wannan Medical College, Wuhu, China
| | - Jingjing Luo
- School of Stomatology, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for dental materials and application, Wannan Medical College, Wuhu, China
| | - Songlin Zhou
- School of Stomatology, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for dental materials and application, Wannan Medical College, Wuhu, China
| | - Chao Deng
- School of Stomatology, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for dental materials and application, Wannan Medical College, Wuhu, China
| |
Collapse
|
10
|
Liu Y, Xiang C, Que Z, Li C, Wang W, Yin L, Chu C, Zhou Y. Neutrophil heterogeneity and aging: implications for COVID-19 and wound healing. Front Immunol 2023; 14:1201651. [PMID: 38090596 PMCID: PMC10715311 DOI: 10.3389/fimmu.2023.1201651] [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: 04/06/2023] [Accepted: 08/02/2023] [Indexed: 12/18/2023] Open
Abstract
Neutrophils play a critical role in the immune response to infection and tissue injury. However, recent studies have shown that neutrophils are a heterogeneous population with distinct subtypes that differ in their functional properties. Moreover, aging can alter neutrophil function and exacerbate immune dysregulation. In this review, we discuss the concept of neutrophil heterogeneity and how it may be affected by aging. We then examine the implications of neutrophil heterogeneity and aging for COVID-19 pathogenesis and wound healing. Specifically, we summarize the evidence for neutrophil involvement in COVID-19 and the potential mechanisms underlying neutrophil recruitment and activation in this disease. We also review the literature on the role of neutrophils in the wound healing process and how aging and neutrophil heterogeneity may impact wound healing outcomes. Finally, we discuss the potential for neutrophil-targeted therapies to improve clinical outcomes in COVID-19 and wound healing.
Collapse
Affiliation(s)
| | | | | | | | - Wen Wang
- Department of Hematology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Medical Cosmetic Center, Chengdu Second People's Hospital; Minhang Hospital, Fudan University, Shanghai, China
| | - Lijuan Yin
- Department of Hematology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Medical Cosmetic Center, Chengdu Second People's Hospital; Minhang Hospital, Fudan University, Shanghai, China
| | - Chenyu Chu
- Department of Hematology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Medical Cosmetic Center, Chengdu Second People's Hospital; Minhang Hospital, Fudan University, Shanghai, China
| | - Yin Zhou
- Department of Hematology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Medical Cosmetic Center, Chengdu Second People's Hospital; Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Panahipour L, Botta S, Abbasabadi AO, Afradi Z, Gruber R. Enamel Matrix Derivative Suppresses Chemokine Expression in Oral Epithelial Cells. Int J Mol Sci 2023; 24:13991. [PMID: 37762294 PMCID: PMC10530986 DOI: 10.3390/ijms241813991] [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: 08/23/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Epithelial cells in periodontitis patients increasingly express chemokines, suggesting their active involvement in the inflammatory process. Enamel matrix derivative (EMD) is an extract of porcine fetal tooth germs clinically applied to support the regrowth of periodontal tissues. Periodontal regeneration might benefit from the potential anti-inflammatory activity of EMD for epithelial cells. Our aim was, therefore, to set up a bioassay where chemokine expression is initiated in the HSC2 oral squamous carcinoma cell line and then test EMD for its capacity to lower the inflammatory response. To establish the bioassay, HSC2 cells being exposed to TNFα and LPS from E. coli (Escherichia coli) or P. gingivalis (Porphyromonas gingivalis) were subjected to RNAseq. Here, TNFα but not LPS caused a robust increase of chemokines, including CXCL1, CXCL2, CXCL8, CCL5, and CCL20 in HSC2 cells. Polymerase chain reaction confirmed the increased expression of the respective chemokines in cells exposed to TNFα and IL-1β. Under these conditions, EMD reduced the expression of all chemokines at the transcriptional level and CXCL8 by immunoassay. The TGF-β receptor type I kinase-inhibitor SB431542 reversed the anti-inflammatory activity. Moreover, EMD-activated TGF-β-canonical signaling was visualized by phosphorylation of smad3 and nuclear translocation of smad2/3 in HSC2 cells and blocked by SB431542. This observation was confirmed with primary oral epithelial cells where EMD significantly lowered the SB431542-dependent expression of CXCL8. In summary, our findings suggest that TGF-β signaling mediates the effects of EMD to lower the forced expression of chemokines in oral epithelial cells.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (S.B.); (A.O.A.); (Z.A.)
| | - Sara Botta
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (S.B.); (A.O.A.); (Z.A.)
| | - Azarakhsh Oladzad Abbasabadi
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (S.B.); (A.O.A.); (Z.A.)
| | - Zohreh Afradi
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (S.B.); (A.O.A.); (Z.A.)
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (S.B.); (A.O.A.); (Z.A.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
12
|
Zheng H, Zhou Y, Zheng Y, Liu G. Advances in hydrogels for the treatment of periodontitis. J Mater Chem B 2023; 11:7321-7333. [PMID: 37431231 DOI: 10.1039/d3tb00835e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
Periodontitis is the second most prevalent oral disease and can cause serious harm to human health. Hydrogels are excellent biomaterials that can be used for periodontitis as drug delivery platforms to achieve inflammation control through high drug delivery efficiency and sustained drug release and as tissue scaffolds to achieve tissue remodelling through encapsulated cell wrapping and effective mass transfer. In this review, we summarize the latest advances in the treatment of periodontitis with hydrogels. The pathogenic mechanisms of periodontitis are introduced first, followed by the recent progress of hydrogels in controlling inflammation and tissue reconstruction, in which the specific performance of hydrogels is discussed in detail. Finally, the challenges and limitations of hydrogels for clinical applications in periodontitis are discussed and possible directions for development are proposed. This review aims to provide a reference for the design and fabrication of hydrogels for the treatment of periodontitis.
Collapse
Affiliation(s)
- Huiyu Zheng
- The State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu 610065, China.
| | - Yuan Zhou
- The State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu 610065, China.
| | - Yu Zheng
- The State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu 610065, China.
| | - Guiting Liu
- The State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu 610065, China.
| |
Collapse
|
13
|
Sordi MB, Panahipour L, Gruber R. Oral squamous carcinoma cell lysates provoke exacerbated inflammatory response in gingival fibroblasts. Clin Oral Investig 2023; 27:4785-4794. [PMID: 37391526 PMCID: PMC10415472 DOI: 10.1007/s00784-023-05107-x] [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: 09/20/2022] [Accepted: 06/05/2023] [Indexed: 07/02/2023]
Abstract
OBJECTIVES To study whether damaged epithelial cells and gingival fibroblast could affect the expression of inflammatory cytokines in healthy cells. MATERIALS AND METHODS Cell suspensions were submitted to different treatments to obtain the lysates: no treatment (supernatant control), sonication, and freeze/thawing. All treatments were centrifuged, and the supernatants of the lysates were used for experimentation. Cell viability assays, RT-qPCR of IL1, IL6 and IL8, IL6 immunoassay, and immunofluorescence of NF-kB p65 were applied to verify the inflammatory crosstalk of damaged cells over healthy plated cells. Furthermore, titanium discs and collagen membranes were treated with lysates and checked for IL8 expression by RT-qPCR. RESULTS Lysates obtained upon sonication or freeze/thawing of oral squamous carcinoma cell lines provoked a robust increase in the expression of IL1, IL6, and IL8 by gingival fibroblasts, which was confirmed by IL6 immunoassays. Lysates obtained from the gingival fibroblasts failed to increase the expression of inflammatory cytokines in oral squamous carcinoma cells. Additionally, oral squamous carcinoma cell lysates caused the activation of the NF-kB signalling cascade in gingival fibroblasts as indicated by the phosphorylation and nuclear translocation of p65. Finally, oral squamous carcinoma cell lysates adhered to the titanium and collagen membrane surfaces and increased IL8 expression by gingival fibroblasts growing in these materials. CONCLUSIONS Injured oral epithelial cells can release factors that incite gingival fibroblasts to become pro-inflammatory. CLINICAL RELEVANCE Injuries affecting the oral mucosa generate epithelial fragments that may reach the underlying connective tissue and provoke inflammation. These injuries are routinely caused by mastication, sonication for teeth cleaning, teeth preparation, prostheses maladaptation, and implant drilling.
Collapse
Affiliation(s)
- Mariane Beatriz Sordi
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
- Department of Dentistry, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria.
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland.
| |
Collapse
|
14
|
Xu X, Li X, Qiu S, Zhou Y, Li L, Chen X, Zheng K, Xu Y. Concentration Selection of Biofriendly Enzyme-Modified Gelatin Hydrogels for Periodontal Bone Regeneration. ACS Biomater Sci Eng 2023; 9:4341-4355. [PMID: 37294274 DOI: 10.1021/acsbiomaterials.3c00166] [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: 06/10/2023]
Abstract
Periodontitis is challenging to cure radically due to its complex periodontal structure and particular microenvironment of dysbiosis and inflammation. However, with the assistance of various materials, cell osteogenic differentiation could be improved, and the ability of hard tissue regeneration could be enhanced. This study aimed to explore the appropriate concentration ratio of biofriendly transglutaminase-modified gelatin hydrogels for promoting periodontal alveolar bone regeneration. Through a series of characterization and cell experiments, we found that all the hydrogels possessed multi-space network structures and demonstrated their biocompatibility. In vivo and in vitro osteogenic differentiation experiments also confirmed that the group 40-5 (transglutaminase-gelatin concentration ratio) possessed a favorable osteogenic potential. In summary, we conclude that such hydrogel with a 40-5 concentration is most conducive to promoting periodontal bone reconstruction, which might be a new route to deal with the dilemma of clinical periodontal treatment.
Collapse
Affiliation(s)
- Xuanwen Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Xinyu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Shuang Qiu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Yi Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Lu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Xu Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Kai Zheng
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Yan Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| |
Collapse
|
15
|
He Y, Wang Y, Jia X, Li Y, Yang Y, Pan L, Zhao R, Han Y, Wang F, Guan X, Hou T. Glycolytic reprogramming controls periodontitis-associated macrophage pyroptosis via AMPK/SIRT1/NF-κB signaling pathway. Int Immunopharmacol 2023; 119:110192. [PMID: 37068341 DOI: 10.1016/j.intimp.2023.110192] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 04/19/2023]
Abstract
Glycolysis has been demonstrated as a crucial metabolic process in bacteria infected diseases via modulating the activity of pyroptosis. Macrophages are the most abundant immune cells that infiltrated in the infected periodontal tissues, which significantly influence the outcome of periodontitis (PD). However, the effect of glycolysis in regulating macrophage pyroptosis during PD development remains unknown. This study aimed to explore the role of glycolysis in PD-associated macrophage pyroptosis and periodontal degeneration. Clinical specimens were used to determine the emergence of macrophage pyroptosis and glycolysis in periodontal tissues by immunohistochemical analysis and western blot. For an in-depth understanding of the regulatory effect of glycolysis in the progression of macrophage pyroptosis associated periodontitis, both in vivo PD model and in vitro PD model were treated with 2-DG (2-Deoxy-d-glucose), a glycolysis inhibitor. The data showed that the blockade of glycolysis could significantly suppress the lipopolysaccharide (LPS) induced macrophage pyroptosis, resulting in an attenuation of the inflammatory response and bone resorption in periodontal lesions. Furthermore, we revealed that the regulatory effect of glycolysis on macrophage pyroptosis can be mediated via AMPK/SIRT1/NF-κB signaling pathway. Our study unveiled that suppressed glycolysis restrains the activity of PD-associated macrophage pyroptosis, osteoclastogenesis, and subsequent periodontal tissue destruction. These findings extend our knowledge of glycolysis in regulating PD-associated macrophage pyroptosis and provide a potential novel target for PD therapy.
Collapse
Affiliation(s)
- Yani He
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuting Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiangbin Jia
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yingxue Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yao Yang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lifei Pan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Rui Zhao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yue Han
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Feng Wang
- Experimental Center of Stomatology, School of Stomatology, Xi'an Medical College, Xi'an, Shaanxi, China
| | - Xiaoyue Guan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Tiezhou Hou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
16
|
Alghamdi B, Jeon HH, Ni J, Qiu D, Liu A, Hong JJ, Ali M, Wang A, Troka M, Graves DT. Osteoimmunology in Periodontitis and Orthodontic Tooth Movement. Curr Osteoporos Rep 2023; 21:128-146. [PMID: 36862360 PMCID: PMC10696608 DOI: 10.1007/s11914-023-00774-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/04/2023] [Indexed: 03/03/2023]
Abstract
PURPOSE OF REVIEW To review the role of the immune cells and their interaction with cells found in gingiva, periodontal ligament, and bone that leads to net bone loss in periodontitis or bone remodeling in orthodontic tooth movement. RECENT FINDINGS Periodontal disease is one of the most common oral diseases causing inflammation in the soft and hard tissues of the periodontium and is initiated by bacteria that induce a host response. Although the innate and adaptive immune response function cooperatively to prevent bacterial dissemination, they also play a major role in gingival inflammation and destruction of the connective tissue, periodontal ligament, and alveolar bone characteristic of periodontitis. The inflammatory response is triggered by bacteria or their products that bind to pattern recognition receptors that induce transcription factor activity to stimulate cytokine and chemokine expression. Epithelial, fibroblast/stromal, and resident leukocytes play a key role in initiating the host response and contribute to periodontal disease. Single-cell RNA-seq (scRNA-seq) experiments have added new insight into the roles of various cell types in the response to bacterial challenge. This response is modified by systemic conditions such as diabetes and smoking. In contrast to periodontitis, orthodontic tooth movement (OTM) is a sterile inflammatory response induced by mechanical force. Orthodontic force application stimulates acute inflammatory responses in the periodontal ligament and alveolar bone stimulated by cytokines and chemokines that produce bone resorption on the compression side. On the tension side, orthodontic forces induce the production of osteogenic factors, stimulating new bone formation. A number of different cell types, cytokines, and signaling/pathways are involved in this complex process. Inflammatory and mechanical force-induced bone remodeling involves bone resorption and bone formation. The interaction of leukocytes with host stromal cells and osteoblastic cells plays a key role in both initiating the inflammatory events as well as inducing a cellular cascade that results in remodeling in orthodontic tooth movement or in tissue destruction in periodontitis.
Collapse
Affiliation(s)
- Bushra Alghamdi
- Department of Endodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, PA, 19104, Philadelphia, USA
- Department of Restorative Dental Sciences, College of Dentistry, Taibah University, Medina, 42353, Kingdom of Saudi Arabia
| | - Hyeran Helen Jeon
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jia Ni
- Department of Periodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Dongxu Qiu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Alyssia Liu
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, PA, 19104, Philadelphia, USA
| | - Julie J Hong
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, PA, 19104, Philadelphia, USA
| | - Mamoon Ali
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, PA, 19104, Philadelphia, USA
| | - Albert Wang
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, PA, 19104, Philadelphia, USA
| | - Michael Troka
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, PA, 19104, Philadelphia, USA
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, PA, 19104, Philadelphia, USA.
| |
Collapse
|
17
|
Guo Q, Zheng J, Lin H, Han Z, Wang Z, Ren J, Zhai J, Zhao H, Du R, Li C. Conditioned media of deer antler stem cells accelerate regeneration of alveolar bone defects in rats. Cell Prolif 2023; 56:e13454. [PMID: 36929672 DOI: 10.1111/cpr.13454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/01/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
The destruction of periodontal alveolar bone (AB) caused by periodontitis is regarded as one of the major reasons for tooth loss. The inhibition of bone resorption and regeneration of lost AB are the desirable outcomes in clinical practice but remain in challenge. The use of mesenchymal stem cells (MSCs) is one current approach for achieving true restoration of AB defects (ABD). Antler stem cells (AnSC) are capable of renewing a huge mammalian bony appendage, the deer antler, suggesting an unparalleled potential for bone regeneration. Herein, we investigated the effectiveness of deer AnSCs conditioned medium (CM, AnSC-CM) for repair of surgically-created ABD using a rat model and sought to define the underlying mechanisms. The results showed that AnSC-CM effectively induced regeneration of AB tissue; the outcome was significantly better than human bone marrow mesenchymal stem cell conditioned medium (hBMSC-CM). AnSC-CM treatment upregulated osteogenic factors and downregulated osteoclastic differentiation factors; stimulated proliferation, migration and differentiation of resident MSCs toward osteogenic lineage cells; modulated macrophage polarization toward the M2 phenotype and suppressed osteoclastogenesis. That AnSC-CM resulted in better outcomes than hBMSC-CM in treating ABD was attributed to the cell compatibility as both AnSCs and AB tissue are neural crest-derived. In conclusion, the effects of AnSC-CM on AB tissue regeneration were achieved through both promotion of osteogenesis and inhibition of osteoclastogenesis. We believe that AnSC-CM is a candidate for effective treatment of ABD in dental clinical practice but will require investment in further development.
Collapse
Affiliation(s)
- Qianqian Guo
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| | - Junjun Zheng
- Institute of Special Economic Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Hongbing Lin
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Zhongming Han
- Jilin Agricultural University, College of Chinese Medicinal Materials, Changchun, Jilin, 130118, China
| | - Zhen Wang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| | - Jing Ren
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| | - Jingjie Zhai
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Haiping Zhao
- Qingdao Agricultural University, College of Animal Science and Technology, Qingdao, Shandong, China
| | - Rui Du
- Jilin Agricultural University, College of Chinese Medicinal Materials, Changchun, Jilin, 130118, China
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| |
Collapse
|
18
|
Wang H, Wang D, Huangfu H, Chen S, Qin Q, Ren S, Zhang Y, Fu L, Zhou Y. Highly efficient photothermal branched Au-Ag nanoparticles containing procyanidins for synergistic antibacterial and anti-inflammatory immunotherapy. Biomater Sci 2023; 11:1335-1349. [PMID: 36594408 DOI: 10.1039/d2bm01212j] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Periodontitis is an inflammatory disease caused by bacterial infection. Excessive immune response and high levels of reactive oxygen species (ROS) further lead to the irreversible destruction of surrounding tissues. Developing new antimicrobial materials that regulate the immune system to resist inflammation can effectively treat periodontal inflammation. A nanoplatform integrating Ag+, photothermal therapy (PTT), and procyanidins (PC) for precision antibacterial and synergistic immunotherapy for periodontitis was proposed. This work loaded PC into AuAg nanoparticles, and the resulting nanocomposite was named AuAg-PC. PTT can effectively remove pathogenic bacteria, but high temperatures can cause tissue damage. Ag+ contributes to the preparation of a nanoparticle branched structure that improves the photothermal efficiency and helps PTT achieve an excellent antibacterial effect and avoid periodontal tissue damage. PC regulates host immunity by eliminating intracellular ROS, inhibiting inflammatory factors, and regulating macrophage polarisation in periodontal disease sites. It enhances the host's resistance to bacterial inflammation. AuAg-PC exerted an excellent anti-inflammatory effect and promoted tissue repair in animal periodontal inflammation models. Hence, AuAg-PC significantly combats periodontal pathogens and shows great application potential in the photothermal-assisted immunotherapy of periodontitis. This design provided a new controllable and efficient treatment platform for controlling persistent inflammation infection and regulating immunity.
Collapse
Affiliation(s)
- Hanchi Wang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China. .,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Dongyang Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Huimin Huangfu
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China. .,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Siyu Chen
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China. .,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Qiuyue Qin
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China. .,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Sicong Ren
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China. .,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Yidi Zhang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China. .,State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Li Fu
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China. .,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Yanmin Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China. .,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| |
Collapse
|
19
|
Cai R, Wang L, Zhang W, Liu B, Wu Y, Pang J, Ma C. The role of extracellular vesicles in periodontitis: pathogenesis, diagnosis, and therapy. Front Immunol 2023; 14:1151322. [PMID: 37114060 PMCID: PMC10126335 DOI: 10.3389/fimmu.2023.1151322] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Periodontitis is a prevalent disease and one of the leading causes of tooth loss. Biofilms are initiating factor of periodontitis, which can destroy periodontal tissue by producing virulence factors. The overactivated host immune response is the primary cause of periodontitis. The clinical examination of periodontal tissues and the patient's medical history are the mainstays of periodontitis diagnosis. However, there is a lack of molecular biomarkers that can be used to identify and predict periodontitis activity precisely. Non-surgical and surgical treatments are currently available for periodontitis, although both have drawbacks. In clinical practice, achieving the ideal therapeutic effect remains a challenge. Studies have revealed that bacteria produce extracellular vesicles (EVs) to export virulence proteins to host cells. Meanwhile, periodontal tissue cells and immune cells produce EVs that have pro- or anti-inflammatory effects. Accordingly, EVs play a critical role in the pathogenesis of periodontitis. Recent studies have also presented that the content and composition of EVs in saliva and gingival crevicular fluid (GCF) can serve as possible periodontitis diagnostic indicators. In addition, studies have indicated that stem cell EVs may encourage periodontal regeneration. In this article, we mainly review the role of EVs in the pathogenesis of periodontitis and discuss their diagnostic and therapeutic potential.
Collapse
Affiliation(s)
- Rong Cai
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
| | - Lu Wang
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wei Zhang
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
| | - Bing Liu
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
| | - Yiqi Wu
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jianliang Pang
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
- *Correspondence: Chufan Ma, ; Jianliang Pang,
| | - Chufan Ma
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
- *Correspondence: Chufan Ma, ; Jianliang Pang,
| |
Collapse
|
20
|
Jeong YJ, Jung JI, Kim Y, Kang CH, Imm JY. Effects of Lactobacillus reuteri MG5346 on Receptor Activator of Nuclear Factor-Kappa B Ligand (RANKL)-Induced Osteoclastogenesis and Ligature-Induced Experimental Periodontitis Rats. Food Sci Anim Resour 2023; 43:157-169. [PMID: 36789196 PMCID: PMC9890358 DOI: 10.5851/kosfa.2022.e68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022] Open
Abstract
Effects of culture supernatants of Lactobacillus reuteri MG5346 (CS-MG5346) on receptor activator of nuclear factor-kappa B ligand (RANKL)-induced osteoclastogenesis were examined. CS-MG5346 treatment up to 400 μg/mL significantly reduced tartrate-resistant acid-phosphatase (TRAP) activity, the phenotype biomarker of osteoclast, without affecting cell viability. CS-MG5346 inhibited the expression of osteoclast specific transcriptional factors (c-fos and nuclear factor-activated T cells c1) and their target genes (TRAP, cathepsin, and matrix metallo-proteinase-9) in a dose-dependent manner (p<0.05). The administration of L. reuteri MG5346 (2×108 CFU/day) for 8 wks significantly improved furcation involvement, but no difference was observed in alveolar bone loss in ligature-induced experimental periodontitis rats. The elevated RANKL/ osteoprotegerin ratio, the biomarker of periodontitis, was significantly lowered in the gingival tissue by administration of L. reuteri MG5346 (p<0.05). L. reuteri MG5346 showed excellent stability in simulated stomach and intestinal fluids and did not have antibiotic resistance. Based on the results, L. reuteri MG5346 has the potential to be a promising probiotic strain for oral health.
Collapse
Affiliation(s)
- Yu-Jin Jeong
- Department of Foods and Nutrition, Kookmin
University, Seoul 02707, Korea
| | - Jae-In Jung
- Department of Foods and Nutrition, Kookmin
University, Seoul 02707, Korea
| | | | | | - Jee-Young Imm
- Department of Foods and Nutrition, Kookmin
University, Seoul 02707, Korea,Corresponding author: Jee-Young
Imm, Department of Foods and Nutrition, Kookmin University, Seoul 02707, Korea,
Tel: +82-2-910-4772, Fax: +82-2-910-5249, E-mail:
| |
Collapse
|
21
|
Fluorescent probes in stomatology. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
22
|
Wang L, Chen X, Zhang L, Niu B, Li L, Sun Y, Yuan X. CAR cell design strategies in solid tumors. Int Immunopharmacol 2022; 113:109345. [DOI: 10.1016/j.intimp.2022.109345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/03/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
23
|
Huang X, Zhu W, Zhang X, Fu Y. Modified gingivoplasty for hereditary gingival fibromatosis: two case reports. BMC Oral Health 2022; 22:523. [DOI: 10.1186/s12903-022-02411-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 08/26/2022] [Indexed: 11/26/2022] Open
Abstract
Abstract
Background
Hereditary gingival fibromatosis (HGF) is characterized by sub-epithelial fibromatosis of keratinized gingiva resulting in a fibrotic enlargement of keratinized gingiva. The treatment choice is gingivectomy, which can be performed with an internal or external bevel incision conventionally. However, both techniques can hardly resume the natural status of gingiva, and have a certain recurrence rate, especially in the cases which have limited width of attached gingiva.
Case description
Two cases of HGF with the chief complaint of difficulty in mastication, pronunciation, and poor esthetics were presented. After the initial periodontal therapy, a novel gingivoplasty modified with a crevicular incision was applied. A full thickness flap above the mucogingival junction and a split flap below the junction were raised. Then, fibrotic connective tissue was completely eliminated and keratinized gingival epithelium was preserved. The fibrotic alveolar bone was shaped by handpiece and bur. Finally, the flap was apically repositioned and sutured. Twelve months after surgery, the gingiva recovered with normal color, contour and consistency.
Conclusions
Compared to traditional gingivectomy, modified gingivoplasty which focuses on eliminating pathological fibrotic connective tissue can completely resume the natural appearance of gingiva and demonstrate no tendency of recurrence.
Collapse
|
24
|
Ramírez Thomé S, Ávila Curiel B, Hernández Huerta MT, Solórzano Mata C. β-defensinas como posibles indicadores de la actividad inflamatoria en la enfermedad periodontal. INVESTIGACIÓN CLÍNICA 2022. [DOI: 10.54817/ic.v63n4a08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Periodontal disease (gingivitis and periodontitis) is an inflam-matory process caused by the activity of pathogenic bacteria and their products on the gingival sulcus, with the consequent activation of the immune response. Saliva and crevicular fluid contain a wide variety of enzymes and antimicrobial factors that are in contact with the supragingival and subgingival region, in-cluding β-defensins (hBDs). hHBDs are non-glycosylated, cysteine-rich cationic peptides produced by epithelial cells with antimicrobial and immunoregulatory effects, thus contributing to maintaining homeostasis in periodontal tissues. The changes in the microbiota and the immune response from a healthy peri-odontium to gingivitis and, finally, to periodontitis are complex. Their sever-ity depends on a dynamic balance between bacteria associated with plaque, genetic and environmental factors. Recent advances have made it possible to understand the implication of hBDs in the detection, diagnosis, and therapy of periodontal disease and the relationship between periodontitis and other inflammatory conditions. This review aims to describe the effect of hBDs on the immune response and its use as a possible marker of the inflammatory activity of the periodontal disease.
Collapse
Affiliation(s)
- Saira Ramírez Thomé
- Facultad de Odontología. Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, México
| | | | | | - Carlos Solórzano Mata
- Facultad de Odontología. Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, México
| |
Collapse
|
25
|
Xu X, Zhou Y, Zheng K, Li X, Li L, Xu Y. 3D Polycaprolactone/Gelatin-Oriented Electrospun Scaffolds Promote Periodontal Regeneration. ACS APPLIED MATERIALS & INTERFACES 2022; 14:46145-46160. [PMID: 36197319 DOI: 10.1021/acsami.2c03705] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Periodontitis is a worldwide chronic inflammatory disease, where surgical treatment still shows an uncertain prognosis. To break through the dilemma of periodontal treatment, we fabricated a three-dimensional (3D) multilayered scaffold by stacking and fixing electrospun polycaprolactone/gelatin (PCL/Gel) fibrous membranes. The biomaterial displayed good hydrophilic and mechanical properties. Besides, we found human periodontal ligament stem cell (hPDLSC) adhesion and proliferation on it. The following scanning electron microscopy (SEM) and cytoskeleton staining results proved the guiding function of fibers to hPDLSCs. Then, we further analyzed periodontal regeneration-related proteins and mRNA expression between groups. In vivo results in a rat acute periodontal defect model confirmed that the topographic cues of materials could directly guide cellular orientation and might provide the prerequisite for further differentiation. In the aligned scaffold group, besides new bone regeneration, we also observed that angular concentrated fiber regeneration in the root surface of the defect is similar to the normal periodontal tissue. To sum up, we have constructed electrospun membrane-based 3D biological scaffolds, which provided a new treatment strategy for patients undergoing periodontal surgery.
Collapse
Affiliation(s)
- Xuanwen Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing210029, China
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing210029, China
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing210029, China
| | - Yi Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing210029, China
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing210029, China
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing210029, China
| | - Kai Zheng
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing210029, China
| | - Xinyu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing210029, China
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing210029, China
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing210029, China
| | - Lu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing210029, China
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing210029, China
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing210029, China
| | - Yan Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing210029, China
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing210029, China
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing210029, China
| |
Collapse
|
26
|
Lin C, Yang Y, Wang Y, Jing H, Bai X, Hong Z, Zhang C, Gao H, Zhang L. Periodontal ligament fibroblasts-derived exosomes induced by PGE 2 inhibit human periodontal ligament stem cells osteogenic differentiation via activating miR-34c-5p/SATB2/ERK. Exp Cell Res 2022; 419:113318. [PMID: 35981635 DOI: 10.1016/j.yexcr.2022.113318] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 11/04/2022]
Abstract
Several studies have confirmed that exosomes containing microRNAs (miRNAs) from the aseptic inflammatory microenvironment play an important role in bone remodeling. But the mechanism that induces changes in the osteogenic ability of periodontal ligament stem cells (PDLSCs) is still unclear. In the present study, the osteogenic function of periodontal ligament fibroblasts-derived exosomes induced by PGE2 on PDLSCs was detected by real-time PCR, alizarin red assay and alkaline phosphatase staining. High-throughput miRNAs sequencing was used to reveal that miR-34c-5p in exosomes-PGE2 was upregulated compared it in exosomes-normal. Real-time PCR and western blotting assay verified that overexpression of miR-34c-5p inhibited osteogenic differentiation, and reduced phosphorylation of ERK1/2. In addition, dual-luciferase reporter assay revealed that miR-34c-5p targeted special AT-rich sequence-binding protein 2 (SATB2). It was shown that exosomal miR-34c-5p inhibited osteogenic differentiation of PDLSCs via SATB2/ERK pathway.
Collapse
Affiliation(s)
- Chen Lin
- School of Clinical Stomatology, Tianjin Medical University, Tianjin, 300070, China; Department of Orthodontics, Tianjin Stomatological Hospital, Tianjin, 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin, 300041, China
| | - Yingying Yang
- School of Clinical Stomatology, Tianjin Medical University, Tianjin, 300070, China; Department of Orthodontics, Tianjin Stomatological Hospital, Tianjin, 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin, 300041, China
| | - Yingxue Wang
- School of Clinical Stomatology, Tianjin Medical University, Tianjin, 300070, China; Department of Orthodontics, Tianjin Stomatological Hospital, Tianjin, 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin, 300041, China
| | - Heng Jing
- Department of Oral and Maxillofacial Surgery, Tianjin Stomatological Hospital, Tianjin, 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin, 300041, China
| | - Xinyi Bai
- Department of Orthodontics, Tianjin Stomatological Hospital, Tianjin, 300041, China
| | - Zheng Hong
- Department of Orthodontics, Tianjin Stomatological Hospital, Tianjin, 300041, China
| | - Chunxiang Zhang
- Department of Orthodontics, Tianjin Stomatological Hospital, Tianjin, 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin, 300041, China.
| | - Hui Gao
- Department of Orthodontics, Tianjin Stomatological Hospital, Tianjin, 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin, 300041, China.
| | - Linkun Zhang
- Department of Orthodontics, Tianjin Stomatological Hospital, Tianjin, 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin, 300041, China.
| |
Collapse
|
27
|
Gonzalez OA, Kirakodu S, Nguyen LM, Orraca L, Novak MJ, Gonzalez-Martinez J, Ebersole JL. Comparative Analysis of Gene Expression Patterns for Oral Epithelial Cell Functions in Periodontitis. FRONTIERS IN ORAL HEALTH 2022; 3:863231. [PMID: 35677025 PMCID: PMC9169451 DOI: 10.3389/froh.2022.863231] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
The structure and function of epithelial cells are critical for the construction and maintenance of intact epithelial surfaces throughout the body. Beyond the mechanical barrier functions, epithelial cells have been identified as active participants in providing warning signals to the host immune and inflammatory cells and in communicating various detailed information on the noxious challenge to help drive specificity in the characteristics of the host response related to health or pathologic inflammation. Rhesus monkeys were used in these studies to evaluate the gingival transcriptome for naturally occurring disease samples (GeneChip® Rhesus Macaque Genome Array) or for ligature-induced disease (GeneChip® Rhesus Gene 1.0 ST Array) to explore up to 452 annotated genes related to epithelial cell structure and functions. Animals were distributed by age into four groups: ≤ 3 years (young), 3–7 years (adolescent), 12–16 years (adult), and 18–23 years (aged). For naturally occurring disease, adult and aged periodontitis animals were used, which comprised 34 animals (14 females and 20 males). Groups of nine animals in similar age groups were included in a ligature-induced periodontitis experiment. A buccal gingival sample from either healthy or periodontitis-affected tissues were collected, and microarray analysis performed. The overall results of this investigation suggested a substantial alteration in epithelial cell functions that occurs rapidly with disease initiation. Many of these changes were prolonged throughout disease progression and generally reflect a disruption of normal cellular functions that would presage the resulting tissue destruction and clinical disease measures. Finally, clinical resolution may not signify biological resolution and represent a continued risk for disease that may require considerations for additional biologically specific interventions to best manage further disease.
Collapse
Affiliation(s)
- Octavio A. Gonzalez
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
- Division of Periodontology, College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - Sreenatha Kirakodu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - Linh M. Nguyen
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, NV, United States
| | - Luis Orraca
- School of Dentistry, University of Puerto Rico, San Juan, Puerto Rico
| | - Michael J. Novak
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - Janis Gonzalez-Martinez
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, NV, United States
| | - Jeffrey L. Ebersole
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, NV, United States
- *Correspondence: Jeffrey L. Ebersole
| |
Collapse
|
28
|
Maquera-Huacho PM, Spolidorio DP, Manthey JA, Grenier D. Eriodictyol Suppresses Porphyromonas gingivalis-Induced Reactive Oxygen Species Production by Gingival Keratinocytes and the Inflammatory Response of Macrophages. FRONTIERS IN ORAL HEALTH 2022; 3:847914. [PMID: 35295880 PMCID: PMC8918503 DOI: 10.3389/froh.2022.847914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Porphyromonas gingivalis is a key pathogen of periodontitis, an inflammatory disease that affects the tooth-supporting tissues. The aim of the present study was to investigate the effects of the flavanone eriodictyol on P. gingivalis-induced reactive oxygen species (ROS) production by gingival keratinocytes and the inflammatory response of macrophages. Porphyromonas gingivalis and H2O2 acted synergistically to induce ROS production by keratinocytes. The presence of eriodictyol significantly attenuated ROS production in a dose-dependent manner. We used a macrophage model to show that eriodictyol decreases the secretion of IL-1β, IL-6, IL-8, and TNF-α induced by P. gingivalis. Evidence has been brought that this anti-inflammatory property of eriodictyol may be related to its ability to prevent the activation of the NF-κB signaling pathway by P. gingivalis. This periodontal pathogen was also found to be a potent inducer of matrix metalloproteinase (MMP) production by macrophages, including MMP-2, MMP-8, and MMP-9. Eriodictyol dose-dependently inhibited the production of all three MMPs. Lastly, eriodictyol inhibited the catalytic activity of both MMP-9 and P. gingivalis collagenase. In conclusion, eriodictyol may be a potential therapeutic agent for preventing and/or treating periodontal disease due to its antioxidant, anti-inflammatory, and anti-proteinase properties.
Collapse
Affiliation(s)
- Patricia Milagros Maquera-Huacho
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, QC, Canada
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (Unesp), Araraquara, Brazil
| | - Denise Palomari Spolidorio
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (Unesp), Araraquara, Brazil
| | - John A. Manthey
- U.S. Horticultural Research Laboratory, Agricultural Research Service, U.S. Department of Agriculture (USDA), Fort Pierce, FL, United States
| | - Daniel Grenier
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, QC, Canada
- *Correspondence: Daniel Grenier
| |
Collapse
|
29
|
Frandsen Lau E, Peterson DE, Leite FRM, Nascimento GG, Robledo‐Sierra J, Porat Ben Amy D, Kerr R, Lopez R, Baelum V, Lodi G, Varoni EM. Embracing multi‐causation of periodontitis: Why aren’t we there yet? Oral Dis 2021; 28:1015-1021. [DOI: 10.1111/odi.14107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/20/2022]
Affiliation(s)
- Ellen Frandsen Lau
- Section for Periodontology Department of Dentistry and Oral Health Faculty of Health Aarhus University Aarhus C Denmark
| | - Douglas E. Peterson
- Section of Oral Medicine Department of Oral Health and Diagnostic Sciences School of Dental Medicine UConn Health Farmington Connecticut USA
| | - Fabio R. M. Leite
- Section for Periodontology Department of Dentistry and Oral Health Faculty of Health Aarhus University Aarhus C Denmark
| | - Gustavo G. Nascimento
- Section for Periodontology Department of Dentistry and Oral Health Faculty of Health Aarhus University Aarhus C Denmark
| | | | - Dalit Porat Ben Amy
- Oral Medicine Unit Department of Oral & Maxillofacial Surgery The Baruch Padeh Medical Center Poriya Israel
| | - Ross Kerr
- Department of Oral and Maxillofacial Pathology, Radiology and Medicine New York University College of Medicine New York City New York USA
| | - Rodrigo Lopez
- Section for Periodontology Department of Dentistry and Oral Health Faculty of Health Aarhus University Aarhus C Denmark
| | - Vibeke Baelum
- Department of Dentistry and Oral Health Faculty of Health Aarhus University Aarhus C Denmark
| | - Giovanni Lodi
- Department of Biomedical Surgical and Dental Sciences University of Milan Milano Italy
| | - Elena M. Varoni
- Department of Biomedical Surgical and Dental Sciences University of Milan Milano Italy
| |
Collapse
|
30
|
Sun X, Gao J, Meng X, Lu X, Zhang L, Chen R. Polarized Macrophages in Periodontitis: Characteristics, Function, and Molecular Signaling. Front Immunol 2021; 12:763334. [PMID: 34950140 PMCID: PMC8688840 DOI: 10.3389/fimmu.2021.763334] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/04/2021] [Indexed: 12/23/2022] Open
Abstract
Periodontitis (PD) is a common chronic infectious disease. The local inflammatory response in the host may cause the destruction of supporting periodontal tissue. Macrophages play a variety of roles in PD, including regulatory and phagocytosis. Moreover, under the induction of different factors, macrophages polarize and form different functional phenotypes. Among them, M1-type macrophages with proinflammatory functions and M2-type macrophages with anti-inflammatory functions are the most representative, and both of them can regulate the tendency of the immune system to exert proinflammatory or anti-inflammatory functions. M1 and M2 macrophages are involved in the destructive and reparative stages of PD. Due to the complex microenvironment of PD, the dynamic development of PD, and various local mediators, increasing attention has been given to the study of macrophage polarization in PD. This review summarizes the role of macrophage polarization in the development of PD and its research progress.
Collapse
Affiliation(s)
- Xiaoyu Sun
- *Correspondence: Lei Zhang, ; Xiaoyu Sun,
| | | | | | | | - Lei Zhang
- Key Laboratory of Oral Diseases Research of Anhui Province, Department of Periodontology, Stomatologic Hospital & College, Anhui Medical University, Hefei, China
| | | |
Collapse
|
31
|
Mansour AM, Yahia S, Elsayed HRH, El-Attar SAE, Grawish ME, El-Hawary YM, El-Sherbiny IM. Efficacy of biocompatible trilayers nanofibrous scaffold with/without allogeneic adipose-derived stem cells on class II furcation defects of dogs' model. Clin Oral Investig 2021; 26:2537-2553. [PMID: 34661742 DOI: 10.1007/s00784-021-04222-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/06/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE This study aimed to evaluate the regenerative capacity of a newly-developed polycaprolactone (PCL)-based nanofibrous composite scaffold either alone or in combination with adipose-derived mesenchymal stem cells (ADSCs) as a treatment modality for class II furcation defects. MATERIALS AND METHODS After ADSCs isolation and scaffold characterization, the mandibular premolars of adult male mongrel dogs were selected and randomly assigned into three equal groups. In group I, class II furcation defects were surgically induced to the inter-radicular bone. While class II furcation defects of group II were induced as in group I. In addition, the defects were filled with the prefabricated scaffold. Moreover, class II furcation defects of group III were induced as in group II and instead the defects were filled with the prefabricated scaffold seeded with ADSCs. The dogs were sacrificed at 30 days or at 60 days. Periodontal wound healing/regeneration was evaluated by radiological examination using cone beam computed tomography and histologically using ordinary, histochemical, and immunohistochemical staining. RESULTS In the two examination periods, group II defects compared to group I, and group III compared to the other groups showed a decrease in defect dimensions radiographically. Histologically, histochemically, and immunohistochemically, they significantly demonstrated better periodontal wound healing/regeneration, predominant collagen type I of newly formed bone and periodontal ligament with a significant increase in the immunoreactivity of vascular endothelial growth factor and osteopontin. CONCLUSIONS The newly fabricated nanofibrous scaffold has enhanced periodontal wound healing/regeneration of class II furcation defects with further enhancement achieved when ADSCs seeded onto the scaffold before implantation. CLINICAL RELEVANCE The implementation of our newly-developed PCL-based nanofibrous composite scaffolds in class II furcation defect either alone or in conjunction with ADSCs can be considered as a suitable treatment modality to allow periodontal tissues regeneration.
Collapse
Affiliation(s)
- Alaa M Mansour
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, 35511, Egypt
| | - Sarah Yahia
- Nanomedicine Labs, Center of Materials Sciences (CMS), Zewail City of Science and Technology, 6th of October, Giza, 12578, Egypt
| | | | - Saied A E El-Attar
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, 35511, Egypt
| | - Mohammed E Grawish
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, 35511, Egypt
| | - Youssry M El-Hawary
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, 35511, Egypt
| | - Ibrahim M El-Sherbiny
- Nanomedicine Labs, Center of Materials Sciences (CMS), Zewail City of Science and Technology, 6th of October, Giza, 12578, Egypt.
| |
Collapse
|
32
|
Abstract
The central theme of this volume of Periodontology 2000 is that the microbial dental plaque biofilm, specifically the subgingival dental plaque biofilm, mimics a human tissue in both structure and function. As a basis for this assertion we use the definition of a tissue as an aggregate of similar cells and cell products forming a defined structure with a specific function, in a multicellular organism. Accordingly, we propose that the dental plaque biofilm represents an acquired human tissue largely of bacterial origin that maintains the health of gingival tissue. Furthermore, we acknowledge that disease can be defined as a deviation from the normal structure or an interruption to the function of any body part, organ, or system, and that is manifested by a characteristic set of symptoms and signs whose etiology, pathology, and prognosis may be known or unknown. Therefore, in this volume we present the concept that periodontitis is a disruption of the normal function of the healthy subgingival plaque biofilm with concomitant disruption to its functional properties in relation to innate defense surveillance and tissue maintenance, leading to excessive, deregulated inflammation and tissue destruction.
Collapse
Affiliation(s)
- Richard P Darveau
- Department of Periodontics, University of Washington, Seattle, Washington, USA
| | - Michael A Curtis
- Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| |
Collapse
|