About all

Inflammation of the membrane. Inflammation of Membranes: From Stomatitis to Neurodegenerative Diseases

How does inflammation affect various membranes in the body. What are the causes and symptoms of stomatitis. Can membrane homeostasis be linked to neurodegenerative diseases. What treatments are available for oral inflammation.

Содержание

Understanding Membrane Inflammation: A Comprehensive Overview

Membrane inflammation is a widespread physiological response that can affect various parts of the body, from the oral cavity to the brain and spinal cord. This complex process plays a crucial role in both protecting the body and, in some cases, contributing to the development of serious conditions.

Types of Membrane Inflammation

  • Stomatitis: Inflammation of the oral mucosa
  • Meningitis: Inflammation of the membranes surrounding the brain and spinal cord
  • Neurodegenerative diseases: Potential link to membrane homeostasis disruption

Understanding these various forms of inflammation and their underlying causes is essential for effective prevention and treatment strategies.

Stomatitis: Causes, Symptoms, and Risk Factors

Stomatitis, the inflammation of the oral mucosa, is a common condition that can significantly impact an individual’s quality of life. Its occurrence is often linked to a variety of factors, both internal and external.

Primary Causes of Stomatitis

  • Viral infections
  • Bacterial infections
  • Fungal infections
  • Allergic reactions
  • Irritants and mechanical trauma

Are certain individuals more susceptible to developing stomatitis? Indeed, several factors can increase the risk of oral inflammation:

  1. Poor oral hygiene
  2. Weakened immune system
  3. Nutritional deficiencies, especially vitamins B and C
  4. Certain systemic diseases, such as celiac disease or inflammatory bowel disease
  5. Blood disorders, including anemia and leukemia

Recognizing these risk factors can help in implementing preventive measures and early intervention strategies.

Identifying Stomatitis: Key Symptoms

How can one recognize the signs of stomatitis? The condition typically manifests with several distinct symptoms:

  • Intense redness of the oral mucosa
  • Swelling of the mucous membrane
  • Coated tongue
  • Slightly enlarged lips
  • Inflamed and bleeding gums
  • Increased salivation
  • Bad breath
  • Enlarged and tender regional lymph nodes
  • Slight fever
  • Reduced appetite
  • Pain during eating or speaking

The presence of these symptoms should prompt individuals to seek medical attention for proper diagnosis and treatment.

Treating Stomatitis: From Antibiotics to Natural Remedies

Effective management of stomatitis often requires a multifaceted approach, combining targeted treatments with supportive care measures.

Antibiotic Therapy

When bacteria are identified as the primary cause of oral inflammation, antibiotic treatment becomes necessary. Healthcare providers typically prescribe specific antibiotics based on the causative organism and the severity of the infection.

Oral Antiseptics: A First Line of Defense

Oral antiseptics play a crucial role in managing stomatitis symptoms and promoting healing. These products, available over-the-counter, contain various active ingredients designed to combat inflammation and reduce discomfort.

One notable ingredient found in certain oral antiseptics is lysozyme, which is unique to Lysobact速 products. These products offer a range of benefits:

  • Action against inflammatory agents (bacteria, viruses, and fungi)
  • Symptom relief
  • Alleviation of the inflammatory process
  • Promotion of healing in damaged oral and throat mucous membranes
  • Protection of oral and throat mucous membranes
  • Strengthening of the local immune response

It’s worth noting that certain Lysobact速 products, such as Lysobact Spray速 and Lysobact Complete Spray速, are alcohol-free, making them suitable for individuals with sensitivities or those who prefer alcohol-free options.

Natural Remedies and Supportive Care

In addition to medical treatments, several natural remedies and supportive care measures can help manage stomatitis:

  1. Salt water rinses to reduce inflammation and promote healing
  2. Honey applications for its antimicrobial properties
  3. Aloe vera gel to soothe irritated tissues
  4. Chamomile tea rinses for their anti-inflammatory effects
  5. Dietary modifications to avoid irritating foods
  6. Proper oral hygiene practices

These complementary approaches can work alongside conventional treatments to enhance recovery and provide symptomatic relief.

Meningitis: When Inflammation Strikes the Brain and Spinal Cord

Meningitis, the inflammation of the membranes surrounding the brain and spinal cord, represents a more severe form of membrane inflammation. This condition can be life-threatening and requires immediate medical attention.

Types of Meningitis

  • Bacterial meningitis: Caused by various bacteria, often requiring intensive antibiotic treatment
  • Viral meningitis: Generally less severe but still serious, caused by viruses such as enteroviruses
  • Fungal meningitis: Rare but potentially fatal, often affecting immunocompromised individuals
  • Parasitic meningitis: Uncommon, typically associated with specific parasitic infections
  • Non-infectious meningitis: Can be caused by certain medications, cancers, or autoimmune disorders

Understanding the different types of meningitis is crucial for proper diagnosis and treatment, as each type may require a specific approach.

Recognizing Meningitis: Key Symptoms

Early recognition of meningitis symptoms is vital for prompt treatment. Common signs include:

  • Sudden high fever
  • Severe headache
  • Stiff neck
  • Sensitivity to light
  • Nausea and vomiting
  • Confusion or altered mental state
  • Seizures (in some cases)
  • Skin rash (particularly in meningococcal meningitis)

If these symptoms are present, especially in combination, immediate medical evaluation is necessary.

The Link Between Membrane Homeostasis and Neurodegenerative Diseases

Recent research has begun to explore the potential connection between membrane homeostasis and the development of neurodegenerative diseases. This emerging field of study offers new insights into the complex mechanisms underlying conditions such as Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis (ALS).

The Role of Protein Misfolding

One key aspect of this potential link involves protein misfolding and aggregation. In many neurodegenerative diseases, specific proteins adopt abnormal conformations, leading to their accumulation and the formation of toxic aggregates. These aggregates can disrupt membrane integrity and function, potentially triggering or exacerbating inflammation and neurodegeneration.

Examples of this phenomenon include:

  • Beta-amyloid plaques in Alzheimer’s disease
  • Alpha-synuclein aggregates in Parkinson’s disease
  • SOD1 and TDP-43 protein aggregates in ALS

Understanding the interplay between these misfolded proteins and cellular membranes could provide new targets for therapeutic interventions.

Membrane Lipid Composition and Neurodegenerative Diseases

Another aspect of the membrane homeostasis-neurodegeneration link involves the lipid composition of cellular membranes. Alterations in membrane lipid profiles have been observed in various neurodegenerative conditions, potentially affecting:

  1. Membrane fluidity and permeability
  2. Protein-lipid interactions
  3. Cellular signaling pathways
  4. Synaptic function and plasticity

These changes in membrane properties could contribute to the progression of neurodegenerative diseases by altering cellular function and vulnerability to stress.

Innovative Approaches to Treating Membrane Inflammation

As our understanding of membrane inflammation and its role in various diseases continues to evolve, researchers are exploring innovative treatment approaches that target specific aspects of the inflammatory process.

Nanotechnology-Based Therapies

Nanotechnology offers promising avenues for delivering anti-inflammatory agents directly to affected tissues. Some potential applications include:

  • Nanoparticles designed to cross the blood-brain barrier for treating neuroinflammation
  • Liposomal drug delivery systems for targeted release of anti-inflammatory compounds
  • Nanoformulations of existing drugs to enhance their efficacy and reduce side effects

These approaches could potentially improve the treatment of conditions ranging from stomatitis to neurodegenerative diseases.

Immunomodulatory Strategies

Modulating the immune response to inflammation represents another promising avenue for treatment. This approach aims to balance the beneficial aspects of inflammation (such as pathogen clearance) with the need to prevent excessive or chronic inflammatory responses.

Some strategies being explored include:

  1. Targeted cytokine inhibition
  2. Enhancement of regulatory T cell function
  3. Modulation of microglial activation in neuroinflammation
  4. Development of novel anti-inflammatory compounds derived from natural sources

These immunomodulatory approaches could offer more precise control over inflammatory processes, potentially leading to better outcomes in various inflammatory conditions.

Future Directions in Membrane Inflammation Research

As we look to the future, several key areas of research hold promise for advancing our understanding and treatment of membrane inflammation:

Personalized Medicine Approaches

The development of personalized treatment strategies based on an individual’s genetic profile, microbiome composition, and specific inflammatory markers could revolutionize the management of inflammatory conditions. This approach may allow for more targeted and effective interventions, minimizing side effects and improving overall outcomes.

Microbiome Modulation

Growing evidence suggests that the microbiome plays a crucial role in regulating inflammation throughout the body, including in the oral cavity and the gut-brain axis. Future research may focus on developing probiotic or prebiotic therapies to modulate the microbiome and reduce inflammatory responses.

Advanced Imaging Techniques

The development of new imaging technologies could provide unprecedented insights into the dynamics of membrane inflammation in real-time. These tools may allow researchers to:

  • Visualize inflammatory processes at the cellular and molecular levels
  • Track the progression of inflammation in various tissues
  • Assess the effectiveness of anti-inflammatory interventions more precisely

Such advancements could lead to earlier detection and more effective management of inflammatory conditions.

Artificial Intelligence and Big Data Analysis

The application of artificial intelligence and machine learning algorithms to large-scale datasets could uncover new patterns and relationships in membrane inflammation research. This approach may help identify:

  1. Novel biomarkers for early disease detection
  2. Previously unknown risk factors for inflammatory conditions
  3. Potential drug targets for anti-inflammatory therapies
  4. Personalized treatment strategies based on individual patient profiles

By harnessing the power of big data, researchers may be able to accelerate the pace of discovery and translation of findings into clinical practice.

Gene Therapy and CRISPR Technology

Advances in gene therapy and CRISPR gene-editing technology offer exciting possibilities for treating membrane inflammation at its genetic roots. Potential applications include:

  • Correcting genetic mutations associated with inflammatory disorders
  • Modifying genes involved in the inflammatory response to reduce excessive inflammation
  • Enhancing the expression of anti-inflammatory factors

While still in its early stages, this approach could potentially lead to long-lasting or even permanent solutions for certain inflammatory conditions.

As research in these areas continues to progress, we can anticipate significant advancements in our ability to understand, prevent, and treat membrane inflammation across a wide range of conditions. From common ailments like stomatitis to complex neurodegenerative diseases, the future holds promise for more effective and personalized approaches to managing inflammatory processes throughout the body.

Inflammation of the mucous membranes (stomatitis)

As the initial part of the digestive tract, the mouth is often exposed to various harmful influences, so it is often the seat of various pathological processes, in which inflammation of the oral mucosa occupies a significant place. The oral mucous membrane is part of the mucous membrane of the digestive tract and coats the oral cavity, and changes in it are also an indicator of many other diseases.

Inflammation of the oral mucosa (stomatitis) as a separate disease occurs due to a direct irritation that acts on the mucosa. The most common causes are viruses, but they can also be bacteria and fungi. In addition to infection as the main cause, inflammation of the mucous membrane of the oral cavity can be a symptom of a disease or it can be caused by allergies or some irritating factors. All factors of chemical, mechanical, and thermal nature that damage the protective surface layer of the oral mucosa affect the development of this disease. General diseases that reduce the body’s resistance, especially with poor oral hygiene, also contribute to the development of stomatitis.

Rough food, foreign bodies (e.g., bones, especially fish), dental diseases as well as tooth changes and congenital and acquired dental irregularities are irritating. The mucous membrane of the mouth can be damaged by various chemicals (acids and bases). Cold foods (from the refrigerator or insufficiently thawed), as well as overheated foods, can also reduce the local resistance of the oral mucosa and favor the development of inflammation.

Diseases that cause stomatitis are anemia and other blood diseases (leukemia, platelet disease), deficiency of certain vitamins (B or C vitamins most commonly), celiac disease or intolerance to gluten protein present in cereals, inflammatory bowel disease and others.

Stomatitis is a disease that affects the entire mucous membrane of the mouth, including the gums. Intense redness occurs, the mucous membrane is swollen, the tongue is coated, and the lips are slightly enlarged. The gums are inflamed, and the parts of the gums between the teeth are enlarged and bleed at the slightest touch. Increased salivation and bad breath are present. Patients have a feeling of rot in their mouth or that their mouth is bitter. Regional lymph nodes are enlarged and tender. The temperature is slightly increased, the appetite is weak, and the pain intensifies during meals or speech.

If the bacterium is the cause of the inflammation of the mucous membrane of the mouth, the therapy is antibiotic as a therapy against the cause itself. Oral antiseptics are often used in the treatment, which can be obtained without a prescription in a pharmacy with a different composition of active ingredients, with the aim of eliminating local symptoms.

Of all available oral antiseptics, the active ingredient lysozyme is found only in Lysobact® products.

Use Lysobact® products (Lysobact® compressed lozenges, Lysobact P Spray®, Lysobact Duo®, Lysobact Spray® or Lysobact Complete Spray®).   The active components of Lysobact® act against inflammatory agents (bacteria, viruses, and fungi), relieve symptoms, alleviate the inflammatory process, and help heal damaged oral and throat  mucous membranes. Lysobact® protects the oral and throat mucous membranes and strengthens the local immune response. Lysobact Spray® and Lysobact Complete Spray® do not contain alcohol.

Ova stranica je također dostupna na:
Bosnian Albanian Macedonian Serbian Ukrainian Croatian

Is membrane homeostasis the missing link between inflammation and neurodegenerative diseases?

1. Carrell RW. Cell toxicity and conformational disease. Trends Cell Biol. 2005;15(11):574–580. doi: 10.1016/j.tcb.2005.09.005. [PubMed] [CrossRef] [Google Scholar]

2. Carrell RW, Lomas DA. Conformational disease. Lancet. 1997;350(9071):134–138. doi: 10.1016/S0140-6736(97)02073-4. [PubMed] [CrossRef] [Google Scholar]

3. Bertram L, Tanzi RE. The genetic epidemiology of neurodegenerative disease. J Clin Invest. 2005;115(6):1449–1457. doi: 10.1172/JCI24761. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

4. Lu JX, Qiang W, Yau WM, Schwieters CD, Meredith SC, Tycko R. Molecular structure of beta-amyloid fibrils in Alzheimer’s disease brain tissue. Cell. 2013;154(6):1257–1268. doi: 10.1016/j.cell.2013.08.035. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

5. Julien JP. Amyotrophic lateral sclerosis. Unfolding the toxicity of the misfolded. Cell. 2001;104(4):581–591. doi: 10.1016/S0092-8674(01)00244-6. [PubMed] [CrossRef] [Google Scholar]

6. Lashuel HA, Overk CR, Oueslati A, Masliah E. The many faces of alpha-synuclein: from structure and toxicity to therapeutic target. Nat Rev Neurosci. 2013;14(1):38–48. doi: 10.1038/nrn3406. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

7. Gidalevitz T, Kikis EA, Morimoto RI. A cellular perspective on conformational disease: the role of genetic background and proteostasis networks. Curr Opin Struct Biol. 2010;20(1):23–32. doi: 10.1016/j.sbi.2009.11.001. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

8. Sankowski R, Mader S, Valdes-Ferrer SI. Systemic inflammation and the brain: novel roles of genetic, molecular, and environmental cues as drivers of neurodegeneration. Front Cell Neurosci. 2015;9:28. doi: 10.3389/fncel.2015.00028. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

9. Amor S, Puentes F, Baker D, van der Valk P. Inflammation in neurodegenerative diseases. Immunology. 2010;129(2):154–169. doi: 10.1111/j.1365-2567.2009.03225.x. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

10. Perry VH. The influence of systemic inflammation on inflammation in the brain: implications for chronic neurodegenerative disease. Brain Behav Immun. 2004;18(5):407–413. doi: 10.1016/j.bbi.2004.01.004. [PubMed] [CrossRef] [Google Scholar]

11. Guyon A, Massa F, Rovere C, Nahon JL. How cytokines can influence the brain: a role for chemokines? J Neuroimmunol. 2008;198(1–2):46–55. doi: 10.1016/j.jneuroim.2008.04.009. [PubMed] [CrossRef] [Google Scholar]

12. Luster AD. The role of chemokines in linking innate and adaptive immunity. Curr Opin Immunol. 2002;14(1):129–135. doi: 10.1016/S0952-7915(01)00308-9. [PubMed] [CrossRef] [Google Scholar]

13. Hanisch UK, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007;10(11):1387–1394. doi: 10.1038/nn1997. [PubMed] [CrossRef] [Google Scholar]

14. Terrando N, Rei Fidalgo A, Vizcaychipi M, Cibelli M, Ma D, Monaco C, Feldmann M, Maze M. The impact of IL-1 modulation on the development of lipopolysaccharide-induced cognitive dysfunction. Crit Care. 2010;14(3):R88. doi: 10.1186/cc9019. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

15. Anderson ST, Commins S, Moynagh PN, Coogan AN. Lipopolysaccharide-induced sepsis induces long-lasting affective changes in the mouse. Brain Behav Immun. 2015;43:98–109. doi: 10. 1016/j.bbi.2014.07.007. [PubMed] [CrossRef] [Google Scholar]

16. Li J, Tang Y, Cai D. IKKbeta/NF-kappaB disrupts adult hypothalamic neural stem cells to mediate a neurodegenerative mechanism of dietary obesity and pre-diabetes. Nat Cell Biol. 2012;14(10):999–1012. doi: 10.1038/ncb2562. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

17. Lemstra AW, Groen in’t Woud JC, Hoozemans JJ, van Haastert ES, Rozemuller AJ, Eikelenboom P, van Gool WA. Microglia activation in sepsis: a case–control study. J Neuroinflamm. 2007;4:4. doi: 10.1186/1742-2094-4-4. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

18. Kuo HK, Yen CJ, Chang CH, Kuo CK, Chen JH, Sorond F. Relation of C-reactive protein to stroke, cognitive disorders, and depression in the general population: systematic review and meta-analysis. Lancet Neurol. 2005;4(6):371–380. doi: 10.1016/S1474-4422(05)70099-5. [PubMed] [CrossRef] [Google Scholar]

19. Buchhave P, Zetterberg H, Blennow K, Minthon L, Janciauskiene S, Hansson O. Soluble TNF receptors are associated with Abeta metabolism and conversion to dementia in subjects with mild cognitive impairment. Neurobiol Aging. 2010;31(11):1877–1884. doi: 10.1016/j.neurobiolaging.2008.10.012. [PubMed] [CrossRef] [Google Scholar]

20. Huang Y, Mucke L. Alzheimer mechanisms and therapeutic strategies. Cell. 2012;148(6):1204–1222. doi: 10.1016/j.cell.2012.02.040. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

21. Ittner LM, Gotz J. Amyloid-beta and tau—a toxic pas de deux in Alzheimer’s disease. Nat Rev Neurosci. 2011;12(2):65–72. doi: 10.1038/nrn2967. [PubMed] [CrossRef] [Google Scholar]

22. Barnett K, Mercer SW, Norbury M, Watt G, Wyke S, Guthrie B. Epidemiology of multimorbidity and implications for health care, research, and medical education: a cross-sectional study. Lancet. 2012;380(9836):37–43. doi: 10.1016/S0140-6736(12)60240-2. [PubMed] [CrossRef] [Google Scholar]

23. Holmes C, Cunningham C, Zotova E, Woolford J, Dean C, Kerr S, Culliford D, Perry VH. Systemic inflammation and disease progression in Alzheimer disease. Neurology. 2009;73(10):768–774. doi: 10.1212/WNL.0b013e3181b6bb95. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

24. Holmes C, Cunningham C, Zotova E, Culliford D, Perry VH. Proinflammatory cytokines, sickness behavior, and Alzheimer disease. Neurology. 2011;77(3):212–218. doi: 10.1212/WNL.0b013e318225ae07. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

25. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, Herrup K, Frautschy SA, Finsen B, Brown GC, Verkhratsky A, Yamanaka K, Koistinaho J, Latz E, Halle A, Petzold GC, Town T, Morgan D, Shinohara ML, Perry VH, Holmes C, Bazan NG, Brooks DJ, Hunot S, Joseph B, Deigendesch N, Garaschuk O, Boddeke E, Dinarello CA, Breitner JC, Cole GM, Golenbock DT, Kummer MP. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14(4):388–405. doi: 10.1016/S1474-4422(15)70016-5. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

26. Krstic D, Madhusudan A, Doehner J, Vogel P, Notter T, Imhof C, Manalastas A, Hilfiker M, Pfister S, Schwerdel C, Riether C, Meyer U, Knuesel I. Systemic immune challenges trigger and drive Alzheimer-like neuropathology in mice. J Neuroinflamm. 2012;9:151. doi: 10.1186/1742-2094-9-151. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

27. Krstic D, Knuesel I. Deciphering the mechanism underlying late-onset Alzheimer disease. Nat Rev Neurol. 2013;9(1):25–34. doi: 10.1038/nrneurol.2012.236. [PubMed] [CrossRef] [Google Scholar]

28. Kitazawa M, Oddo S, Yamasaki TR, Green KN, LaFerla FM. Lipopolysaccharide-induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated pathway in a transgenic model of Alzheimer’s disease. J Neurosci. 2005;25(39):8843–8853. doi: 10.1523/JNEUROSCI.2868-05.2005. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

29. Bhaskar K, Konerth M, Kokiko-Cochran ON, Cardona A, Ransohoff RM, Lamb BT. Regulation of tau pathology by the microglial fractalkine receptor. Neuron. 2010;68(1):19–31. doi: 10.1016/j.neuron.2010.08.023. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

30. Cardona AE, Pioro EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra IM, Huang D, Kidd G, Dombrowski S, Dutta R, Lee JC, Cook DN, Jung S, Lira SA, Littman DR, Ransohoff RM. Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci. 2006;9(7):917–924. doi: 10.1038/nn1715. [PubMed] [CrossRef] [Google Scholar]

31. Cho SH, Sun B, Zhou Y, Kauppinen TM, Halabisky B, Wes P, Ransohoff RM, Gan L. CX3CR1 protein signaling modulates microglial activation and protects against plaque-independent cognitive deficits in a mouse model of Alzheimer disease. J Biol Chem. 2011;286(37):32713–32722. doi: 10.1074/jbc.M111.254268. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

32. Ozawa Y. Neurodegenerative disease: pieces of the Parkinson’s puzzle. Nat Rev Neurosci. 2010;11(12):787. doi: 10.1038/nrn2945. [PubMed] [CrossRef] [Google Scholar]

33. Hirsch EC, Hunot S. Neuroinflammation in Parkinson’s disease: a target for neuroprotection? Lancet Neurol. 2009;8(4):382–397. doi: 10.1016/S1474-4422(09)70062-6. [PubMed] [CrossRef] [Google Scholar]

34. Choi DY, Liu M, Hunter RL, Cass WA, Pandya JD, Sullivan PG, Shin EJ, Kim HC, Gash DM, Bing G. Striatal neuroinflammation promotes Parkinsonism in rats. PLoS ONE. 2009;4(5):e5482. doi: 10.1371/journal.pone.0005482. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

35. Tanaka S, Ishii A, Ohtaki H, Shioda S, Yoshida T, Numazawa S. Activation of microglia induces symptoms of Parkinson’s disease in wild-type, but not in IL-1 knockout mice. J Neuroinflamm. 2013;10:143. doi: 10.1186/1742-2094-10-143. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

36. Nash KR, Moran P, Finneran DJ, Hudson C, Robinson J, Morgan D, Bickford PC. Fractalkine over expression suppresses alpha-synuclein-mediated neurodegeneration. Mol Ther. 2015;23(1):17–23. doi: 10.1038/mt.2014.175. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

37. Ajroud-Driss S, Siddique-Driss T. Sporadic and hereditary amyotrophic lateral sclerosis (ALS) Biochim Biophys Acta. 2015;1852(4):679–684. doi: 10.1016/j.bbadis.2014.08.010. [PubMed] [CrossRef] [Google Scholar]

38. Frischer JM, Bramow S, Dal-Bianco A, Lucchinetti CF, Rauschka H, Schmidbauer M, Laursen H, Sorensen PS, Lassmann H. The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain. 2009;132(Pt 5):1175–1189. doi: 10.1093/brain/awp070. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

39. Frakes AE, Ferraiuolo L, Haidet-Phillips AM, Schmelzer L, Braun L, Miranda CJ, Ladner KJ, Bevan AK, Foust KD, Godbout JP, Popovich PG, Guttridge DC, Kaspar BK. Microglia induce motor neuron death via the classical NF-kappaB pathway in amyotrophic lateral sclerosis. Neuron. 2014;81(5):1009–1023. doi: 10.1016/j.neuron.2014.01.013. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

40. Solberg NO, Chamberlin R, Vigil JR, Deck LM, Heidrich JE, Brown DC, Brady CI, Vander Jagt TA, Garwood M, Bisoffi M, Severns V, Vander Jagt DL, Sillerud LO. Optical and SPION-enhanced MR imaging shows that trans-stilbene inhibitors of NF-kappaB concomitantly lower Alzheimer’s disease plaque formation and microglial activation in AbetaPP/PS-1 transgenic mouse brain. J Alzheimers Dis. 2014;40(1):191–212. [PMC free article] [PubMed] [Google Scholar]

41. Wang S, Jing H, Yang H, Liu Z, Guo H, Chai L, Hu L. Tanshinone I selectively suppresses pro-inflammatory genes expression in activated microglia and prevents nigrostriatal dopaminergic neurodegeneration in a mouse model of Parkinsons disease. J Ethnopharmacol. 2015;164:247–255. doi: 10.1016/j.jep.2015.01.042. [PubMed] [CrossRef] [Google Scholar]

42. Marchetto MC, Muotri AR, Mu Y, Smith AM, Cezar GG, Gage FH. Non-cell-autonomous effect of human SOD1 G37R astrocytes on motor neurons derived from human embryonic stem cells. Cell Stem Cell. 2008;3(6):649–657. doi: 10.1016/j.stem.2008.10.001. [PubMed] [CrossRef] [Google Scholar]

43. Saijo K, Winner B, Carson CT, Collier JG, Boyer L, Rosenfeld MG, Gage FH, Glass CK. A Nurr1/CoREST pathway in microglia and astrocytes protects dopaminergic neurons from inflammation-induced death. Cell. 2009;137(1):47–59. doi: 10.1016/j.cell.2009.01.038. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

44. Moon M, Jeong I, Kim CH, Kim J, Lee PK, Mook-Jung I, Leblanc P, Kim KS. Correlation between orphan nuclear receptor Nurr1 expression and amyloid deposition in 5XFAD mice, an animal model of Alzheimer’s disease. J Neurochem. 2015;132(2):254–262. doi: 10.1111/jnc.12935. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

45. Bates GP. History of genetic disease: the molecular genetics of Huntington disease—a history. Nat Rev Genet. 2005;6(10):766–773. doi: 10.1038/nrg1686. [PubMed] [CrossRef] [Google Scholar]

46. Khoshnan A, Ko J, Watkin EE, Paige LA, Reinhart PH, Patterson PH. Activation of the IkappaB kinase complex and nuclear factor-kappaB contributes to mutant huntingtin neurotoxicity. J Neurosci. 2004;24(37):7999–8008. doi: 10.1523/JNEUROSCI.2675-04.2004. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

47. Kraft AD, Kaltenbach LS, Lo DC, Harry GJ. Activated microglia proliferate at neurites of mutant huntingtin-expressing neurons. Neurobiol Aging. 2012;33(3):621.e617-633. doi: 10.1016/j.neurobiolaging.2011.02.015. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

48. Nicolson GL. The fluid-mosaic model of membrane structure: still relevant to understanding the structure, function and dynamics of biological membranes after more than 40 years. Biochim Biophys Acta. 2014;1838(6):1451–1466. doi: 10.1016/j.bbamem.2013.10.019. [PubMed] [CrossRef] [Google Scholar]

49. Pamplona R. Membrane phospholipids, lipoxidative damage and molecular integrity: a causal role in aging and longevity. Biochim Biophys Acta. 2008;1777(10):1249–1262. doi: 10.1016/j.bbabio.2008.07.003. [PubMed] [CrossRef] [Google Scholar]

50. Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8(1):57–69. doi: 10.1038/nrn2038. [PubMed] [CrossRef] [Google Scholar]

51. Lull ME, Block ML. Microglial activation and chronic neurodegeneration. Neurotherapeutics. 2010;7(4):354–365. doi: 10.1016/j.nurt.2010.05.014. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

52. de la Haba C, Palacio JR, Martinez P, Morros A. Effect of oxidative stress on plasma membrane fluidity of THP-1 induced macrophages. Biochim Biophys Acta. 2013;1828(2):357–364. doi: 10.1016/j.bbamem.2012.08.013. [PubMed] [CrossRef] [Google Scholar]

53. Lu SP, Lin Feng MH, Huang HL, Huang YC, Tsou WI, Lai MZ. Reactive oxygen species promote raft formation in T lymphocytes. Free Radic Biol Med. 2007;42(7):936–944. doi: 10.1016/j.freeradbiomed.2006.11.027. [PubMed] [CrossRef] [Google Scholar]

54. Yang B, Oo TN, Rizzo V. Lipid rafts mediate h3O2 prosurvival effects in cultured endothelial cells. FASEB J. 2006;20(9):1501–1503. doi: 10.1096/fj.05-5359fje. [PubMed] [CrossRef] [Google Scholar]

55. Epand RF, Ramamoorthy A, Epand RM. Membrane lipid composition and the interaction of pardaxin: the role of cholesterol. Protein Pept Lett. 2006;13(1):1–5. [PubMed] [Google Scholar]

56. Torrent M, Valle J, Nogues MV, Boix E, Andreu D. The generation of antimicrobial peptide activity: a trade-off between charge and aggregation? Angew Chem Int Ed Engl. 2011;50(45):10686–10689. doi: 10.1002/anie.201103589. [PubMed] [CrossRef] [Google Scholar]

57. Torrent M, Andreu D, Nogues VM, Boix E. Connecting peptide physicochemical and antimicrobial properties by a rational prediction model. PLoS ONE. 2011;6(2):e16968. doi: 10.1371/journal.pone.0016968. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

58. Ledesma MD, Martin MG, Dotti CG. Lipid changes in the aged brain: effect on synaptic function and neuronal survival. Prog Lipid Res. 2012;51(1):23–35. doi: 10.1016/j.plipres.2011.11.004. [PubMed] [CrossRef] [Google Scholar]

59. Gauthier-Kemper A, Weissmann C, Golovyashkina N, Sebo-Lemke Z, Drewes G, Gerke V, Heinisch JJ, Brandt R. The frontotemporal dementia mutation R406W blocks tau’s interaction with the membrane in an annexin A2-dependent manner. J Cell Biol. 2011;192(4):647–661. doi: 10.1083/jcb.201007161. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

60. Auluck PK, Caraveo G, Lindquist S. alpha-Synuclein: membrane interactions and toxicity in Parkinson’s disease. Annu Rev Cell Dev Biol. 2010;26:211–233. doi: 10.1146/annurev.cellbio.042308.113313. [PubMed] [CrossRef] [Google Scholar]

61. Pope CR, De Feo CJ, Unger VM. Cellular distribution of copper to superoxide dismutase involves scaffolding by membranes. Proc Natl Acad Sci USA. 2013;110(51):20491–20496. doi: 10.1073/pnas.1309820110. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

62. Butterfield SM, Lashuel HA. Amyloidogenic protein-membrane interactions: mechanistic insight from model systems. Angew Chem Int Ed Engl. 2010;49(33):5628–5654. doi: 10.1002/anie.200906670. [PubMed] [CrossRef] [Google Scholar]

63. Kotarek JA, Moss MA. Impact of phospholipid bilayer saturation on amyloid-beta protein aggregation intermediate growth: a quartz crystal microbalance analysis. Anal Biochem. 2010;399(1):30–38. doi: 10.1016/j.ab.2009.12.016. [PubMed] [CrossRef] [Google Scholar]

64. Williamson R, Sutherland C. Neuronal membranes are key to the pathogenesis of Alzheimer’s disease: the role of both raft and non-raft membrane domains. Curr Alzheimer Res. 2011;8(2):213–221. doi: 10.2174/156720511795256008. [PubMed] [CrossRef] [Google Scholar]

65. Engel MF, Khemtemourian L, Kleijer CC, Meeldijk HJ, Jacobs J, Verkleij AJ, de Kruijff B, Killian JA, Hoppener JW. Membrane damage by human islet amyloid polypeptide through fibril growth at the membrane. Proc Natl Acad Sci USA. 2008;105(16):6033–6038. doi: 10.1073/pnas.0708354105. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

66. Rivest S. Regulation of innate immune responses in the brain. Nat Rev Immunol. 2009;9(6):429–439. doi: 10.1038/nri2565. [PubMed] [CrossRef] [Google Scholar]

67. Rushworth JV, Hooper NM. Lipid rafts: linking Alzheimer’s amyloid-beta production, aggregation, and toxicity at neuronal membranes. Int J Alzheimers Dis. 2010;2011:603052. [PMC free article] [PubMed] [Google Scholar]

68. Yoda M, Miura T, Takeuchi H. Non-electrostatic binding and self-association of amyloid beta-peptide on the surface of tightly packed phosphatidylcholine membranes. Biochem Biophys Res Commun. 2008;376(1):56–59. doi: 10.1016/j.bbrc.2008.08.093. [PubMed] [CrossRef] [Google Scholar]

69. Malnar M, Kosicek M, Lisica A, Posavec M, Krolo A, Njavro J, Omerbasic D, Tahirovic S, Hecimovic S. Cholesterol-depletion corrects APP and BACE1 misstrafficking in NPC1-deficient cells. Biochim Biophys Acta. 2012;1822(8):1270–1283. doi: 10.1016/j.bbadis.2012.04.002. [PubMed] [CrossRef] [Google Scholar]

70. Marquer C, Devauges V, Cossec JC, Liot G, Lecart S, Saudou F, Duyckaerts C, Leveque-Fort S, Potier MC. Local cholesterol increase triggers amyloid precursor protein-Bace1 clustering in lipid rafts and rapid endocytosis. FASEB J. 2011;25(4):1295–1305. doi: 10.1096/fj.10-168633. [PubMed] [CrossRef] [Google Scholar]

71. Chochina SV, Avdulov NA, Igbavboa U, Cleary JP, O’Hare EO, Wood WG. Amyloid beta-peptide1-40 increases neuronal membrane fluidity: role of cholesterol and brain region. J Lipid Res. 2001;42(8):1292–1297. [PubMed] [Google Scholar]

72. Oksman M, Iivonen H, Hogyes E, Amtul Z, Penke B, Leenders I, Broersen L, Lutjohann D, Hartmann T, Tanila H. Impact of different saturated fatty acid, polyunsaturated fatty acid and cholesterol containing diets on beta-amyloid accumulation in APP/PS1 transgenic mice. Neurobiol Dis. 2006;23(3):563–572. doi: 10.1016/j.nbd.2006.04.013. [PubMed] [CrossRef] [Google Scholar]

73. Maas T, Eidenmuller J, Brandt R. Interaction of tau with the neural membrane cortex is regulated by phosphorylation at sites that are modified in paired helical filaments. J Biol Chem. 2000;275(21):15733–15740. doi: 10.1074/jbc.M000389200. [PubMed] [CrossRef] [Google Scholar]

74. Hernandez P, Lee G, Sjoberg M, Maccioni RB. Tau phosphorylation by cdk5 and Fyn in response to amyloid peptide Abeta (25-35): involvement of lipid rafts. J Alzheimers Dis. 2009;16(1):149–156. [PubMed] [Google Scholar]

75. Elbaum-Garfinkle S, Ramlall T, Rhoades E. The role of the lipid bilayer in tau aggregation. Biophys J. 2010;98(11):2722–2730. doi: 10.1016/j.bpj.2010.03.013. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

76. Jones EM, Dubey M, Camp PJ, Vernon BC, Biernat J, Mandelkow E, Majewski J, Chi EY. Interaction of tau protein with model lipid membranes induces tau structural compaction and membrane disruption. Biochemistry. 2012;51(12):2539–2550. doi: 10.1021/bi201857v. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

77. Kuhla B, Haase C, Flach K, Luth HJ, Arendt T, Munch G. Effect of pseudophosphorylation and cross-linking by lipid peroxidation and advanced glycation end product precursors on tau aggregation and filament formation. J Biol Chem. 2007;282(10):6984–6991. doi: 10.1074/jbc.M609521200. [PubMed] [CrossRef] [Google Scholar]

78. Lotharius J, Brundin P. Pathogenesis of Parkinson’s disease: dopamine, vesicles and alpha-synuclein. Nat Rev Neurosci. 2002;3(12):932–942. doi: 10.1038/nrn983. [PubMed] [CrossRef] [Google Scholar]

79. Stefanis L. alpha-Synuclein in Parkinson’s disease. Cold Spring Harb Perspect Med. 2012;2(2):a009399. doi: 10.1101/cshperspect.a009399. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

80. Lee HJ, Choi C, Lee SJ. Membrane-bound alpha-synuclein has a high aggregation propensity and the ability to seed the aggregation of the cytosolic form. J Biol Chem. 2002;277(1):671–678. doi: 10.1074/jbc.M107045200. [PubMed] [CrossRef] [Google Scholar]

81. Fortin DL, Troyer MD, Nakamura K, Kubo S, Anthony MD, Edwards RH. Lipid rafts mediate the synaptic localization of alpha-synuclein. J Neurosci. 2004;24(30):6715–6723. doi: 10.1523/JNEUROSCI.1594-04.2004. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

82. Pranke IM, Morello V, Bigay J, Gibson K, Verbavatz JM, Antonny B, Jackson CL. alpha-Synuclein and ALPS motifs are membrane curvature sensors whose contrasting chemistry mediates selective vesicle binding. J Cell Biol. 2011;194(1):89–103. doi: 10.1083/jcb.201011118. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

83. Dexter DT, Carter CJ, Wells FR, Javoy-Agid F, Agid Y, Lees A, Jenner P, Marsden CD. Basal lipid peroxidation in substantia nigra is increased in Parkinson’s disease. J Neurochem. 1989;52(2):381–389. doi: 10.1111/j.1471-4159.1989.tb09133.x. [PubMed] [CrossRef] [Google Scholar]

84. Bosco DA, Fowler DM, Zhang Q, Nieva J, Powers ET, Wentworth P, Jr, Lerner RA, Kelly JW. Elevated levels of oxidized cholesterol metabolites in Lewy body disease brains accelerate alpha-synuclein fibrilization. Nat Chem Biol. 2006;2(5):249–253. doi: 10.1038/nchembio782. [PubMed] [CrossRef] [Google Scholar]

85. Giasson BI, Duda JE, Murray IV, Chen Q, Souza JM, Hurtig HI, Ischiropoulos H, Trojanowski JQ, Lee VM. Oxidative damage linked to neurodegeneration by selective alpha-synuclein nitration in synucleinopathy lesions. Science. 2000;290(5493):985–989. doi: 10.1126/science.290.5493.985. [PubMed] [CrossRef] [Google Scholar]

86. Sevcsik E, Trexler AJ, Dunn JM, Rhoades E. Allostery in a disordered protein: oxidative modifications to alpha-synuclein act distally to regulate membrane binding. J Am Chem Soc. 2011;133(18):7152–7158. doi: 10.1021/ja2009554. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

87. Pasinelli P, Brown RH. Molecular biology of amyotrophic lateral sclerosis: insights from genetics. Nat Rev Neurosci. 2006;7(9):710–723. doi: 10.1038/nrn1971. [PubMed] [CrossRef] [Google Scholar]

88. Zhai J, Strom AL, Kilty R, Venkatakrishnan P, White J, Everson WV, Smart EJ, Zhu H. Proteomic characterization of lipid raft proteins in amyotrophic lateral sclerosis mouse spinal cord. FEBS J. 2009;276(12):3308–3323. doi: 10.1111/j.1742-4658.2009.07057.x. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

89. Karch CM, Prudencio M, Winkler DD, Hart PJ, Borchelt DR. Role of mutant SOD1 disulfide oxidation and aggregation in the pathogenesis of familial ALS. Proc Natl Acad Sci USA. 2009;106(19):7774–7779. doi: 10.1073/pnas.0902505106. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

90. Kim YJ, Nakatomi R, Akagi T, Hashikawa T, Takahashi R. Unsaturated fatty acids induce cytotoxic aggregate formation of amyotrophic lateral sclerosis-linked superoxide dismutase 1 mutants. J Biol Chem. 2005;280(22):21515–21521. doi: 10.1074/jbc.M502230200. [PubMed] [CrossRef] [Google Scholar]

91. Hadano S, Kunita R, Otomo A, Suzuki-Utsunomiya K, Ikeda JE. Molecular and cellular function of ALS2/alsin: implication of membrane dynamics in neuronal development and degeneration. Neurochem Int. 2007;51(2–4):74–84. doi: 10.1016/j.neuint.2007.04.010. [PubMed] [CrossRef] [Google Scholar]

92. Kumar S, Zhou Y, Plamann M. Dynactin-membrane interaction is regulated by the C-terminal domains of p150(Glued) EMBO Rep. 2001;2(10):939–944. doi: 10.1093/embo-reports/kve202. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

93. Guan WJ, Xia KD, Ma YT, Liu YT, Shi YT, Jiang H, Shen L, Xia K, Li JD, Tang BS, Wang JL. Transglutaminase 6 interacts with polyQ proteins and promotes the formation of polyQ aggregates. Biochem Biophys Res Commun. 2013;437(1):94–100. doi: 10.1016/j.bbrc.2013.06.044. [PubMed] [CrossRef] [Google Scholar]

94. Park SH, Kukushkin Y, Gupta R, Chen T, Konagai A, Hipp MS, Hayer-Hartl M, Hartl FU. PolyQ proteins interfere with nuclear degradation of cytosolic proteins by sequestering the Sis1p chaperone. Cell. 2013;154(1):134–145. doi: 10.1016/j.cell.2013.06.003. [PubMed] [CrossRef] [Google Scholar]

95. Yanai A, Huang K, Kang R, Singaraja RR, Arstikaitis P, Gan L, Orban PC, Mullard A, Cowan CM, Raymond LA, Drisdel RC, Green WN, Ravikumar B, Rubinsztein DC, El-Husseini A, Hayden MR. Palmitoylation of huntingtin by HIP14 is essential for its trafficking and function. Nat Neurosci. 2006;9(6):824–831. doi: 10.1038/nn1702. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

96. Bertoni A, Giuliano P, Galgani M, Rotoli D, Ulianich L, Adornetto A, Santillo MR, Porcellini A, Avvedimento VE. Early and late events induced by polyQ-expanded proteins: identification of a common pathogenic property of polYQ-expanded proteins. J Biol Chem. 2011;286(6):4727–4741. doi: 10.1074/jbc.M110.156521. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

97. Burke KA, Hensal KM, Umbaugh CS, Chaibva M. Legleiter J (2013) Huntingtin disrupts lipid bilayers in a polyQ-length dependent manner. Biochim Biophys Acta. 1828;8:1953–1961. [PubMed] [Google Scholar]

98. Hajieva P, Bayatti N, Granold M, Behl C, Moosmann B. Membrane protein oxidation determines neuronal degeneration. J Neurochem. 2015;133(3):352–367. doi: 10.1111/jnc.12987. [PubMed] [CrossRef] [Google Scholar]

99. Fallahi-Sichani M, Linderman JJ. Lipid raft-mediated regulation of G-protein coupled receptor signaling by ligands which influence receptor dimerization: a computational study. PLoS ONE. 2009;4(8):e6604. doi: 10.1371/journal.pone.0006604. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

100. Thathiah A, De Strooper B. The role of G protein-coupled receptors in the pathology of Alzheimer’s disease. Nat Rev Neurosci. 2011;12(2):73–87. doi: 10.1038/nrn2977. [PubMed] [CrossRef] [Google Scholar]

101. Baglioni S, Casamenti F, Bucciantini M, Luheshi LM, Taddei N, Chiti F, Dobson CM, Stefani M. Prefibrillar amyloid aggregates could be generic toxins in higher organisms. J Neurosci. 2006;26(31):8160–8167. doi: 10.1523/JNEUROSCI.4809-05.2006. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

102. Reynolds NP, Soragni A, Rabe M, Verdes D, Liverani E, Handschin S, Riek R, Seeger S. Mechanism of membrane interaction and disruption by alpha-synuclein. J Am Chem Soc. 2011;133(48):19366–19375. doi: 10.1021/ja2029848. [PubMed] [CrossRef] [Google Scholar]

103. Gustot A, Raussens V, Dehousse M, Dumoulin M, Bryant CE, Ruysschaert JM, Lonez C. Activation of innate immunity by lysozyme fibrils is critically dependent on cross-beta sheet structure. Cell Mol Life Sci. 2013;70(16):2999–3012. doi: 10.1007/s00018-012-1245-5. [PubMed] [CrossRef] [Google Scholar]

104. Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D, Remus A, Tzeng TC, Gelpi E, Halle A, Korte M, Latz E, Golenbock DT. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493(7434):674–678. doi: 10.1038/nature11729. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

105. Coll RC, Robertson AA, Chae JJ, Higgins SC, Munoz-Planillo R, Inserra MC, Vetter I, Dungan LS, Monks BG, Stutz A, Croker DE, Butler MS, Haneklaus M, Sutton CE, Nunez G, Latz E, Kastner DL, Mills KH, Masters SL, Schroder K, Cooper MA, O’Neill LA. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med. 2015;21(3):248–255. [PMC free article] [PubMed] [Google Scholar]

Treatment of otitis media, how to treat otitis media

  1. EAR DISEASES
  2. Otitis media

Otitis media is a disease in which there is inflammation of the mucous membrane in the middle ear cavity. The cause of this condition is a violation of the patency of the so-called Eustachian tube – a thin channel connecting the middle ear cavity with the nasal cavity.

The Eustachian tube normally performs a drainage function – through it, mucus is discharged from the middle ear into the nasal cavity and atmospheric pressure is equalized. The patency of the Eustachian tube can be impaired in the presence of any volumetric formations in the nasal cavity – adenoid, polyps, neoplasms, as well as in any inflammatory diseases of the nasal mucosa (acute and chronic).

The Eustachian tube is a canal that connects the tympanic cavity with the nasopharynx.

The tube, if you look at the transverse section, consists of two halves – a dense cartilaginous semi-canal and a soft membranous. These halves are most of the time in a “sticky” state, and “unstick” only during the swallowing movement.

Eardrum normal.

This is a thin, translucent membrane that captures sound vibrations and transmits them through the ossicular chain to the sensory nerve cells (receptors) in the inner ear. The membrane is firmly connected to the auditory ossicle with the malleus, or rather with its process, called the handle of the malleus. The chain of auditory ossicles “hammer-anvil-stapes” creates a system of levers and amplifies sound vibrations by 2.5 times.

And here is how both mouths in the nasopharynx look at once when comparing endoscopic images.

What is all this for?

Indeed, why such difficulties? Why should nature create such a complex mechanism if it did not initially create a person either for flying in the air or for diving under water. The fact is that the mucous membrane of the ear cavity, like any other, contains a huge number of blood vessels. These vessels come very close to the surface. Blood flows through the vessels. Red blood cells are contained in the blood. Red blood cells contain a special protein, hemoglobin, which binds oxygen and transports it through tissues and organs.

That’s the problem! It turns out that the blood vessels “suck” oxygen from the air in the tympanic cavity. And the air pressure inside the tympanic cavity decreases. If the auditory tube is closed and the pressure does not equalize, the vacuum inside the tympanic cavity causes the compliant eardrum to retract inward. This makes it difficult to transmit sounds and the person feels stuffy in the ear.

What can block the Eustachian tube?

  1. Edema of the mucous membrane of the nasal cavity with any (!) inflammation in the nose and paranasal sinuses.
  2. Any volumetric processes in the nasopharynx (adenoid, polyposis, benign and malignant neoplasms).
  3. Anatomical narrowness of the Eustachian tube
  4. Impaired mobility of the muscles that open the Eustachian tube (palatolingual and palatopharyngeal)

If the eustachian tube block continues, the negative pressure inside the tympanic cavity causes blood plasma to leak through the blood vessel wall. This is a non-inflammatory fluid or so-called transudate. The transudate in the tympanic cavity further disrupts the movements of the tympanic membrane and auditory ossicles, which leads to even greater hearing loss (up to 30% of the norm).

Fragments of an enlarged adenoid (grade 2) obstructing the orifice of the Eustachian tube (arrow).

Retracted tympanic membrane.

This is especially evident in the upper section (arrow), where the membrane is anatomically thinner.

Transudate behind the eardrum.

Tympanic cavity 3/4 full. The liquid level is clearly visible.

In the presence of fluid in the tympanic cavity, a nasal endoscopy is mandatory to determine the cause of the blockade of the Eustachian tube. In the presence of an inflammatory process in the nasopharynx, it is treated and, as a rule, this leads to the recovery of the ear. If there is an enlarged adenoid covering the mouth of the tube, its surgical removal is recommended. In the absence of obvious inflammatory phenomena in the nose and volumetric processes in the area of ​​​​the mouth of the Eustachian tube and while maintaining fluid in the tympanic cavity, they resort to shunting the eardrum.

Bypass surgery is the insertion of a special plastic tube into a small incision in the eardrum. It is shaped like a spool of thread. This tube allows fluid to escape into the ear canal and the patient does not suffer from hearing loss.

Endoscopic picture in acute otitis media.

If the accumulation of mucus behind the eardrum is provoked by an acute respiratory infection, then the transudate can become a breeding ground for the development of pathogenic microbes and cause acute inflammation of the mucous membrane of the middle ear – acute otitis media.

This is due to the fact that viral respiratory infections suppress the activity of local immunity and bacterial inflammation is always “superimposed” on any SARS.

Remember how a runny nose occurs during a cold: first, a stage of clear serous discharge (caused by a reaction to a virus), then, after 5-7 days, a stage of thick mucopurulent discharge (caused by a reaction to a bacterial infection).

Accumulated thick discharge cannot pass through a closed Eustachian tube. It bursts the tympanic cavity from the inside, which causes excruciating pains in the ear, often pulsating, and the body temperature rises. The tympanic membrane is hyperemic (reddened), often bulges outward.

If the accumulation of transudate in the tympanic cavity is caused by a chronic blockade of the Eustachian tube (adenoid, neoplasms), it can continue without changes for a long time and is called secretory otitis (aka tubootitis, aka eustachitis).

NOTE: None of the three names are inherently incorrect, as there is no inflammation indicated by the “-itis” suffix. But since it happened historically, we will call it “secretory otitis.”

But since it happened historically, we will call it “secretory otitis”. The only symptom of secretory otitis media will be hearing loss. And no pain. Sometimes the patient feels the sound of “overflowing liquid” in the head. Parents of sick children pay attention to the fact that the child often asks again, asks to make the sound of the TV or radio louder. However, if such a child gets ARVI, the likelihood of developing acute otitis media according to the first scenario is very high.

DIAGNOSIS:

When consulting patients with hearing impairments, an endoscopic examination of the ear and nose is mandatory. In addition, they are sent to the hardware study of the organ of hearing – audiometry and impedancemetry.

Audiometry is a test that shows how much hearing is reduced at certain frequencies. The method is not very applicable for children under 8-10 years old. The patient here must give a signal to the researcher himself when he begins to hear the sound in the headphones. And a small child may be distracted or simply not understand what they want from him.

Impedancemetry – measuring the compliance of the tympanic membrane. The method is absolutely objective and does not depend on the reaction of the child. It allows you to know for sure exactly whether there is fluid behind the eardrum or not.

TREATMENT:

In the treatment of all diseases of the middle ear, the doctor must solve 3 tasks:

  1. Restore drainage through the Eustachian tube
  2. Infection control
  3. Restore the integrity of the tympanic membrane (if it is perforated).

These tasks are solved depending on each specific situation, either medically or surgically. Accordingly, the algorithm of actions can be determined only during a face-to-face consultation with a specialist. Here it is advisable to show life wisdom and not be tempted by the diagnosis and treatment regimen recommended in the pharmacy 🙂!

*** Use of site materials only with the permission of the author and a link to the source.

How to find us

Consultative reception and operations of the otolaryngologist Zaporozhchenko Pavel Aleksandrovich are carried out at the Artmedium clinic.

Clinic “Artmedium” is located at Odessa, st. Nedelina, 78

Appointment for a consultation from Monday to Saturday,
from 8.00 to 19.00, by phone:
+38 (096) 757-90-99.

Candidate of Medical Sciences, Assistant of the Department of Otorhinolaryngology, ONMedU,
otorhinolaryngologist of the highest category, allergist

    • Allergist (Immunologist) deals with the diagnosis and treatment of pathologies of the immune system, in particular bronchial asthma, urticaria and other allergic diseases.
    • An otorhinolaryngologist (ENT) specializes in diseases of the ear, nose and throat. Diagnoses and treats diseases such as otitis, sinusitis, sinusitis, pharyngitis, laryngitis, rhinitis, tonsillitis.

Graduated with honors from Odessa State Medical University and master’s degree in otorhinolaryngology (2001 degree).

15-year certificate of endoscopic diagnosis and surgery in older children.

Course of promotion:

  • Laser surgery of ENT organs, head and neck” in 2000 (m. Berlin, Nіmechchina).
  • “Endoscopic surgery of the nose and paranasal sinuses” under prof. S.Z. Piskunov in 2001 (m. Kursk, Russia).
  • “Microsurgery of the ear” – under the supervision of prof. Yu.A.Sushko and O.N. Borisenko, NDI of otorhinolaryngology (city of Kiev).

Scientific activity:

  • Double winner of the Olympiad for Young Otorhinolaryngologists under the International Academy of Otorhinolaryngology ї, Head and Neck Surgery (IAO-HNS).
  • Laureate of the award “Odesit to rock 2000” in the nomination “young students of rock”.
  • Author of 30 scientific works and 3 rational propositions.

Internship for specialty:

  • ENT department of Balcali University Clinic, Cukurova University, p.d. prof. Levent Soylu (m. Adana, Turkey) in 1999
  • ENT Department of Charite University Clinic, Oleksandr Humboldt University, prof. Volker Jahnke (m. Berlin, Nіmechchina) in 2000
  • ENT Department of the Steiglitz University Clinic, Vilniy University, prof. Hans Sherer (m. Berlin, Nіmechchina) in 2000 rotation.
  • Mayo Clinic Arizona (USA) 2012 Transoral surgery of the head and neck.
  • Austria. Graz-course of nasal endoscopy. 2013
  • Received the Kingdom of Great Britain and Irish Ireland. Cambridge. University spital. Endoscopic surgery – dissection course 2014
  • Pediatric anesthesiologist – resuscitator at the ENT Clinic of Dr. Zaporozhchenko.
  • Medical experience over 5 years.
  • He completed his internship in 2018 with a degree in Pediatric Anesthesiology.
  • Graduated from the Chisinau State Medical University. N. Testemitanu, 2001–2007
  • Passed residency “General Oncology”, in Chisinau, 2007-2009.
  • Passed the internship “Adult otolaryngology”, in Odessa, 2012–2013.
  • Specialty “Pediatric otolaryngology”, 2014 – 2018

Treatment of peri-implantitis – inflammation after implantation

With the development of implant technology, more and more patients prefer this method of restoring lost teeth, as the most reliable, aesthetic and convenient. Set and forget! This is exactly what happens in most cases, unless the plans of the doctor and his patient are violated by the most formidable complication due to which the implant can be lost – peri-implantitis. It is them who scare each other, visitors to numerous dental forums, so much so that some even refuse to think about installing an implant. Let’s figure out in this article what peri-implantitis is, how to avoid it, and, if necessary, treat it.

What is peri-implantitis?

Peri-implantitis is an inflammation of the tissues surrounding the implant and the bone, which, ultimately, without timely intensive treatment, leads to bone loss and loss of the implant.

Peri-implantitis is much easier to prevent than to stop the process that has already begun. The main cause of the disease is a bacterial infection, which can join both at the very moment of implantation, and then the symptoms of the disease appear a few days after the operation, or after some time – from several months to several years.

Why can peri-implantitis start?

According to statistics, during implantation, more than 95% of implants are successfully installed and take root. 5% of failures are due to various reasons, but in about 1% of cases, peri-implantitis is “to blame” for the loss of an implant, i.e. inflammation affects about 1 artificial tooth root out of 100. It must be understood that complications are possible with any surgical intervention. It depends on the state of health of the patient and his immune system. If the patient has disorders or there may be a decrease in immunity under the influence of adverse external factors, this must be taken into account. That is why implantation requires a preliminary thorough diagnosis by the surgeon.

Other causes of infection of the implant and the development of peri-implantitis can be:

  1. Misbehavior of the patient himself after implantation:
  • poor hygiene,
  • violation of the recommendations of the attending dentist for the care of implants,
  • increased chewing load, careless attitude to one’s artificial teeth, gum injury near the implant,
  • smoking,
  • ignoring scheduled inspections to monitor the condition of the roots of implants.
  1. Errors of specialists during implantation:
  • an unsuitable prosthetic technique was chosen,
  • implant placed in the wrong place,
  • the implant is placed in a socket with a larger diameter than required,
  • Implant load was incorrectly calculated,
  • a full diagnosis was not carried out and / or the patient’s health status was incorrectly assessed before implantation,
  • during the operation, asepsis was violated.
  1. A poor quality implant was placed. This can happen due to the fault of the dentist or at the insistence of the patient himself.
  2. The patient’s mouth had a source of infection – cavities, periodontal disease, dental plaque and stones. In order for implantation to proceed without complications, all sources of possible infection in the oral cavity must be eliminated at the preparation stage.

Why is peri-implantitis dangerous?

In addition to pain and discomfort in the affected area, the most dangerous consequence of peri-implantitis is the loss of the implant. Re-implantation will be possible only after a long process of restoration and bone grafting, because. it will be necessary to increase the volume of bone tissue.

Symptoms of peri-implantitis:

  1. The disease begins with redness, discomfort and swelling of the gums in the area of ​​the implant.
  2. There is bleeding of the gums in the problem area.
  3. At the site of inflammation, the proliferation of connective tissue begins.
  4. The gum moves away from the implant, as in periodontal disease, a periodontal pocket is formed around the titanium rod.
  5. Serous fluid and pus may be discharged from the pocket, a fistula is formed.
  6. X-ray shows marked bone loss around the implant.
  7. The implant becomes loose, the patient feels its mobility, this provokes further destruction of the bone around the titanium rod.
  8. Ultimately, if the inflammatory process is not controlled, the implant will fail.

How is peri-implantitis treated?

The success of the treatment of peri-implantitis depends on the stage of the development of the disease at which it is started: the earlier, the better the prognosis.

The treatment is directed, first of all, to the relief of inflammation in the focus and to the restoration of bone volume, with the onset of its loss. Therefore, in the treatment, 2 main stages are distinguished – the sanitation of the inflamed area and surgical bone augmentation.

  1. Before starting treatment, the doctor makes a diagnosis. The main stage of such a diagnosis will be a 3D CT scan – computed tomography to accurately determine the affected area and the state of the bone tissue.
  2. Then professional hygiene of the implant and adjacent areas is carried out – removal of soft dental deposits and tartar from the dental crown and from the subgingival space using ultrasound.
  3. Next, surgical debridement of the area of ​​inflammation is carried out – abscesses are opened. Periodontal pockets are cleaned in the same way as in periodontal diseases – with the help of special curettes or the Vector device. It is desirable that the problematic implant is not loaded.
  4. At the same time, bone grafting can be performed, using the method of guided bone regeneration using bone chips and regenerating membranes.
  5. In parallel, the patient is given local and general antibiotic therapy, antibiotics are prescribed.
  6. It is very important in the treatment of peri-implantitis to maintain daily hygiene with the use of antiseptic preparations.

The result of the treatment is necessarily controlled by repeated X-ray diagnostics.

It must be remembered that peri-implantitis is prone to frequent relapses, therefore, after treatment, monitoring the condition of the implants and increased attention to proper hygiene is MANDATORY.

Prevention of peri-implantitis

From the above it follows that peri-implantitis is much easier to prevent than to treat later. In order to protect yourself as much as possible from such a complication, both immediately after implantation and during subsequent life with an implant, you must:

  1. Carefully monitor oral hygiene, use not only a toothbrush and paste to care for your teeth and implant, but also special tools – a mono-bundle brush, a toothbrush, special dental floss and an irrigator.