DT News - India - How open architecture CAD/CAM can benefit your practice

Search Dental Tribune

How open architecture CAD/CAM can benefit your practice

With exocad’s ChairsideCAD, clinicians have the freedom to choose the best hardware and software for same-day dentistry regardless of the product or system they prefer. The technology has been selected as a Cellerant Best of Class Technology Award recipient in 2019 and 2020. (Photo: exocad)

Tue. 13 April 2021

save

ChairsideCAD, available from exocad, is the choice of software for leading manufacturers of dental CAD/CAM systems, according to the company, and it has been selected as a Cellerant Best of Class Technology Award recipient in 2019 and 2020.

“We are entering a new era in dentistry — one that will change how we diagnose, treat and manage our patients and practices,” said Dr. Lou Shuman, CEO of Cellerant and founder of the Best of Class Technology Awards. “This was a breakthrough year in product and services technologies. The panel spent hundreds of hours in close discussion reviewing and analyzing the corporate landscape. Pay close attention to our winners as they are truly leading the way to provide you what is best in today’s contemporary practice.”

With exocad’s ChairsideCAD, clinicians have the freedom to choose the best hardware and software for same-day dentistry regardless of the product or system they prefer. This is a groundbreaking evolution, according to the company, since historically the industry has been limited to specific workflows and specific hardware/software working together and adhering to a more closed architecture.

Derived from exocad DentalCAD, a signature software solution within the dental laboratory marketplace for more than a decade, ChairsideCAD includes dentalshare, a powerful collaboration tool for clinicians and labs, according to the company.

“We are honored to receive this prestigious industry award from our clinical audience. With exocad ChairsideCAD, clinicians can access labs, design and production centers with freedom of choice,” said Larry Bodony, president of exocad America. “With this open and flexible workflow, clinicians can maximize their return on investment as well as valuable chair time.”

“Our goal is to help the doctor make the best decisions for their office, which, in the end, benefits the patients that we all serve,” said John Flucke, DDS, Best of Class panel member. “I’m honored to be able to help my peers with the decision-making process and helping them wade through the plethora of high-tech products that can change offices and lives for the better.”

Bodony said, “Having received the Best of Class Award from the Cellerant Company and recognition from their exclusive panel of experts is truly an honor.”

(Source: exocad)

Tags:
To post a reply please login or register

Periodontitis can disrupt gut microbiota by inducing dysbiosis

Recent studies have shown that periodontitis may disrupt the homeostasis of gut microbiota by inducing dysbiosis (Image: Canva)

Fri. 29 March 2024

save

Periodontitis is a chronic oral inflammatory condition, commenced by the accumulation of pathogenic dental plaque biofilm above and below the gingival margin. Microbial dysbiosis may result in a chronic non-resolving and destructive inflammatory response which is characterized by the destruction of tooth-supporting tissues, such as the alveolar bone and periodontal ligament, and further leading to tooth loss. Recent studies suggest that periodontitis can disrupt the homeostasis of the healthy gut microbiota.

Potential pathways of gut translocation of oral pathobionts

The oral bacteria from the oral cavity can be translocated to the gut mucosa through the following two potential routes:

  1. Hematogenous spread
    • Oral bacteria can spread systemically through a hematogenous route.
    • Mechanical injuries in the oral cavity can result in the dissemination of oral bacteria into the systemic circulation.
    • Hematogenously inoculated Fusobacteria strains are more thriving in colonization in the gut tumor than gavaged strains, implying the circulatory system plays a major route in oral bacteria dissemination.
    • Oral bacteria can seize and remain inside dendritic cells and macrophages, indicating the hijacking of host immune cells to serve as Trojan horses for the spread of bacteria from the oral to the gut mucosa.
  2. Enteral dissemination
    • An individual can swallow around 600 times a day and synthesize ~1.5 L of saliva containing 1.5x10^12 oral bacteria.
    • More than half of the oral bacterial species have been detected in the gut, indicating oral–gut translocation of oral bacteria even in healthy people.
    • Generally, oral bacteria poorly colonize in the healthy gastrointestinal tract (GIT) because of the multiple barriers conferred by the healthy gut.
    • Gastric acidity is the first barrier against the translocation of oral bacteria to the gut mucosa.
    • Oral bacteria translocation can also be observed in patients who have gastric achlorhydria, gastroesophageal reflux disease, and gastritis after gastric surgery.
    • Various types of oral pathogens, like P. gingivalis, can survive in the acidic environment of the stomach and pass through the gut barrier.
    • Thus, the prevention of the enteral transmission of oral pathogens is the primary defense mechanism.
    • Secondly, maintaining the gut's harmonious microbial structure is crucial for avoiding ectopic colonization by ingested oral bacteria, since the gut resident microbiota confers resistance to colonization.
    • Factors responsible for gut microbiota dysbiosis include gut inflammation, artificial sweeteners, diets, and antibiotic treatment.

Microbial routes of direct gut colonization by oral pathogens

Multiple oral pathogens that colonize the gut may induce abnormal immune responses in GIT, thus leading to intestinal inflammation. Following oral bacteria are majorly responsible for gut inflammation:

  1. Fusobacterium spp.
    • The role of Fusobacterium spp. is still debatable in gut inflammation.
    • F. varium and F. nucleatum can permeate the intestinal epithelium and induce the synthesis of proinflammatory cytokines.
    • F. nucleatum can invade intestinal epithelial cells and promote the expression of proinflammatory cytokines.
    • Additionally, F. nucleatum can disrupt the integrity of the epithelial barrier.
  2. Porphyromonas Gingivalis
    • P. gingivalis is the main periodontopathic bacteria involved in the pathogenesis of periodontitis.
    • Studies showed that the orogastric administration of P. gingivalis to mice can disrupt the integrity of the gut epithelium.
    • Continuous administration of P. gingivalis to mice leads to endotoxemia, reduced gene expression of tight junction proteins, and increased proinflammatory cytokines in the gut.
  3. Staphylococcus aureus
    • S. aureus is observed to adhere to intestinal epithelial cells.
    • Oral administration of S. aureus strain RN8098 into antibiotic–penetrated mice produces epithelial damage in the small intestine.
  4. Klebsiella spp. and Enterobacter spp.
    • Klebsiella spp. and Enterobacter spp. colonization have been isolated from the saliva of patients with Crohn’s disease, resulting in potent Th1 cell differentiation in the gut of gnotobic animals.
    • Oral inflammation fosters blooms of Enterobacteriaceae, including Klebsiella spp. and Enterobacter spp., in genetically susceptible mice, leading to exacerbation of intestinal inflammation.

Conclusion

Over the last decade, the research area of intestinal inflammation and oral microorganisms has widely expanded by studies that have mainly focused on the effect of direct colonization of oral pathobionts in the gut. Moreover, the usage of murine models showed the novel aspects of the complicated intermucosal connection between the gut and the oral cavity.

References:

  1. Kistler JO, Booth V, Bradshaw DJ, Wade WG. Bacterial community development in experimental gingivitis. PLOS ONE 8(8), e71227 (2013).
  2. Bao J, Li L, Zhang Y, Wang M, Chen F, Ge S, Chen B, Yan F. Periodontitis may induce gut microbiota dysbiosis via salivary microbiota. International Journal of Oral Science 2022; 14:32.
  3. Kitamoto S, Kamada N. Periodontal connection with intestinal inflammation: Microbiological and immunological mechanisms. Periodontology 2000. 2022; 89: 142-153.
  4. Nakajima M, Arimatsu K, Kato T, et al. Oral administration of P gingivalis induces dysbiosis of gut microbiota and impaired barrier function leading to dissemination of enterobacteria to the liver. PLoS One. 2015;10(7):e0134234.
  5. Arimatsu K, Yamada H, Miyazawa H, et al. Oral pathobiont induces systemic inflammation and metabolic changes associated with alteration of gut microbiota. Sci Rep. 2014; 4:4828.
  6. Atarashi K, Suda W, Luo C, et al. Ectopic colonization of oral bacteria in the intestine drives TH1 cell induction and inflammation. Science. 2017;358(6361):359-365.
  7. Kitamoto S, Nagao-Kitamoto H, Jiao Y, et al. The intermucosal connection between the mouth and gut in commensal pathobiont-driven colitis. Cell. 2020;182(2):447-62 e14.
Topics:
Tags:
To post a reply please login or register
advertisement
advertisement