Manuel Ruiz Profile picture
๐Ÿ”ฌDiving deep into #MECFS, #LongCOVID, #LongEBV & post-vaccination syndrome research. ๐Ÿฆ Dual role:Researcher and EBV ME/CFS patient. Seeking answers #Antivirals
Jul 9 โ€ข 4 tweets โ€ข 16 min read
๐ŸŒŸ ๐๐ž๐ฐ ๐š๐ซ๐ญ๐ข๐œ๐ฅ๐ž: ๐ญ๐ก๐ž ๐œ๐จ๐ง๐ง๐ž๐œ๐ญ๐ข๐จ๐ง ๐›๐ž๐ญ๐ฐ๐ž๐ž๐ง ๐‹๐จ๐ง๐  ๐‚๐Ž๐•๐ˆ๐ƒ, ๐Œ๐ฒ๐š๐ฅ๐ ๐ข๐œ ๐„๐ง๐œ๐ž๐ฉ๐ก๐š๐ฅ๐จ๐ฆ๐ฒ๐ž๐ฅ๐ข๐ญ๐ข๐ฌ ๐š๐ง๐ ๐ฉ๐จ๐ฌ๐ญ-๐ฏ๐š๐œ๐œ๐ข๐ง๐š๐ฅ ๐ฌ๐ฒ๐ง๐๐ซ๐จ๐ฆ๐ž๐ฌ ๐ฅ๐ข๐ž๐ฌ ๐ข๐ง ๐ญ๐ก๐ž ๐๐ž๐ฏ๐ž๐ฅ๐จ๐ฉ๐ฆ๐ž๐ง๐ญ ๐จ๐Ÿ ๐š๐ง ๐€๐ฎ๐ญ๐จ๐ข๐ฆ๐ฆ๐ฎ๐ง๐ž ๐‡๐ฒ๐ฉ๐จ๐œ๐จ๐ซ๐ญ๐ข๐ฌ๐จ๐ฅ๐ž๐ฆ๐ข๐œ ๐’๐ฒ๐ง๐๐ซ๐จ๐ฆ๐ž๐ŸŒŸ

๐ŸŒ I am excited to share with you our latest paper entitled "๐‡๐ฒ๐ฉ๐จ๐œ๐จ๐ซ๐ญ๐ข๐ฌ๐จ๐ฅ๐ž๐ฆ๐ข๐œ ๐€๐’๐ˆ๐€: ๐€ ๐ฏ๐š๐œ๐œ๐ข๐ง๐ž- ๐š๐ง๐ ๐œ๐ก๐ซ๐จ๐ง๐ข๐œ ๐ข๐ง๐Ÿ๐ž๐œ๐ญ๐ข๐จ๐ง-๐ข๐ง๐๐ฎ๐œ๐ž๐ ๐ฌ๐ฒ๐ง๐๐ซ๐จ๐ฆ๐ž ๐›๐ž๐ก๐ข๐ง๐ ๐ญ๐ก๐ž ๐จ๐ซ๐ข๐ ๐ข๐ง ๐จ๐Ÿ ๐ฅ๐จ๐ง๐  ๐‚๐Ž๐•๐ˆ๐ƒ ๐š๐ง๐ ๐ฆ๐ฒ๐š๐ฅ๐ ๐ข๐œ ๐ž๐ง๐œ๐ž๐ฉ๐ก๐š๐ฅ๐จ๐ฆ๐ฒ๐ž๐ฅ๐ข๐ญ๐ข๐ฌ". In this paper, we explain the links between Long COVID, Chronic Fatigue Syndrome/Myalgic Encephalomyelitis (ME/CFS) and COVID-19 post-vaccine syndromes. ๐Ÿค” Stay reading to the end and you will also find our treatment proposal that could improve symptoms.๐Ÿ’Š

โžก๏ธ๐‹๐ข๐ง๐ค ๐จ๐Ÿ ๐จ๐ฎ๐ซ ๐ซ๐ž๐ฏ๐ข๐ž๐ฐ ๐š๐ซ๐ญ๐ข๐œ๐ฅ๐ž:

๐Ÿ” ๐€๐›๐ฌ๐ญ๐ซ๐š๐œ๐ญ: We present a model for the development of these diseases that involves a complex interplay between immune hyperactivation, autoimmune hypophysitis or pituitary hypofunction, and immune exhaustion. We believe that the starting point is a deficient CD4 T-cell response to viral infections in genetically predisposed individuals (HLA-DRB1). This would follow from an uncontrolled immune response with hyperactivation of CD8 T cells and elevated antibody production, some of which could be directed against self-antigens, triggering autoimmune hypophysitis or direct damage to the pituitary, resulting in decreased production of pituitary hormones, such as ACTH. ๐Ÿงฌ

๐Ÿ”ฌ ๐–๐ก๐š๐ญ'๐ฌ ๐ญ๐ก๐ž ๐›๐ข๐  ๐๐ž๐š๐ฅ?
1๏ธโƒฃ ๐‘๐ž๐ฅ๐š๐ญ๐ข๐จ๐ง๐ฌ๐ก๐ข๐ฉ ๐ญ๐จ ๐€๐’๐ˆ๐€ ๐’๐ฒ๐ง๐๐ซ๐จ๐ฆ๐ž: We propose that Long COVID, ME/CFS and post-vaccine COVID-19 syndrome could be included in adjuvant-induced autoimmune/inflammatory syndrome (ASIA) due to their similar clinical manifestations and possible relationship to genetic factors, such as polymorphisms in the HLA-DRB1 gene.
2๏ธโƒฃ ๐ƒ๐ž๐ฏ๐ž๐ฅ๐จ๐ฉ๐ฆ๐ž๐ง๐ญ๐š๐ฅ ๐Œ๐จ๐๐ž๐ฅ: We suggest that these diseases begin with a deficient immune response and progress to uncontrolled immune hyperactivation, followed by immune exhaustion, exacerbating symptoms and pathology.
3๏ธโƒฃ ๐‡๐ฒ๐ฉ๐จ๐œ๐จ๐ซ๐ญ๐ข๐ฌ๐จ๐ฅ๐ž๐ฆ๐ข๐š: We highlight the decrease in ACTH production and its impact on immune function and clinical symptoms, establishing a direct link with pituitary dysfunction.
4๏ธโƒฃ ๐“๐ซ๐ž๐š๐ญ๐ฆ๐ž๐ง๐ญ ๐๐ซ๐จ๐ฉ๐จ๐ฌ๐š๐ฅ: We propose a treatment approach including antivirals, corticosteroids/ginseng, antioxidants and metabolic precursors to improve symptoms by modulating immune response, pituitary function, inflammation and oxidative stress.

๐Ÿ’ก ๐ˆ๐ฆ๐ฉ๐ฅ๐ข๐œ๐š๐ญ๐ข๐จ๐ง๐ฌ ๐š๐ง๐ ๐‚๐จ๐ง๐œ๐ฅ๐ฎ๐ฌ๐ข๐จ๐ง๐ฌ:
๐Ÿ”นThese disorders could have an autoimmune origin against the adenohypophysis.
๐Ÿ”นTreatment with antivirals and corticosteroid replacement therapy in patients with permanent pituitary damage could improve symptoms by addressing immune and hormonal dysfunction.

๐Ÿง  ๐“๐ก๐ž ๐ค๐ž๐ฒ ๐ข๐ฌ ๐ฉ๐ข๐ญ๐ฎ๐ข๐ญ๐š๐ซ๐ฒ ๐๐š๐ฆ๐š๐ ๐ž: ๐ก๐จ๐ฐ ๐๐จ๐ž๐ฌ ๐ญ๐ก๐ข๐ฌ ๐ซ๐ž๐ฅ๐š๐ญ๐ž ๐ญ๐จ ๐ญ๐ก๐ž ๐๐ž๐ฏ๐ž๐ฅ๐จ๐ฉ๐ฆ๐ž๐ง๐ญ ๐จ๐Ÿ ๐Œ๐„/๐‚๐…๐’, ๐‹๐จ๐ง๐  ๐‚๐Ž๐•๐ˆ๐ƒ ๐š๐ง๐ ๐จ๐ญ๐ก๐ž๐ซ ๐ฉ๐จ๐ฌ๐ญ-๐ฏ๐ข๐ซ๐š๐ฅ ๐š๐ง๐ ๐ฉ๐จ๐ฌ๐ญ-๐ฏ๐š๐œ๐œ๐ข๐ง๐ž ๐ฌ๐ฒ๐ง๐๐ซ๐จ๐ฆ๐ž๐ฌ?

๐Ÿ’‰ Certain viruses (and other pathogens) and vaccines can affect the pituitary gland, interfering with cortisol production and triggering a cascade of complex symptoms. In patients with weak HLA-DRB1 alleles, such as DR15, immune hyperactivation can trigger an autoimmune response against ACTH, crucial for cortisol production. This is exactly analogous to how other autoimmune diseases such as multiple sclerosis or lupus develop, where the immune system attacks other antigens in the body, but in the syndromes we are discussing, the autoimmunity is specifically directed against pituitary ACTH.

๐Ÿฆ  This link explains why patients with chronic infections often experience persistent hypocortisolemia, as the pathogen continues to produce ACTH-mimicking antigens, maintaining the active autoimmune response or generates direct pituitary damage. In contrast, patients without chronic infections and with the same weak alleles treated with immune checkpoint inhibitors (ICIs) may develop temporary hypophysitis and similar cortisol deficits, but discontinuation of treatment usually allows recovery.

This also explains why patients experience chronic fatigue, dysautonomia, orthostatic intolerance, exercise intolerance, intolerance to stressful events and mild hypoglycemia due to low cortisol. Cortisol is crucial in providing the body with needed energy and regulating the stress response. When cortisol levels are low, as they are in these syndromes, the body cannot respond effectively to physical and emotional demands.

๐ŸฉธCortisol plays a crucial role in maintaining stable blood sugar levels by promoting gluconeogenesis (glucose production) and stimulating the release of stored glucose in the form of glycogen in the liver. When there is insufficient cortisol, the body faces difficulties in increasing glucose levels in demand situations, such as during physical exercise or in response to stress, which can lead to episodes of mild hypoglycemia. In times of fright or anger, adrenaline release may temporarily improve symptoms by temporarily increasing glucose availability, briefly compensating for cortisol deficiency. However, this response does not adequately replace the long-term regulatory functions of cortisol, so symptoms may return once adrenaline subsides.

๐Ÿ‹๏ธโ€โ™‚๏ธ In the case of physical exercise, which naturally increases cortisol levels to mobilize energy and respond to physical exertion, the lack of this hormone limits the body's ability to maintain sustained physical activity. Patients may experience rapid muscle fatigue, feelings of weakness and slower recovery after exercise.

๐Ÿ˜ฐ As for stressful events (exams, travel, surgical operations, etc) , cortisol also plays a crucial role in the body's response to emotional or physical stress. When cortisol levels are insufficient, the body has difficulty handling stressful situations effectively. This can manifest itself in an exacerbation of existing symptoms, such as intense fatigue, dizziness, difficulty concentrating and a generalized feeling of malaise.

โžก๏ธ For years, many of these patients have been misunderstood and mislabeled as having psychosomatic illness. This is because their symptoms tend to worsen during periods of stress, which has led to the suggestion that the origin of their problems lies in psychological factors. However, the reality is that these patients are not experiencing symptoms due to an underlying psychological disorder, but as a direct result of insufficient cortisol. The lack of this vital hormone prevents the body from adapting and responding appropriately to stress, which perpetuates and aggravates their physical symptoms.

๐Ÿšจ ๐“๐ก๐ž ๐ƒ๐ž๐ฏ๐ž๐ฅ๐จ๐ฉ๐ฆ๐ž๐ง๐ญ ๐จ๐Ÿ ๐€๐ฎ๐ญ๐จ๐ข๐ฆ๐ฆ๐ฎ๐ง๐ข๐ญ๐ฒ ๐ญ๐จ ๐€๐‚๐“๐‡: ๐€ ๐๐ซ๐จ๐œ๐ž๐ฌ๐ฌ ๐’๐ข๐ฆ๐ข๐ฅ๐š๐ซ ๐ญ๐จ ๐Ž๐ญ๐ก๐ž๐ซ ๐€๐ฎ๐ญ๐จ๐ข๐ฆ๐ฆ๐ฎ๐ง๐ž ๐ƒ๐ข๐ฌ๐ž๐š๐ฌ๐ž๐ฌ ๐ฌ๐ฎ๐œ๐ก ๐š๐ฌ ๐Œ๐ฎ๐ฅ๐ญ๐ข๐ฉ๐ฅ๐ž ๐’๐œ๐ฅ๐ž๐ซ๐จ๐ฌ๐ข๐ฌ ๐Ÿšจ

This same mechanism occurs in other autoimmune diseases. Some HLA-II alleles, such as the DR15 variant, are associated with an impaired ability to recognize cells infected with certain pathogens, such as Epstein-Barr virus (EBV). In multiple sclerosis this poor recognition ability specifically affects CD4 T cells, which are crucial for coordinating the immune response. When CD4 T cells cannot correctly recognize infected cells, this leads to hyperactivation of CD8 T cells and an increase in antibodies against the pathogen to compensate for the deficient CD4 T cell response. Without the coordinated help of CD4 T cells, CD8 T cells cannot eliminate all EBV-infected cells, thus never effectively eliminating or controlling the infection and resulting in chronic infection. This results in an increase of infected cells, an exhaustion of CD8 T cells and an increased risk of developing autoimmune diseases, since CD4 T cells, by misrecognizing these viral antigens presented on the HLA-II antigen-presenting cells, can confuse them with the body's own proteins, generating an autoimmune disease. In multiple sclerosis, autoimmunity develops when the EBNA-1 antigen of the Epstein-Barr virus is mistaken for myelin, due to a similar amino acid sequence and molecular mimicry in patients with DR15 alleles. The same could occur in patients with Long COVID, myalgic encephalomyelitis and post-vaccinal syndromes, where autoimmunity against ACTH develops.

#LongCovid #MECFS #Vaccinesfrontiersin.org/journals/immunโ€ฆImage ๐Ÿ“ข ๐–๐ก๐ฒ ๐•๐š๐œ๐œ๐ข๐ง๐ž๐ฌ ๐‚๐š๐ง ๐ƒ๐ž๐ฏ๐ž๐ฅ๐จ๐ฉ ๐๐š๐ญ๐ก๐จ๐ฅ๐จ๐ ๐ข๐ž๐ฌ ๐’๐ข๐ฆ๐ข๐ฅ๐š๐ซ ๐ญ๐จ ๐๐ž๐ซ๐ฌ๐ข๐ฌ๐ญ๐ž๐ง๐ญ ๐‚๐Ž๐•๐ˆ๐ƒ ๐š๐ง๐ ๐๐จ๐ฌ๐ญ-๐ˆ๐ง๐Ÿ๐ž๐œ๐ญ๐ข๐จ๐ฎ๐ฌ ๐Œ๐„/๐‚๐…๐’? ๐Ÿ“ข

๐Ÿ”ฌ ๐‚๐จ๐ฆ๐ฆ๐จ๐ง ๐Œ๐ž๐œ๐ก๐š๐ง๐ข๐ฌ๐ฆ: ๐”๐ง๐œ๐จ๐ง๐ญ๐ซ๐จ๐ฅ๐ฅ๐ž๐ ๐ˆ๐ฆ๐ฆ๐ฎ๐ง๐ž ๐€๐œ๐ญ๐ข๐ฏ๐š๐ญ๐ข๐จ๐ง.
Both persistent COVID and post-infectious ME/CFS and post-vaccine syndrome share a common basis: an overactive immune response. In our study, we propose the presence of:

1๏ธโƒฃ ๐๐ž๐ซ๐ฌ๐ข๐ฌ๐ญ๐ž๐ง๐œ๐ž ๐จ๐Ÿ ๐•๐ข๐ซ๐š๐ฅ ๐€๐ง๐ญ๐ข๐ ๐ž๐ง๐ฌ ๐จ๐ซ ๐€๐๐ฃ๐ฎ๐ฏ๐š๐ง๐ญ๐ฌ:
โ–ช๏ธ ๐‚๐ก๐ซ๐จ๐ง๐ข๐œ ๐ฏ๐ข๐ซ๐š๐ฅ ๐ข๐ง๐Ÿ๐ž๐œ๐ญ๐ข๐จ๐ง: after a viral infection, such as SARS-CoV-2 or Epstein-Barr virus (EBV), viral antigens may remain in the body, triggering a continuous immune response.
โ–ช๏ธ ๐•๐š๐œ๐œ๐ข๐ง๐š๐ญ๐ข๐จ๐ง: Vaccines contain adjuvants designed to stimulate the immune system. In genetically predisposed individuals (HLA-DRB1), this stimulation may be excessive, leading to uncontrolled immune activation similar to a viral infection. In addition, the introduced viral antigens could have molecular mimicry with ACTH, exacerbating the immune response and potentially contributing to the development of autoimmunity against ACTH.

2๏ธโƒฃ ๐€๐ฎ๐ญ๐จ๐ข๐ฆ๐ฆ๐ฎ๐ง๐ข๐ญ๐ฒ ๐€๐ ๐š๐ข๐ง๐ฌ๐ญ ๐ญ๐ก๐ž ๐€๐๐ž๐ง๐จ๐ก๐ฒ๐ฉ๐จ๐ฉ๐ก๐ฒ๐ฌ๐ข๐ฌ:
* Both conditions can lead to autoimmune inflammation of the adenohypophysis (autoimmune hypophysitis), resulting in hormonal dysfunction, especially ACTH production.

3๏ธโƒฃ ๐‡๐ฒ๐ฉ๐ž๐ซ๐œ๐จ๐ซ๐ญ๐ข๐ฌ๐จ๐ฅ๐ž๐ฆ๐ข๐š ๐š๐ง๐ ๐‡๐๐€ ๐€๐ฑ๐ข๐ฌ ๐‡๐ฒ๐ฉ๐จ๐Ÿ๐ฎ๐ง๐œ๐ญ๐ข๐จ๐ง:
* Chronic inflammation and proinflammatory cytokine production can alter the hypothalamic-pituitary-adrenal (HPA) axis, leading to low cortisol levels, which exacerbate fatigue and other symptoms.

๐Ÿ”„ ๐‚๐ฒ๐œ๐ฅ๐ž ๐จ๐Ÿ ๐‡๐ฒ๐ฉ๐ž๐ซ๐š๐œ๐ญ๐ข๐ฏ๐š๐ญ๐ข๐จ๐ง ๐š๐ง๐ ๐ˆ๐ฆ๐ฆ๐ฎ๐ง๐ž ๐ƒ๐ž๐ฉ๐ฅ๐ž๐ญ๐ข๐จ๐ง:
โ–ช๏ธ ๐ˆ๐ง๐ข๐ญ๐ข๐š๐ฅ ๐‡๐ฒ๐ฉ๐ž๐ซ๐š๐œ๐ญ๐ข๐ฏ๐š๐ญ๐ข๐จ๐ง: Initial immune response is excessive, with high cytokine production and CD8 T-cell activation.
โ–ช๏ธ ๐ˆ๐ฆ๐ฆ๐ฎ๐ง๐ž ๐ž๐ฑ๐ก๐š๐ฎ๐ฌ๐ญ๐ข๐จ๐ง: Over time, this hyperactivation leads to T-cell exhaustion, decreasing the body's ability to control infection and exacerbating the pathology.
โ–ช๏ธ ๐‡๐ฒ๐ฉ๐จ๐œ๐จ๐ซ๐ญ๐ข๐ฌ๐จ๐ฅ๐ข๐ฌ๐ฆ: Insufficient cortisol maintains the state of immune hyperactivation, perpetuating inflammation and hindering resolution of the immune cycle.

๐Ÿ“Š ๐„๐ฏ๐ข๐๐ž๐ง๐œ๐ž ๐š๐ง๐ ๐‚๐ฅ๐ข๐ง๐ข๐œ๐š๐ฅ ๐’๐ข๐ฆ๐ข๐ฅ๐š๐ซ๐ข๐ญ๐ข๐ž๐ฌ:
โ–ช๏ธ ๐’๐ก๐š๐ซ๐ž๐ ๐’๐ฒ๐ฆ๐ฉ๐ญ๐จ๐ฆ๐ฌ: chronic fatigue, muscle and joint pain, sleep disturbances, cognitive problems, and post-exertional malaise.
โ–ช๏ธ ๐‚๐จ๐ฆ๐ฆ๐จ๐ง ๐†๐ž๐ง๐ž๐ญ๐ข๐œ: Polymorphisms in the HLA-DRB1 gene predispose to an exaggerated immune response to both viral infections and vaccine components.

๐Ÿ” ๐‚๐จ๐ง๐œ๐ฅ๐ฎ๐ฌ๐ข๐จ๐ง: Our review suggests that both Long COVID and post-infectious ME/CFS and post-vaccination syndromes could be considered as part of the autoimmune/autoinflammatory syndrome induced by adjuvants (ASIA). These findings underline the need for a personalized therapeutic approach that takes into account the genetic predisposition and immune status of the patient.

๐ŸŒ๐Ÿ’‰ ๐‡๐จ๐ฐ ๐‚๐š๐ง ๐’๐จ๐ฆ๐ž ๐•๐š๐œ๐œ๐ข๐ง๐ž๐ฌ ๐“๐ซ๐ข๐ ๐ ๐ž๐ซ ๐๐ž๐ซ๐ฌ๐ข๐ฌ๐ญ๐ž๐ง๐ญ ๐‡๐ž๐š๐ฅ๐ญ๐ก ๐๐ซ๐จ๐›๐ฅ๐ž๐ฆ๐ฌ? ๐Ÿ’‰๐ŸŒ
1๏ธโƒฃ ๐Œ๐ž๐œ๐ก๐š๐ง๐ข๐ฌ๐ฆ ๐จ๐Ÿ ๐€๐œ๐ญ๐ข๐จ๐ง:
โ–ช๏ธ Vaccines contain adjuvants and antigens designed to stimulate robust immune responses.
โ–ช๏ธ In some cases, this stimulation can lead to a dysregulated autoimmune response, similar to that seen in diseases such as systemic lupus erythematosus.

2๏ธโƒฃ ๐•๐š๐œ๐œ๐ข๐ง๐ž๐ฌ ๐ˆ๐ง๐ฏ๐จ๐ฅ๐ฏ๐ž๐:
โ–ช๏ธ COVID-19: Cases of prolonged symptoms after infection or vaccination, known as Long COVID or post-COVID syndrome, have been reported.
โ–ช๏ธ Hepatitis B and C, HPV: Some individuals develop ME/CFS after vaccination, related to autoimmunity and immune alterations.

3๏ธโƒฃ ๐‘๐ข๐ฌ๐ค ๐…๐š๐œ๐ญ๐จ๐ซ๐ฌ:
โ–ช๏ธ Genetic predisposition such as polymorphisms in the HLA-DRB1 gene may increase susceptibility to abnormal immune responses.
โ–ช๏ธ Viral persistence or chronic antigen exposure may trigger a continuous and pathologic immune response.

๐ŸŒ๐Ÿ’‰ ๐ƒ๐ข๐ ๐ฒ๐จ๐ฎ ๐ค๐ง๐จ๐ฐ ๐ญ๐ก๐š๐ญ ๐ฏ๐š๐œ๐œ๐ข๐ง๐ž๐ฌ ๐๐ž๐ฌ๐ข๐ ๐ง๐ž๐ ๐ฐ๐ข๐ญ๐ก ๐š๐๐ž๐ง๐จ๐ฏ๐ข๐ซ๐ฎ๐ฌ๐ž๐ฌ ๐ฆ๐š๐ฒ ๐ก๐š๐ฏ๐ž ๐š ๐ฌ๐ฅ๐ข๐ ๐ก๐ญ๐ฅ๐ฒ ๐ข๐ง๐œ๐ซ๐ž๐š๐ฌ๐ž๐ ๐ซ๐ข๐ฌ๐ค ๐จ๐Ÿ ๐ญ๐ซ๐ข๐ ๐ ๐ž๐ซ๐ข๐ง๐  ๐ฉ๐ซ๐จ๐ฅ๐จ๐ง๐ ๐ž๐ ๐ข๐ฆ๐ฆ๐ฎ๐ง๐ž ๐ซ๐ž๐ฌ๐ฉ๐จ๐ง๐ฌ๐ž๐ฌ ๐œ๐จ๐ฆ๐ฉ๐š๐ซ๐ž๐ ๐ญ๐จ ๐ฆ๐ž๐ฌ๐ฌ๐ž๐ง๐ ๐ž๐ซ ๐‘๐๐€ ๐ฏ๐š๐œ๐œ๐ข๐ง๐ž๐ฌ?

Adenoviruses used as vectors may persist longer in the body, exposing the immune system to viral components for extended periods.

๐Ÿฆ Adenoviruses are linear double-stranded DNA viruses and remain in an episomal condition in the nucleus. The mechanism of adenovirus vector persistence has not yet been elucidated. But they can persist for a long time in the organism. It is thought that it could be by their integration into the genome or by episomal gene duplication facilitated by the transcription machinery of the host cells. That is, it could happen that it transmits episomes to its cells as occurs in Epstein-Barr virus without lytic phase. During mitosis, EBV episomes are bound to chromosomes in metaphase through EBNA-1, allowing efficient segregation of episomes into daughter cells.

๐Ÿ”ฌ This theoretical phenomenon could predispose certain genetically susceptible individuals to develop chronic autoimmune or inflammatory disorders, as discussed in the context of ASIA syndrome. In contrast, messenger RNA vaccines, by rapidly degrading after inducing the desired immune response, limit this prolonged exposure.

๐Ÿ’‰โš ๏ธ Regardless of the type of vaccine against COVID-19 or other pathologies, whether messenger RNA or adenoviral vector, there is a theoretical possible risk of developing autoimmune disease only in individuals with certain genetic alleles, such as HLA-DRB1. The key lies in prolonged exposure to viral antigens, which could trigger persistent autoimmune responses.

๐Ÿ”ฌ mRNA vaccines are characterized by rapidly degrading after inducing the desired immune response, thus limiting prolonged exposure to viral components. In contrast, adenoviral vector-based vaccines can persist in the body longer, potentially increasing the risk of a longer-lasting autoimmune reaction in susceptible individuals.

๐Ÿ’ก It is crucial to remember that these risks are rare when compared to the total number of vaccinated individuals and that all vaccines, including adenovirus and messenger RNA vaccines, are critical to combat pandemic diseases such as COVID-19 at the species level. However, understanding these differences may help improve future vaccine design and surveillance for potential adverse effects in these susceptible individuals.

๐ŸŒŸ Recall that vaccines save millions of lives and prevent serious consequences of infections in the majority of the population. However, it is crucial to recognize that they may also present risks in some susceptible individuals.

๐Ÿงฌ๐Ÿ’‰ HLA-DRB1 genetic alleles play a crucial role in predisposition to diseases such as Long COVID, myalgic encephalomyelitis (ME/CFS) and ASIA syndrome, as well as in post-vaccine syndromes.

๐Ÿ” These HLA-DRB1 alleles are part of the HLA (human leukocyte antigen) system, which regulates how the immune system recognizes and responds to foreign antigens, such as viruses and vaccine components. In individuals with certain HLA-DRB1 alleles, the immune system may trigger excessive or inappropriate responses to these antigens, leading to autoimmune conditions and inflammatory syndromes.

๐Ÿฆ  This explains why some patients, after significant viral infection or vaccination, may develop persistent autoimmune reactions. In the case of Long COVID and ME/CFS, persistence of viral antigens could trigger an autoimmune response against self tissues, including the pituitary gland.

๐Ÿ”โœจ In a ๐ค๐ž๐ฒ ๐ฌ๐ญ๐ฎ๐๐ฒ, patients who developed hypophysitis after infection with the first SARS-CoV recovered their health months after treatment with replacement corticosteroids. This finding underscores the importance of detecting hypophysitis early to prevent permanent damage.โžก๏ธ

๐Ÿง  Hypophysitis can cause a decrease in ACTH production, crucial for adrenal function and immune response. Controlling ACTH levels is critical if symptoms such as chronic fatigue, orthostatic hypotension and sleep disturbances are present after a viral infection.

๐Ÿ’Š Early administration of replacement corticosteroids can stop ACTH secretion and prevent autoimmunity against this hormone, thus protecting the pituitary from further damage. Over time, as hypophysitis and viral infection subside, the dose of replacement corticosteroids can be gradually reduced until normal pituitary function is restored.

๐Ÿ”Challenges in HPA Axis Evaluation in Patients with Long COVID, ME/CFS and Post-Vaccine Syndromes.
Studies are often limited to single morning cortisol measurements, ignoring diurnal variability and cortisol response to physical/mental stress, which may underestimate problems in the HPA axis. In addition, CRH or ACTH stimulation tests, crucial for assessing pituitary and adrenal function, are not frequently performed.

It is crucial to detect HPA axis dysfunctions as early as possible to prevent adrenal atrophy due to low ACTH levels. Newly diagnosed patients may recover HPA axis function if autoimmune or direct damage by infection to the pituitary is detected early. Conversely, as time passes, the risk of pituitary damage from autoimmunity against ACTH increases, leading to further adrenal atrophy and the need for hydrocortisone replacement therapy. Early detection and treatment of these dysfunctions is crucial to improve the clinical management and quality of life of these patients.

#Research #Health #Autoimmunityncbi.nlm.nih.gov/pmc/articles/Pโ€ฆImage
Mar 28 โ€ข 18 tweets โ€ข 3 min read
๐Ÿ”ดNuevo artรญculo confirma lo que hemos discutido: ยก๐ฅ๐จ๐ฌ ๐ญ๐ž๐ฌ๐ญ ๐๐ž ๐ฆ๐ข๐œ๐ซ๐จ๐›๐ข๐จ๐ญ๐š ๐ฌ๐จ๐ง ๐ข๐ง๐ฎฬ๐ญ๐ข๐ฅ๐ž๐ฌ! El mundo de la microbiota estรก siendo explotado como una estafa, especialmente entre pacientes desesperados por resolver problemas intestinales. Aquรญ te explico: Image 1/La diversidad de microbiota varรญa entre personas, lo que hace imposible obtener resultados consistentes de los test de microbiota. No se sabe cuรกl es la microbiota normal. #Microbiota #SIBO #LongEBV #microE2324
Oct 2, 2023 โ€ข 6 tweets โ€ข 19 min read
๐Ÿฆ  ๐Ÿ” ๐ƒ๐ข๐ ๐ฒ๐จ๐ฎ ๐ค๐ง๐จ๐ฐ ๐ญ๐ก๐š๐ญ ๐„๐ฌ๐ฉ๐ญ๐ž๐ข๐ง-๐๐š๐ซ๐ซ ๐ฏ๐ข๐ซ๐ฎ๐ฌ ๐œ๐จ๐ฎ๐ฅ๐ ๐›๐ž ๐š ๐ญ๐ซ๐ข๐ ๐ ๐ž๐ซ ๐Ÿ๐จ๐ซ ๐ฌ๐ฒ๐ฆ๐ฉ๐ญ๐จ๐ฆ๐ฌ ๐ข๐ง ๐Œ๐„/๐‚๐…๐’ ๐š๐ง๐ ๐‹๐จ๐ง๐  ๐‚๐Ž๐•๐ˆ๐ƒ?
A thread ๐Ÿงต

Below, I summarize the points of our recent published review and how it could explain the development of ๐๐ฒ๐ฌ๐š๐ฎ๐ญ๐จ๐ง๐จ๐ฆ๐ข๐š, ๐ฆ๐ข๐œ๐ซ๐จ๐œ๐ฅ๐จ๐ญ๐ฌ, ๐š๐ฎ๐ญ๐จ๐ข๐ฆ๐ฆ๐ฎ๐ง๐ž ๐๐ข๐ฌ๐ž๐š๐ฌ๐ž๐ฌ, ๐ž๐ฑ๐ž๐ซ๐œ๐ข๐ฌ๐ž ๐ข๐ง๐ญ๐จ๐ฅ๐ž๐ซ๐š๐ง๐œ๐ž, ๐ฆ๐ข๐ญ๐จ๐œ๐ก๐จ๐ง๐๐ซ๐ข๐š๐ฅ ๐๐ฒ๐ฌ๐Ÿ๐ฎ๐ง๐œ๐ญ๐ข๐จ๐ง, ๐ก๐ฒ๐ฉ๐จ๐œ๐จ๐ซ๐ญ๐ข๐ฌ๐จ๐ฅ๐ข๐ฌ๐ฆ ๐š๐ง๐ ๐ฆ๐จ๐ซ๐ž ๐œ๐จ๐ง๐ฌ๐ž๐ช๐ฎ๐ž๐ง๐œ๐ž๐ฌ. Share with other patients so they can understand their illness. ๐Š๐ง๐จ๐ฐ๐ฅ๐ž๐๐ ๐ž ๐ข๐ฌ ๐ฉ๐จ๐ฐ๐ž๐ซ! ๐Ÿ“š

Many people wonder why EBV, the Epstein-Barr virus, and not another virus, is the main trigger of the symptomatic picture of ME/CFS and Long COVID, when there are other factors, such as different infections, metal intoxication, among others. In the following lines, I will explain the fundamentals of this phenomenon, summarizing the essential points of the review article we recently published.

The common denominator of all the triggering factors of both diseases is their capacity to immunosuppress. Any intracellular infection, whether bacterial, viral or parasitic, that is not controlled, leads to immunosuppression due to chronic exposure to antigens. Usually, the ability to control an infection is due to genetic factors, particularly the HLA genes (I and II). These genes encode proteins called human leukocyte antigen (HLA), which are essential for distinguishing self from non-self in our body. If this "alert" does not function properly, the immune system cannot efficiently eliminate pathogens. HLAs are like a scanner that allows us to identify things that are not from the body and respond to them. If this scanner is not working properly and does not recognize something that is bad, it would prevent your immune system from eliminating it and therefore prevent it from moving freely through your body without being eliminated.
In the context of ME/CFS, there are ๐ญ๐ฐ๐จ ๐ฉ๐จ๐ฌ๐ฌ๐ข๐›๐ฅ๐ž ๐ฌ๐œ๐ž๐ง๐š๐ซ๐ข๐จ๐ฌ: the individual may have a direct genetic predisposition to EBV, or they may lose control over EBV due to an underlying immune deficiency caused by another infection or exposure to chemicals or metals. If it is another infection, we would also be talking about a "genetic weakness" (HLA) to those specific pathogens.
If the individual is genetically susceptible to EBV, it is because he or she has old HLA-II haplotypes, to which EBV has co-evolved. This is mainly because EBV infects by binding to HLA-II proteins in cells. This virus is cunning: when it infects a cell, it introduces its DNA into it latently without generating new virions, avoiding detection by the immune system and using B cells as "Trojan horses".

While 95% of the world's population has EBV, only a minority develop problems. This is where the "weak" HLA-II haplotypes against EBV come into play again.
Although it is said that the majority of the population does not have problems with this virus, this is not entirely true. Think that this virus takes advantage of every time you become immunosuppressed for any reason. An example is its brother herpes labialis, that every time that person is immunosuppressed for any reason, it takes advantage and infects more cells, making the lesion visible on the lips. But every time the immune system recovers from the first event that immunosuppressed him, this virus is controlled again and the lesion disappears. This is exactly the same thing that happens in healthy people who are able to control EBV.

The main difference between these healthy people and ME/CFS patients who have problems with EBV, is that by having strong HLA-II haplotypes against EBV they are able to recognize all EBV latency types and control them. In contrast, ๐ฉ๐š๐ญ๐ข๐ž๐ง๐ญ๐ฌ ๐ฐ๐ข๐ญ๐ก ๐„๐๐• ๐Œ๐„/๐‚๐…๐’, ๐ก๐š๐ฏ๐ข๐ง๐  ๐ฐ๐ž๐š๐ค ๐ก๐š๐ฉ๐ฅ๐จ๐ญ๐ฒ๐ฉ๐ž๐ฌ ๐š๐ ๐š๐ข๐ง๐ฌ๐ญ ๐„๐๐•, ๐ญ๐ก๐ž๐ข๐ซ ๐“๐‚๐ƒ๐Ÿ’ ๐œ๐ž๐ฅ๐ฅ๐ฌ ๐š๐ซ๐ž ๐ง๐จ๐ญ ๐š๐›๐ฅ๐ž ๐ญ๐จ ๐ซ๐ž๐œ๐จ๐ ๐ง๐ข๐ณ๐ž ๐„๐๐• ๐ฅ๐š๐ญ๐ž๐ง๐œ๐ฒ ๐ˆ ๐œ๐ž๐ฅ๐ฅ๐ฌ. The rest of the latency types are able to be recognized since they are controlled by TCD8 cells, as they would not have weak HLA-I haplotypes against EBV.

Let us explain this better. TCD4 cells recognize antigens presented on HLA-II proteins and TCD8 cells recognize antigens presented on HLA-I proteins. Normally, any intracellular infection is controlled by TCD8 cells because the infected cells present on the HLA-I proteins of their membrane the antigens of the pathogen inside. On the other hand, HLA-II proteins are found mostly on antigen-presenting cells that take antigens from outside the cell and present them on their HLA-II proteins so that they are recognized by T-CD4 cells. So, we would think that EBV, being an intracellular pathogen, its antigens should always be presented on the HLA-I proteins of the infected cell. This is not always the case; this virus has evolved to evade this system by generating latency. One of the evasion mechanisms to remain unrecognized is to prevent a latency antigen, called EBNA-1, from being presented on HLA-I proteins and from being presented on HLA-II proteins. This is of utmost importance for the virus because it prevents, for example, latency I cells (only expressing EBNA-1 from the virus) from being recognized by TCD8 cells. On the other hand, the rest of the latency cells (II and III) and lytic cells without would be recognized and eliminated by CD8 T cells. So we would think that no one would be able to control the latency I cells if they evade CD8 T cells. Here our immune system is also intelligent and this is where CD4 T cells play the differential role between healthy and sick people with this virus. The importance of HLA-II haplotypes reappears. Those individuals with EBV-resistant HLA-II haplotypes will be able to present the EBNA-1 antigen well on latency I cells and will be recognized and eliminated by TCD4 cells. In contrast, those with EBV-weak HLA-II haplotypes will not be able to present EBNA-1 well on HLA-II proteins and therefore CD4 T cells will not recognize the latency I cells. These latency I cells, when left unchecked, will multiply and cause inflammation and damage. But every time they go to another type of latency or lytic phase they will be recognized by CD8 T cells.

๐’๐จ ๐ฐ๐ก๐š๐ญ ๐ก๐š๐ฉ๐ฉ๐ž๐ง๐ฌ ๐ญ๐จ ๐ญ๐ก๐จ๐ฌ๐ž ๐ข๐ง๐๐ข๐ฏ๐ข๐๐ฎ๐š๐ฅ๐ฌ ๐ฐ๐ก๐จ ๐ก๐š๐ฏ๐ž ๐›๐ž๐ž๐ง ๐ข๐ง๐Ÿ๐ž๐œ๐ญ๐ž๐ ๐ฐ๐ข๐ญ๐ก ๐จ๐ญ๐ก๐ž๐ซ ๐ฉ๐š๐ญ๐ก๐จ๐ ๐ž๐ง๐ฌ (๐ฌ๐ฎ๐œ๐ก ๐š๐ฌ ๐‹๐จ๐ง๐  ๐‚๐Ž๐•๐ˆ๐ƒ) ๐š๐ง๐ ๐Ÿ๐š๐ข๐ฅ ๐ญ๐จ ๐œ๐จ๐ง๐ญ๐ซ๐จ๐ฅ ๐ญ๐ก๐ž๐ฆ? Well, in the end the same thing happens. Being genetically weak against these pathogens, they also end up presenting an immunodeficiency due to chronic exposure to antigens, decreasing the effective response of CD4 T cells. As these cells are the main cells that control EBV latency I, cells with latency I end up evading the immune system and multiplying, generating the same problems as in the case of EBV ME/CFS.

Therefore, in any subtype of patient with ME/CFS and in Long COVID, they end up presenting a viral syndrome due to EBV. Once EBV latency I cells are out of control, it allows any inflammatory stimulus in any tissue to recruit leukocytes (including EBV latency cells), ultimately leading to the ๐Ÿ๐จ๐ซ๐ฆ๐š๐ญ๐ข๐จ๐ง ๐จ๐Ÿ ๐„๐๐•-๐ข๐ง๐Ÿ๐ž๐œ๐ญ๐ž๐ ๐ž๐œ๐ญ๐จ๐ฉ๐ข๐œ ๐ฅ๐ฒ๐ฆ๐ฉ๐ก๐จ๐ข๐ ๐š๐ ๐ ๐ซ๐ž๐ ๐š๐ญ๐ž๐ฌ.

These formations are "ectopic" because they are outside their usual location, i.e., they do not form in primary (such as thymus and bone marrow) or secondary (such as lymph nodes and spleen) lymphoid tissues. They form transiently to cope with infection or inflammation and disappear when the stimulus is resolved.

The B cells with EBV latency use these lymphoid aggregates to their advantage, since as there is antigen presentation in these structures, they take advantage of it to pass from latency to lytic phase, generating foci of viral reactivations in these inflamed tissues. This causes that, although the initial inflammatory stimulus that provoked the formation of these aggregates has been resolved, these aggregates remain continuously in these tissues due to another inflammatory stimulus due to the molecules released by the cells with latency, as well as the viral reactivations. This would occur mainly in the mucous membranes of our organism but can occur in different tissues and would be the basis for the ๐๐ž๐ฏ๐ž๐ฅ๐จ๐ฉ๐ฆ๐ž๐ง๐ญ ๐จ๐Ÿ ๐š๐ฎ๐ญ๐จ๐ข๐ฆ๐ฆ๐ฎ๐ง๐ž ๐๐ข๐ฌ๐ž๐š๐ฌ๐ž๐ฌ.

These autoimmune diseases are generated due to the presentation of cellular autoantigens from those tissues or by the presentation of viral EBNA-1 through HLA-II proteins. The antigens when presented on HLA-II proteins undergo a series of modifications that can lead to the formation of new antigens "different" from the previous ones. In addition, EBNA-1 has a sequence similar to different proteins in our body, which can confuse our immune system and generate an autoimmune response. Here again appears the implication of having weak HLA-II haplotypes against EBV, since most of the diseases associated with this virus such as multiple sclerosis, lupus, rheumatoid arthritis, Sjรถgren's, etc. are associated with the same old HLA-II haplotypes as those weak against EBV. So having these autoimmune diseases implies that they do not control well these cells with EBV latency and therefore are responsible for the development of their autoimmunity.

๐–๐ก๐ฒ ๐๐จ ๐ฐ๐จ๐ฆ๐ž๐ง ๐ก๐š๐ฏ๐ž ๐š ๐ก๐ข๐ ๐ก๐ž๐ซ ๐ฉ๐ซ๐ž๐ฏ๐š๐ฅ๐ž๐ง๐œ๐ž ๐จ๐Ÿ ๐Œ๐„/๐‚๐…๐’, ๐‹๐จ๐ง๐  ๐‚๐Ž๐•๐ˆ๐ƒ ๐š๐ง๐ ๐š๐ฎ๐ญ๐จ๐ข๐ฆ๐ฆ๐ฎ๐ง๐ž ๐๐ข๐ฌ๐ž๐š๐ฌ๐ž๐ฌ?
Hormones play a crucial role. Estrogens, in particular, affect the CD4/CD8 T-cell ratio by reducing it, increase B-cell longevity, enhance antibody release and amplify the expression of HLA-II proteins. Under normal conditions, this increase in antibody levels in women enhances their resistance to viral infections. However, under pathological circumstances, this hormonal balance leads to a prolonged survival of B cells and a decrease in CD4 T cells, in addition to intensifying the expression of HLA-II. This situation leads to increased vulnerability to EBV due to increased survival of virus-transformed B cells and increased expression of HLA-II, which facilitates infection of a greater number of cells. In addition, elevated HLA-II expression can lead to a more robust presentation of cellular autoantigens or viral antigens that, after undergoing post-translational changes, generate neoantigens. These can activate autoreactive cells. Therefore, both the increased survival of transformed B cells and the increased antigenic presentation generated by the increased expression of HLA-II by estradiol may favor an increase in the presentation of both self and foreign antigens during an infectious process, and those abnormal plasma cells that produce autoantibodies survive longer, which consequently increases the likelihood of women developing autoimmune diseases or even cancer.

On the male side, testosterone modulates the immune system by promoting CD4 Th1 (antiviral) response and CD8 T-cell activation, while inhibiting NK cell response and HLA-II expression. Since antigen-presenting cells are essential for the differentiation of CD4 T cells toward Th1 or Th2, based on the cytokines they release, sex hormones may influence this differentiation. Women, having higher levels of antibodies (Th2 response), show a more efficient response to extracellular infections. However, against intracellular pathogens, their immune system may not be as efficient as the male immune system, which benefits from a stronger Th1 cellular response to fight virus-infected cells.

๐ƒ๐จ๐ž๐ฌ ๐ญ๐ก๐ข๐ฌ ๐ฆ๐จ๐๐ž๐ฅ ๐ž๐ฑ๐ฉ๐ฅ๐š๐ข๐ง ๐ญ๐ก๐ž ๐จ๐ฏ๐ž๐ซ๐š๐œ๐ญ๐ข๐ฏ๐ž ๐ฒ๐ž๐ญ ๐ข๐ง๐ž๐Ÿ๐Ÿ๐ž๐œ๐ญ๐ข๐ฏ๐ž ๐ข๐ฆ๐ฆ๐ฎ๐ง๐ž ๐ซ๐ž๐ฌ๐ฉ๐จ๐ง๐ฌ๐ž?
Yes, this model describes a situation in which the body's immune system is working excessively, but inefficiently, against the EBV virus:

1๏ธโƒฃ ๐ƒ๐ž๐Ÿ๐ข๐œ๐ข๐ž๐ง๐ญ ๐š๐๐š๐ฉ๐ญ๐ข๐ฏ๐ž ๐ซ๐ž๐ฌ๐ฉ๐จ๐ง๐ฌ๐ž: CD4 T cells, are not correctly recognizing and dealing with EBV latency I cells. This leads to more infected cells circulating and, at the same time, to a fatigue or exhaustion of the T cells, reducing their ability to fight the virus.

2๏ธโƒฃ ๐Ž๐ฏ๐ž๐ซ-๐š๐œ๐ญ๐ข๐ฏ๐š๐ญ๐ข๐จ๐ง ๐จ๐Ÿ ๐ข๐ง๐ง๐š๐ญ๐ž ๐ข๐ฆ๐ฆ๐ฎ๐ง๐ข๐ญ๐ฒ: Because of this failure of the adaptive response, another part of the immune system, called innate immunity, becomes over-activated because it continually detects that there is an infection but that it cannot be resolved by adaptive immunity. This results in the constant production of inflammatory substances that, instead of helping, can generate more problems and maintain chronic inflammation.

3๏ธโƒฃ ๐ˆ๐ฆ๐›๐š๐ฅ๐š๐ง๐œ๐ž ๐ข๐ง ๐ข๐ฆ๐ฆ๐ฎ๐ง๐ž ๐ซ๐ž๐ฌ๐ฉ๐จ๐ง๐ฌ๐ž๐ฌ: The body tends to favor an immune response (known as Th2) that is not best suited to fight this type of infection. This is due in part to the production of a substance called IL-6, which redirects the body's defensive response towards the production of antibodies instead of an antiviral cellular response (Th1). The increased Th2 response favors the latency and lytic cycles of EBV by activating more B cells.

๐ƒ๐จ๐ž๐ฌ ๐ญ๐ก๐ข๐ฌ ๐ฆ๐จ๐๐ž๐ฅ ๐ž๐ฑ๐ฉ๐ฅ๐š๐ข๐ง ๐ฏ๐ข๐ซ๐š๐ฅ ๐ซ๐ž๐š๐œ๐ญ๐ข๐ฏ๐š๐ญ๐ข๐จ๐ง๐ฌ ๐š๐ง๐ ๐ญ๐ก๐š๐ญ ๐จ๐Ÿ ๐จ๐ญ๐ก๐ž๐ซ ๐ฅ๐š๐ญ๐ž๐ง๐ญ ๐ฉ๐š๐ญ๐ก๐จ๐ ๐ž๐ง๐ฌ?
Yes, the model explains that, due to decreased activation and function of cytotoxic CD4 T cells, there is a loss of immune surveillance over latent infections of other pathogens. These CD4 T cells are necessary to control latent or lytic phase cells of pathogens such as Parvovirus B19, EBV, cytomegalovirus and other herpesviruses. As a result, increased viral reactivation will be observed, especially in individuals with a higher degree of immunodeficiency.

๐ƒ๐จ๐ž๐ฌ ๐ญ๐ก๐ข๐ฌ ๐ฆ๐จ๐๐ž๐ฅ ๐ž๐ฑ๐ฉ๐ฅ๐š๐ข๐ง ๐ก๐จ๐ฐ ๐ญ๐ก๐ž๐ซ๐ž ๐œ๐จ๐ฎ๐ฅ๐ ๐›๐ž ๐ข๐ง๐œ๐ซ๐ž๐š๐ฌ๐ž๐ ๐ฆ๐ž๐ญ๐š๐ฅ ๐ข๐ง๐ญ๐จ๐ฑ๐ข๐œ๐š๐ญ๐ข๐จ๐ง ๐ข๐ง ๐ญ๐ก๐ž๐ฌ๐ž ๐ฉ๐š๐ญ๐ข๐ž๐ง๐ญ๐ฌ?
If there is increased expression of MTs due to elevated intracellular zinc levels, as described in the model, those MTs will be busy binding and regulating zinc and, potentially, copper. As a result, there would be fewer MTs available to bind and detoxify heavy metals that may be present. This could lead to an accumulation of unregulated and potentially toxic heavy metals (such as cadmium and mercury) in the body.

๐ƒ๐จ๐ž๐ฌ ๐ญ๐ก๐ข๐ฌ ๐ฆ๐จ๐๐ž๐ฅ ๐ž๐ฑ๐ฉ๐ฅ๐š๐ข๐ง ๐ญ๐ก๐ž ๐ข๐ง๐œ๐ซ๐ž๐š๐ฌ๐ž๐ ๐จ๐ฑ๐ข๐๐š๐ญ๐ข๐ฏ๐ž ๐ฌ๐ญ๐ซ๐ž๐ฌ๐ฌ ๐ข๐ง ๐ญ๐ก๐ž๐ฌ๐ž ๐๐ข๐ฌ๐ž๐š๐ฌ๐ž๐ฌ?
Yes, the model explains the increased oxidative stress. Infected ectopic lymphoid structures trigger inflammatory responses by releasing certain viral components. This activation induces the release of proinflammatory cytokines, which, in turn, promote excessive production of reactive oxygen species, leading to marked oxidative stress. In addition, perturbation in the homeostasis of certain metals contributes to the disruption of intracellular antioxidant responses. Specifically, there is evidence that an antioxidant enzyme (superoxide dismutase) is affected by altered copper and zinc concentration. Briefly, the model describes how the combination of chronic inflammatory responses, together with imbalances in the homeostasis of certain metals and the persistent release of proinflammatory agents, culminates in a significant increase in oxidative and nitrosative stress in the body.

๐ƒ๐จ๐ž๐ฌ ๐ญ๐ก๐ข๐ฌ ๐ฆ๐จ๐๐ž๐ฅ ๐ž๐ฑ๐ฉ๐ฅ๐š๐ข๐ง ๐ž๐ฑ๐ž๐ซ๐œ๐ข๐ฌ๐ž ๐ข๐ง๐ญ๐จ๐ฅ๐ž๐ซ๐š๐ง๐œ๐ž ๐š๐ง๐ ๐ฉ๐จ๐ฌ๐ญ-๐ž๐ฑ๐ž๐ซ๐ญ๐ข๐จ๐ง๐š๐ฅ ๐ฆ๐š๐ฅ๐š๐ข๐ฌ๐ž?
Yes, this model explains exercise intolerance and post-exertional distress in the context of persistent EBV infection and its metabolic, immunological and neurophysiological interactions. The following is a breakdown of how the model addresses this phenomenon:

1๏ธโƒฃ ๐Œ๐ž๐ญ๐š๐›๐จ๐ฅ๐ข๐œ ๐š๐ฅ๐ญ๐ž๐ซ๐š๐ญ๐ข๐จ๐ง๐ฌ: the model describes how EBV infection can lead to insulin resistance through elevated IFN-ฮณ production. This resistance, accompanied by compensatory hyperinsulinemia, can lead to transient hypoglycemia and decreased peripheral tissue metabolism. These factors contribute to exercise intolerance, as muscles are unable to obtain the necessary energy efficiently, resulting in early fatigue.

2๏ธโƒฃ ๐€๐ฅ๐ญ๐ž๐ซ๐š๐ญ๐ข๐จ๐ง๐ฌ ๐ข๐ง ๐œ๐š๐ซ๐๐ข๐จ๐ฏ๐š๐ฌ๐œ๐ฎ๐ฅ๐š๐ซ ๐Ÿ๐ฎ๐ง๐œ๐ญ๐ข๐จ๐ง: High levels of serotonin and activation of certain receptors, such as TLR3 and TLR2, can alter cardiovascular function, affecting blood distribution and the body's ability to meet oxygen demands during exercise.

3๏ธโƒฃ ๐‚๐จ๐ฆ๐ฉ๐ซ๐จ๐ฆ๐ข๐ฌ๐ž๐ ๐ญ๐ก๐ž๐ซ๐ฆ๐จ๐ซ๐ž๐ ๐ฎ๐ฅ๐š๐ญ๐ข๐จ๐ง: The body's ability to dissipate heat generated during exercise may be impaired, which could lead to overheating.

4๏ธโƒฃ ๐Ž๐ฑ๐ข๐๐š๐ญ๐ข๐ฏ๐ž ๐ฌ๐ญ๐ซ๐ž๐ฌ๐ฌ: Chronic infection with EBV generates constant oxidative stress, which can impair mitochondrial function and reduce the ability of muscle tissue to generate energy efficiently. This oxidative stress exacerbated during exercise can lead to cell damage and muscle fatigue.

5๏ธโƒฃ ๐€๐ฅ๐ญ๐ž๐ซ๐š๐ญ๐ข๐จ๐ง๐ฌ ๐ข๐ง ๐ซ๐ž๐ฌ๐ฉ๐ข๐ซ๐š๐ญ๐จ๐ซ๐ฒ ๐Ÿ๐ฎ๐ง๐œ๐ญ๐ข๐จ๐ง: Respiratory function may be impaired, limiting adequate oxygenation during exercise and contributing to fatigue.

6๏ธโƒฃ ๐’๐ฒ๐ฌ๐ญ๐ž๐ฆ๐ข๐œ ๐ข๐ง๐Ÿ๐ฅ๐š๐ฆ๐ฆ๐š๐ญ๐ข๐จ๐ง: Activation of certain receptors, release of proinflammatory cytokines and other mechanisms associated with chronic infection can generate systemic inflammation. This inflammation can negatively affect the body's ability to recover after exercise, contributing to post-exertional malaise.

7๏ธโƒฃ ๐€๐ฅ๐ญ๐ž๐ซ๐š๐ญ๐ข๐จ๐ง๐ฌ ๐ข๐ง ๐ง๐ž๐ฎ๐ซ๐จ๐ฅ๐จ๐ ๐ข๐œ๐š๐ฅ ๐Ÿ๐ฎ๐ง๐œ๐ญ๐ข๐จ๐ง: Metabolic changes and systemic inflammation can have an impact on the central nervous system. Reduced serotonin availability and other alterations may contribute to feelings of fatigue and lethargy.

๐ƒ๐จ๐ž๐ฌ ๐ญ๐ก๐ข๐ฌ ๐ฆ๐จ๐๐ž๐ฅ ๐ž๐ฑ๐ฉ๐ฅ๐š๐ข๐ง ๐ญ๐ก๐ž ๐๐ฒ๐ฌ๐š๐ฎ๐ญ๐จ๐ง๐จ๐ฆ๐ข๐š ๐ฉ๐ซ๐ž๐ฌ๐ž๐ง๐ญ ๐ข๐ง ๐ญ๐ก๐ž๐ฌ๐ž ๐ฉ๐š๐ญ๐ข๐ž๐ง๐ญ๐ฌ?
Yes, here is a breakdown of how the model can generate dysautonomia:

1๏ธโƒฃ ๐„๐๐• ๐ข๐ง๐Ÿ๐ž๐œ๐ญ๐ข๐จ๐ง: the inability to adequately control EBV latency I cells could result in chronic inflammatory responses, which disrupts immune system homeostasis and, by extension, affects the autonomic nervous system (ANS).

2๏ธโƒฃ ๐ˆ๐ง๐Ÿ๐ฅ๐š๐ฆ๐ฆ๐š๐ญ๐จ๐ซ๐ฒ ๐ซ๐ž๐ฌ๐ฉ๐จ๐ง๐ฌ๐ž๐ฌ: proinflammatory cytokines released in response to EBV, such as IL-1ฮฒ, IL-6 and TNF-ฮฑ, can act on the brain and other organs, disrupting ANS function, leading to symptoms of dysautonomia.

3๏ธโƒฃ ๐Œ๐ž๐ญ๐š๐›๐จ๐ฅ๐ข๐œ ๐š๐ฅ๐ญ๐ž๐ซ๐š๐ญ๐ข๐จ๐ง๐ฌ: Hypozincemia and alterations in copper transport can imbalance the function of key enzymes, such as DAO. Impaired DAO function leads to an accumulation of histamine, a mediator that can cause symptoms of dysautonomia, such as vasodilatation and arrhythmias.

4๏ธโƒฃ ๐†๐š๐ฌ๐ญ๐ซ๐จ๐ข๐ง๐ญ๐ž๐ฌ๐ญ๐ข๐ง๐š๐ฅ ๐๐ข๐ฌ๐ญ๐ฎ๐ซ๐›๐š๐ง๐œ๐ž๐ฌ: Serotonin accumulation in the gut can stimulate the vagus nerve, a primary connection between the gut and the brain. Overstimulation of the vagus nerve can trigger symptoms of dysautonomia, such as bradycardia.

5๏ธโƒฃ ๐๐ž๐ฎ๐ซ๐จ๐ฅ๐จ๐ ๐ข๐œ๐š๐ฅ ๐š๐ฅ๐ญ๐ž๐ซ๐š๐ญ๐ข๐จ๐ง๐ฌ: A decrease in brain extracellular serotonin and an increase in neurotoxic metabolites of kynurenine may alter neuronal and ANS function, which could manifest as fatigue, exercise intolerance and other symptoms of dysautonomia.

6๏ธโƒฃ ๐•๐š๐ฌ๐œ๐ฎ๐ฅ๐š๐ซ ๐š๐ฅ๐ญ๐ž๐ซ๐š๐ญ๐ข๐จ๐ง๐ฌ: Microclot formation can affect adequate blood perfusion in organs and tissues, including the brain. Inadequate perfusion can result in neurological and autonomic symptoms.

7๏ธโƒฃ ๐„๐ง๐๐จ๐œ๐ซ๐ข๐ง๐ž ๐๐ข๐ฌ๐ญ๐ฎ๐ซ๐›๐š๐ง๐œ๐ž๐ฌ: Hyperinsulinemia and possible reduction in cortisol secretion may affect the balance of the ANS. For example, hypoglycemia may trigger an acute sympathetic response, while decreased cortisol may affect the body's ability to handle stress.

๐ƒ๐จ๐ž๐ฌ ๐ญ๐ก๐ข๐ฌ ๐ฆ๐จ๐๐ž๐ฅ ๐ž๐ฑ๐ฉ๐ฅ๐š๐ข๐ง ๐ง๐ž๐ฎ๐ซ๐จ๐ข๐ง๐Ÿ๐ฅ๐š๐ฆ๐ฆ๐š๐ญ๐ข๐จ๐ง, ๐ฆ๐ž๐ง๐ญ๐š๐ฅ ๐Ÿ๐จ๐  ๐š๐ง๐ ๐œ๐จ๐ ๐ง๐ข๐ญ๐ข๐ฏ๐ž ๐ข๐ฆ๐ฉ๐š๐ข๐ซ๐ฆ๐ž๐ง๐ญ?
Yes, this presented model could explain neuroinflammation, mental fog and cognitive impairment as follows:

1๏ธโƒฃ ๐๐ž๐ฎ๐ซ๐จ๐ข๐ง๐Ÿ๐ฅ๐š๐ฆ๐ฆ๐š๐ญ๐ข๐จ๐ง: In patients with ME/CFS and LC, there is evidence of chronic viral infection or virus reactivation, especially EBV. When viral genetic material is present, especially EBV EBERs, TLR3 receptors in microglia (immune cells of the central nervous system) are activated. This activation results in the release of proinflammatory cytokines such as IL-1ฮฒ and TNF-ฮฑ. These cytokines may contribute to chronic inflammation of the central nervous system, characterizing neuroinflammation.

2๏ธโƒฃ ๐Œ๐ž๐ง๐ญ๐š๐ฅ ๐Ÿ๐จ๐  ๐š๐ง๐ ๐œ๐จ๐ ๐ง๐ข๐ญ๐ข๐ฏ๐ž ๐ข๐ฆ๐ฉ๐š๐ข๐ซ๐ฆ๐ž๐ง๐ญ: several pathways mentioned in the model may contribute to these symptoms. For example:

- Viral infection can cause damage to the blood-brain barrier, allowing entry of viral genetic material that could further activate microglia and contribute to neuroinflammation.

- Increased IDO activity and decreased tryptophan levels lead to a reduction in serotonin (5-HT) and melatonin synthesis. Since serotonin plays a role in the regulation of mood, cognition and alertness, its reduction could contribute to mental fog.

- Quinolinic acid, a metabolite of tryptophan, has neurotoxic properties by binding to the NMDA receptor, which may increase nitrosative stress and contribute to cognitive impairment.

- Increased oxidative and nitrosative stress in EBV-infected cells, together with neuroinflammation, may interfere with proper neuronal functioning and contribute to cognitive impairment.

- Alterations in the serotonergic system may also directly affect cognitive function and perception of fatigue.

๐‹๐ข๐ง๐ค:

๐‚๐จ๐ง๐ญ๐ข๐ง๐ฎ๐ž ๐ญ๐ก๐ž ๐ญ๐ก๐ซ๐ž๐š๐ ๐Ÿงต๐Ÿ‘‡๐Ÿฝ

#EpsteinBarrVirus #EBV #LongCovid #MECFS #MyalgicEncephalomyelitis #Health #Research #News #microE2324 #medicine #Microbiology #VIRUS #ChronicDiseases #SARSCOV2 #COVID19 #LongHaulers #MCAS #ME #MyE #CFSME

@microbeminded2 @MVGutierrezMD @elisaperego78 @TomKindlon @Dan_Wyke @research_long @agy_lena @SarahOC_MECFS @joshuamcclure @Lymenews @JanetDafoe @EricTopol
@SGriffin_Lab
Image ๐ƒ๐จ๐ž๐ฌ ๐ญ๐ก๐ข๐ฌ ๐ฆ๐จ๐๐ž๐ฅ ๐ž๐ฑ๐ฉ๐ฅ๐š๐ข๐ง ๐ญ๐ก๐ž ๐จ๐œ๐œ๐ฎ๐ซ๐ซ๐ž๐ง๐œ๐ž ๐จ๐Ÿ ๐๐ข๐ ๐ž๐ฌ๐ญ๐ข๐ฏ๐ž ๐ฉ๐ซ๐จ๐›๐ฅ๐ž๐ฆ๐ฌ, ๐Ÿ๐จ๐จ๐ ๐ข๐ง๐ญ๐จ๐ฅ๐ž๐ซ๐š๐ง๐œ๐ž๐ฌ ๐š๐ง๐ ๐š๐ฅ๐ญ๐ž๐ซ๐š๐ญ๐ข๐จ๐ง๐ฌ ๐ข๐ง ๐ญ๐ก๐ž ๐ฆ๐ข๐œ๐ซ๐จ๐›๐ข๐จ๐ญ๐š ๐ข๐ง ๐ญ๐ก๐ž๐ฌ๐ž ๐ฉ๐š๐ญ๐ข๐ž๐ง๐ญ๐ฌ?
Yes, I will summarize the key points below:

1๏ธโƒฃ ๐ƒ๐ข๐ ๐ž๐ฌ๐ญ๐ข๐ฏ๐ž ๐ฉ๐ซ๐จ๐›๐ฅ๐ž๐ฆ๐ฌ:
- The accumulation of 5-HT (serotonin) in the intestinal mucosa causes chronic inflammation.

- This intestinal inflammation leads to a decrease in stomach acid secretion and in the expression of enzymes needed to digest carbohydrates. As a result, more carbohydrates reach the intestinal microbiota without being digested or absorbed, which can cause digestive problems such as bloating, gas and diarrhea.

- The serotonergic alteration, together with the increase in proinflammatory cytokines, causes an alteration in acid secretion, breakdown of the intestinal barrier and decreased expression of enzymes needed to digest carbohydrates.

2๏ธโƒฃ ๐…๐จ๐จ๐ ๐ข๐ง๐ญ๐จ๐ฅ๐ž๐ซ๐š๐ง๐œ๐ž๐ฌ:
- Dysfunction of adaptive immunity, combined with increased nutrient input for commensal bacteria and decreased acid secretion, leads to bacterial proliferation, resulting in small intestinal bacterial overgrowth (SIBO) and the development of food intolerances.

- Alterations in the intestinal barrier allow substances and microorganisms that should not normally cross it to do so, further activating innate immunity and leading to increased accumulation of 5-HT, which aggravates symptoms.

3๏ธโƒฃ ๐€๐ฅ๐ญ๐ž๐ซ๐š๐ญ๐ข๐จ๐ง๐ฌ ๐ข๐ง ๐ญ๐ก๐ž ๐ฆ๐ข๐œ๐ซ๐จ๐›๐ข๐จ๐ญ๐š:
- The disruption of tight junctions between enterocytes, caused by increased proinflammatory cytokines, leads to increased intestinal permeability. As a result, bacteria and substances that should not cross the barrier do so, which alters the microbiota and leads to increased activation of the innate immune system.

- SIBO has other consequences, such as bacterial deconjugation of bile salts, leading to poor micelle formation and fat malabsorption, as well as deficiencies in fat-soluble vitamins (A, D, E, and K). In addition, competitive absorption of vitamin B12 by bacteria results in less binding to intrinsic factor and, therefore, less absorption in the terminal ileum.

๐ƒ๐จ๐ž๐ฌ ๐ญ๐ก๐ข๐ฌ ๐ฆ๐จ๐๐ž๐ฅ ๐ž๐ฑ๐ฉ๐ฅ๐š๐ข๐ง ๐ญ๐ก๐ž ๐ฆ๐ž๐ญ๐š๐›๐จ๐ฅ๐ข๐œ ๐š๐ฅ๐ญ๐ž๐ซ๐š๐ญ๐ข๐จ๐ง๐ฌ ๐š๐ง๐ ๐ฆ๐ข๐ญ๐จ๐œ๐ก๐จ๐ง๐๐ซ๐ข๐š๐ฅ ๐๐ฒ๐ฌ๐Ÿ๐ฎ๐ง๐œ๐ญ๐ข๐จ๐ง ๐ฉ๐ซ๐ž๐ฌ๐ž๐ง๐ญ ๐ข๐ง ๐ญ๐ก๐ž๐ฌ๐ž ๐ฉ๐š๐ญ๐ข๐ž๐ง๐ญ๐ฌ?
Yes, let's see how:

1๏ธโƒฃ ๐Œ๐ž๐ญ๐š๐›๐จ๐ฅ๐ข๐œ ๐š๐ฅ๐ญ๐ž๐ซ๐š๐ญ๐ข๐จ๐ง๐ฌ:
- EBV infection, especially in individuals with "weak" HLA-II haplotypes, leads to immune evasion, which promotes inflammatory responses in the body. This inflammation can affect various metabolic pathways.

- Increased release of proinflammatory cytokines, such as IL-1ฮฒ, IL-6, IL-8, IL-12, TNF-ฮฑ, and IFN-ฮณ, has metabolic consequences. These cytokines can alter the balance of certain minerals such as zinc and copper, which can disrupt several essential enzymatic functions.

- Insulin resistance induced by elevated levels of IFN-ฮณ affects glucose metabolism. To compensate, the pancreas produces more insulin, leading to hyperinsulinemia, which has various consequences, including an effect on hepatic glycogen metabolism.

2๏ธโƒฃ ๐Œ๐ข๐ญ๐จ๐œ๐ก๐จ๐ง๐๐ซ๐ข๐š๐ฅ ๐๐ฒ๐ฌ๐Ÿ๐ฎ๐ง๐œ๐ญ๐ข๐จ๐ง:
- Serotonergic disturbances caused by infections can affect mitochondrial function. Serotonin depletion in the central nervous system (CNS) plays a role in the regulation of appetite and energy metabolism.

- Constant activation of the anaerobic glycolytic pathway, either due to the Warburg effect in infected cells or due to mitochondrial dysfunction, results in elevated lactic acid production.

- The generation of quinolinic, a metabolite of tryptophan, may be neurotoxic and contribute to nitrosative stress, which may further impair mitochondrial function.

- Overstimulation of NMDA receptors, whether by quinolinic acid or other pathways, can lead to increased nitric oxide/peroxynitrite levels, which has direct consequences on mitochondrial function and health.

3๏ธโƒฃ ๐†๐ฅ๐จ๐›๐š๐ฅ ๐œ๐จ๐ง๐ฌ๐ž๐ช๐ฎ๐ž๐ง๐œ๐ž๐ฌ:
- Energy depletion, as a result of metabolic alterations and mitochondrial dysfunction, can manifest as chronic fatigue and other associated symptoms.

- Alterations in the serotonergic system, along with metabolic imbalances and chronic inflammation, may contribute to neurological symptoms, including fatigue and depression.

๐ƒ๐จ๐ž๐ฌ ๐ญ๐ก๐ข๐ฌ ๐ฆ๐จ๐๐ž๐ฅ ๐ž๐ฑ๐ฉ๐ฅ๐š๐ข๐ง ๐ญ๐ก๐ž ๐Ÿ๐จ๐ซ๐ฆ๐š๐ญ๐ข๐จ๐ง ๐จ๐Ÿ ๐ฆ๐ข๐œ๐ซ๐จ๐œ๐ฅ๐จ๐ญ ๐Ÿ๐จ๐ซ๐ฆ๐š๐ญ๐ข๐จ๐ง?
Yes, the following are the key points of this process according to the model:

1๏ธโƒฃ ๐๐ซ๐จ๐ข๐ง๐Ÿ๐ฅ๐š๐ฆ๐ฆ๐š๐ญ๐จ๐ซ๐ฒ ๐œ๐ฒ๐ญ๐จ๐ค๐ข๐ง๐ž๐ฌ ๐š๐ง๐ ๐ก๐ฒ๐ฉ๐ž๐ซ๐œ๐จ๐š๐ ๐ฎ๐ฅ๐š๐ญ๐ข๐จ๐ง:
- Overproduction of proinflammatory cytokines, such as TNFฮฑ, IL-6 and IL-1ฮฒ during infectious processes, leads to hypercoagulation, platelet activation, leukocyte infiltration and vascular hyperpermeability.

2๏ธโƒฃ ๐๐ฅ๐š๐ญ๐ž๐ฅ๐ž๐ญ ๐š๐œ๐ญ๐ข๐ฏ๐š๐ญ๐ข๐จ๐ง:
- Platelets, when activated through 5-HT2A receptors due to increased 5-HT in peripheral blood or by binding of EBERs to TLR3, bind fibrinogen, which enhances aggregation and coagulation processes.

- Activated platelets can release ฮฒ-amyloid (Aฮฒ) peptide, which, interacting with fibrinogen, causes fibrinogen oligomerization, fibrin deposition and Aฮฒ fibrillation, favoring the formation of abnormal microclots resistant to degradation by fibrinolytic enzymes.

3๏ธโƒฃ ๐€๐ฆ๐ฒ๐ฅ๐จ๐ข๐๐จ๐ ๐ž๐ง๐ž๐ฌ๐ข๐ฌ ๐š๐ง๐ ๐Ÿ๐ข๐›๐ซ๐ข๐ง๐จ๐ ๐ž๐ง๐ž๐ฌ๐ข๐ฌ:
- Elevated levels of IL-1ฮฒ, IL-6 and TNF-ฮฑ stimulate the production of serum amyloid A (SAA) protein in hepatocytes.

- Increased SAA in the blood may favor its interaction with fibrinogen, leading to amyloidogenic changes in fibrinogen, resulting in the formation of fibrin amyloid microaggregates resistant to fibrinolysis.

4๏ธโƒฃ ๐‚๐š๐ฉ๐ข๐ฅ๐ฅ๐š๐ซ๐ฒ ๐จ๐›๐ฌ๐ญ๐ซ๐ฎ๐œ๐ญ๐ข๐จ๐ง ๐š๐ง๐ ๐ž๐ง๐๐จ๐ญ๐ก๐ž๐ฅ๐ข๐š๐ฅ ๐๐š๐ฆ๐š๐ ๐ž:
- The formation of these micro-clots leads to capillary obstruction, which compromises blood flow and increases inflammation, contributing to the appearance of various symptoms.

- In addition, endothelial damage caused by EBV infection, either through positive activation of NOX2 by EBNA-1 in EBV-transformed endothelial cells or through activation of TLR3 by EBERs, may influence microclot formation. NOX2 activation may cause vasoconstriction and thrombosis through platelet aggregation via overproduction of hydrogen peroxide, isoprostanes, or inactivation of nitric oxide.

๐ƒ๐จ๐ž๐ฌ ๐ญ๐ก๐ข๐ฌ ๐ฆ๐จ๐๐ž๐ฅ ๐ž๐ฑ๐ฉ๐ฅ๐š๐ข๐ง ๐ญ๐ก๐ž ๐š๐ฅ๐ญ๐ž๐ซ๐š๐ญ๐ข๐จ๐ง ๐ข๐ง ๐œ๐จ๐ซ๐ญ๐ข๐ฌ๐จ๐ฅ ๐ฅ๐ž๐ฏ๐ž๐ฅ๐ฌ?
Yes. The following are the key points of this process according to the model:

1๏ธโƒฃ ๐ˆ๐ง๐ข๐ญ๐ข๐š๐ฅ ๐ฌ๐ญ๐ข๐ฆ๐ฎ๐ฅ๐ฎ๐ฌ:
- Under acute conditions, proinflammatory cytokines such as IFN-ฮณ, TNFฮฑ, IL-1 and IL-6 stimulate the hypothalamic-pituitary-adrenal (HPA) axis by activating ACTH secretion. IFN-ฮณ, for example, not only activates macrophages, but also allows an increase in glucocorticoid receptor expression for subsequent feedback inhibition. So that the inflammatory response is not exaggerated, there is a stimulation in cortisol secretion due to the increase in ACTH, which results from the action of IFN-ฮณ and the direct stimulation of the adrenal gland by IL-6.

2๏ธโƒฃ ๐’๐ฎ๐ฉ๐ฉ๐ซ๐ž๐ฌ๐ฌ๐ข๐จ๐ง ๐จ๐Ÿ ๐œ๐จ๐ซ๐ญ๐ข๐ฌ๐จ๐ฅ ๐ฌ๐ž๐œ๐ซ๐ž๐ญ๐ข๐จ๐ง ๐๐ฎ๐ซ๐ข๐ง๐  ๐ฉ๐ž๐ซ๐ฌ๐ข๐ฌ๐ญ๐ž๐ง๐ญ ๐ข๐ง๐Ÿ๐ž๐œ๐ญ๐ข๐จ๐ง๐ฌ:
- During persistent infections, chronic exposure to IL-10, TGF-ฮฒ1 and TNFฮฑ could suppress ACTH-stimulated cortisol secretion in the adrenal gland, leading to relative hypocortisolism.

- The anti-inflammatory and immunosuppressive cytokines, IL-10 and TGF-ฮฒ1, are elevated in EBV infections. These cytokines are secreted by EBV latent cells and regulatory T cells to counteract proinflammatory cytokines and evade CD4 T cells. Therefore, the greater the number of EBV reservoir-infected tissues, the greater the secretion of these anti-inflammatory cytokines and thus the greater the negative feedback on cortisol secretion.

3๏ธโƒฃ ๐๐จ๐ฌ๐ฌ๐ข๐›๐ฅ๐ž ๐ข๐ง๐Ÿ๐ž๐œ๐ญ๐ข๐จ๐ง ๐ข๐ง ๐ญ๐ก๐ž ๐š๐๐ซ๐ž๐ง๐š๐ฅ ๐จ๐ซ ๐ฉ๐ข๐ญ๐ฎ๐ข๐ญ๐š๐ซ๐ฒ ๐ ๐ฅ๐š๐ง๐๐ฌ:
- Disease progression and acquired immunodeficiency in CD4 T-cell function may lead to an increase in the replicative rate of the virus and infection in the adrenal or pituitary glands, depleting cortisol stores.

4๏ธโƒฃ ๐ˆ๐ฆ๐ฉ๐š๐œ๐ญ ๐จ๐Ÿ ๐œ๐ก๐ซ๐จ๐ง๐ข๐œ ๐ข๐ง๐ฌ๐ฎ๐ฅ๐ข๐ง๐ข๐ฌ๐ฆ:
- Chronic high insulin may also play a role in the development of hypocortisolism, as insulin may inhibit the secretion of corticotropin-releasing hormone in the hypothalamus, which in turn would inhibit ACTH secretion in the pituitary gland.

๐Ÿ”ตIn the next days I will be uploading more consequences that our model responds. As a patient with ME/CFS after EBV infection, I see the urgent need to find treatments against this virus.
#Science #virology #Immunology
Image
Sep 29, 2023 โ€ข 15 tweets โ€ข 18 min read
๐Ÿฆ  ๐Ÿ” ยฟ๐’๐š๐›๐ขฬ๐š๐ฌ ๐ช๐ฎ๐ž ๐ž๐ฅ ๐ฏ๐ข๐ซ๐ฎ๐ฌ ๐๐ž ๐„๐ฉ๐ฌ๐ญ๐ž๐ข๐ง-๐๐š๐ซ๐ซ ๐ฉ๐จ๐๐ซ๐ขฬ๐š ๐ฌ๐ž๐ซ ๐ฎ๐ง ๐๐ž๐ฌ๐ž๐ง๐œ๐š๐๐ž๐ง๐š๐ง๐ญ๐ž ๐๐ž ๐ฅ๐จ๐ฌ ๐ฌ๐ขฬ๐ง๐ญ๐จ๐ฆ๐š๐ฌ ๐ž๐ง ๐ฅ๐š ๐„๐Œ/๐’๐…๐‚ ๐ฒ ๐ž๐ง ๐ž๐ฅ ๐‹๐จ๐ง๐  ๐‚๐Ž๐•๐ˆ๐ƒ? Abro ๐Ÿงต

A continuaciรณn, os resumo los puntos de nuestra reciente revisiรณn publicada y como podrรญa explicar el desarrollo de ๐๐ข๐ฌ๐š๐ฎ๐ญ๐จ๐ง๐จ๐ฆ๐ขฬ๐š, ๐ฆ๐ข๐œ๐ซ๐จ๐œ๐จ๐šฬ๐ ๐ฎ๐ฅ๐จ๐ฌ, ๐ž๐ง๐Ÿ๐ž๐ซ๐ฆ๐ž๐๐š๐๐ž๐ฌ ๐š๐ฎ๐ญ๐จ๐ข๐ง๐ฆ๐ฎ๐ง๐ž๐ฌ, ๐ข๐ง๐ญ๐จ๐ฅ๐ž๐ซ๐š๐ง๐œ๐ข๐š ๐š๐ฅ ๐ž๐ฃ๐ž๐ซ๐œ๐ข๐œ๐ข๐จ, ๐๐ข๐ฌ๐Ÿ๐ฎ๐ง๐œ๐ข๐จฬ๐ง ๐ฆ๐ข๐ญ๐จ๐œ๐จ๐ง๐๐ซ๐ข๐š๐ฅ, ๐ก๐ข๐ฉ๐จ๐œ๐จ๐ซ๐ญ๐ข๐ฌ๐จ๐ฅ๐ข๐ฌ๐ฆ๐จ ๐ฒ ๐ฆ๐ฎ๐œ๐ก๐š๐ฌ ๐ฆ๐šฬ๐ฌ ๐œ๐จ๐ง๐ฌ๐ž๐œ๐ฎ๐ž๐ง๐œ๐ข๐š๐ฌ.

Muchos se preguntan por quรฉ el EBV, el virus de Epstein-Barr, y no otro virus, es el principal desencadenante del cuadro sintomรกtico de la EM/SFC y del Long COVID, cuando existen otros factores, como distintas infecciones, intoxicaciรณn por metales, entre otros. En las siguientes lรญneas, expondrรฉ los fundamentos de este fenรณmeno, resumiendo los puntos esenciales del artรญculo de revisiรณn que publicamos recientemente.

El comรบn denominador de todos los factores desencadenantes de ambas enfermedades es su capacidad para inmunodeprimir. Toda infecciรณn intracelular, sea bacteriana, viral o parasitaria, que no sea controlada, lleva a la inmunosupresiรณn debido a la exposiciรณn crรณnica a antรญgenos. Normalmente, la habilidad para controlar una infecciรณn se debe a factores genรฉticos, particularmente a los genes HLA (I y II). Estos genes codifican proteรญnas llamadas antรญgeno leucocitario humano (HLA), esenciales para distinguir lo propio de lo ajeno en nuestro cuerpo. Si esta "alerta" no funciona adecuadamente, el sistema inmune no puede eliminar eficientemente patรณgenos. Los HLA son como un escรกner que permite identificar las cosas que no son del cuerpo y responder a ellas. Si este escรกner no funciona correctamente y no reconoce algo que es malo, impedirรญa que tu sistema inmune lo elimine y por tanto, que se mueva libremente por tu organismo sin ser eliminado.

En el contexto de la EM/SFC, existen ๐๐จ๐ฌ ๐ž๐ฌ๐œ๐ž๐ง๐š๐ซ๐ข๐จ๐ฌ ๐ฉ๐จ๐ฌ๐ข๐›๐ฅ๐ž๐ฌ: el individuo puede tener una predisposiciรณn genรฉtica directa frente al EBV, o puede perder el control sobre el EBV debido a una inmunodeficiencia subyacente causada por otra infecciรณn o exposiciรณn a quรญmicos o metales. Si se trata de otra infecciรณn, tambiรฉn estarรญamos hablando de una "debilidad genรฉtica" (HLA) frente a esos patรณgenos especรญficos.

Si el individuo es genรฉticamente susceptible al EBV, es porque posee haplotipos HLA-II antiguos, a los que el EBV ha coevolucionado. Esto se debe principalmente porque el EBV infecta uniรฉndose a las proteinas HLA-II de las cรฉlulas. Este virus es astuto: al infectar una cรฉlula, introduce su ADN en ella de manera latente sin generar nuevos viriones, evitando la detecciรณn del sistema inmune y utilizando las cรฉlulas B como "caballos de Troya".

Si bien el 95% de la poblaciรณn mundial tiene EBV, solo una minorรญa desarrolla problemas. Aquรญ es donde entran en juego de nuevo los haplotipos HLA-II โ€œdรฉbilesโ€ frente al EBV.

Aunque se diga que la mayorรญa de la poblaciรณn no tiene problemas con este virus, eso no del todo cierto. Pensad que este virus se aprovecha cada vez que os inmunodeprimรญs por cualquier motivo. Un ejemplo lo tenรฉis con su hermano herpes labial, que cada vez que esa persona se inmunodeprime por cualquier motivo, aprovecha e infecta mรกs cรฉlulas haciendose visible la lesiรณn en los labios. Pero cada vez que vuelve a remontar el sistema inmunologico del primer evento que lo inmunodeprimiรณ, se vuelve a controlar este virus y desaparece la lesiรณn. Esto es exactamente lo mismo que ocurre en las personas sanas que son capaces de controlar el EBV.

La principal diferencia entre estas personas sanas y los pacientes de EM/SFC que tienen problemas con el EBV, es que al tener haplotipos HLA-II fuertes contra el EBV son capaces de reconocer todos los tipos de latencia del EBV y controlarlos. En cambio, ๐ฅ๐จ๐ฌ ๐ฉ๐š๐œ๐ข๐ž๐ง๐ญ๐ž๐ฌ ๐œ๐จ๐ง ๐„๐Œ/๐’๐…๐‚ ๐ฉ๐จ๐ซ ๐„๐๐•, ๐š๐ฅ ๐ญ๐ž๐ง๐ž๐ซ ๐ก๐š๐ฉ๐ฅ๐จ๐ญ๐ข๐ฉ๐จ๐ฌ ๐๐žฬ๐›๐ข๐ฅ๐ž๐ฌ ๐œ๐จ๐ง๐ญ๐ซ๐š ๐ž๐ฅ ๐„๐๐•, ๐ฌ๐ฎ๐ฌ ๐œ๐žฬ๐ฅ๐ฎ๐ฅ๐š๐ฌ ๐“๐‚๐ƒ๐Ÿ’ ๐ง๐จ ๐ฌ๐จ๐ง ๐œ๐š๐ฉ๐š๐œ๐ž๐ฌ ๐๐ž ๐ซ๐ž๐œ๐จ๐ง๐จ๐œ๐ž๐ซ ๐ฅ๐š๐ฌ ๐œ๐žฬ๐ฅ๐ฎ๐ฅ๐š๐ฌ ๐œ๐จ๐ง ๐ฅ๐š๐ญ๐ž๐ง๐œ๐ข๐š ๐ˆ ๐๐ž๐ฅ ๐„๐๐•. El resto de tipos de latencias si son capaces de ser reconocidas puesto que son controladas por las cรฉlulas TCD8, ya que no tendrรญan haplotipos HLA-I dรฉbiles contra el EBV.

Vamos a explicar mejor esto. Las cรฉlulas TCD4 reconocen antรญgenos presentados en proteรญnas HLA-II y las cรฉlulas TCD8 reconocen antรญgenos presentados en proteรญnas HLA-I. Normalmente, cualquier infecciรณn intracelular es controlada por las cรฉlulas TCD8 porque las cรฉlulas infectadas presentan en las proteรญnas HLA-I de su membrana los antรญgenos del patรณgeno que tienen dentro. Por otro lado, las proteรญnas HLA-II se encuentran sobre todo en cรฉlulas presentadoras de antรญgenos que se encargan de coger antรญgenos que se encuentran fuera de la cรฉlula y presentarlos en sus proteรญnas HLA-II para que sean reconocidas por las cรฉlulas T-CD4. Entonces, pensarรญamos que el EBV al ser un patรณgeno intracelular, sus antรญgenos deberรญan ser presentados siempre en las proteรญnas HLA-I de la cรฉlula infectada. Pues esto no es asรญ siempre, este virus ha evolucionado para esquivar este sistema al generar latencia. Uno de los mecanismos de evasiรณn para permanecer sin ser reconocido es evitar que un antรญgeno de latencia, llamado EBNA-1, sea presentado en las proteรญnas HLA-I y pase a ser presentado en las HLA-II. Esto es de suma importancia para el virus porque evita por ejemplo, que las cรฉlulas de latencia I (solo expresan EBNA-1 del virus) sean reconocidas por las cรฉlulas TCD8. En cambio, el resto de las cรฉlulas con latencia (II y III) y cรฉlulas lรญticas sin serรญan reconocidas y eliminadas por las cรฉlulas T CD8. Entonces pensarรญamos que nadie podrรญa controlar las cรฉlulas con latencia I si evaden a las cรฉlulas T CD8. Aquรญ nuestro sistema inmune tambiรฉn es inteligente y es donde las cรฉlulas T CD4 juegan el papel diferencial entre las personas sanas y enfermas con este virus. Vuelve a aparecer la importancia de los haplotipos HLA-II. Aquellos individuos que tengan haplotipos HLA-II resistentes al EBV serรกn capaces de presentar bien el antรญgeno EBNA-1 en las cรฉlulas con latencia I y serรกn reconocidas y eliminadas por las cรฉlulas TCD4. En cambio, aquellos que tengan los haplotipos HLA-II dรฉbiles frente al EBV, no podrรกn presentar bien EBNA-1 en las proteรญnas HLA-II y por tanto, las cรฉlulas T CD4 no reconocerรกn las cรฉlulas con latencia I. Estas cรฉlulas con latencia I al no ser controladas, se multiplicarรกn y causarรกn inflamaciรณn y daรฑo. Pero cada vez que pasen a otro tipo de latencia o a fase lรญtica si serรกn reconocidas por las cรฉlulas T CD8.

Entonces ยฟ๐ช๐ฎ๐žฬ ๐จ๐œ๐ฎ๐ซ๐ซ๐ž ๐œ๐จ๐ง ๐š๐ช๐ฎ๐ž๐ฅ๐ฅ๐จ๐ฌ ๐ข๐ง๐๐ข๐ฏ๐ข๐๐ฎ๐จ๐ฌ ๐ช๐ฎ๐ž ๐ฌ๐ž ๐ก๐š๐ง ๐ข๐ง๐Ÿ๐ž๐œ๐ญ๐š๐๐จ ๐œ๐จ๐ง ๐จ๐ญ๐ซ๐จ๐ฌ ๐ฉ๐š๐ญ๐จฬ๐ ๐ž๐ง๐จ๐ฌ (๐œ๐จ๐ฆ๐จ ๐‹๐จ๐ง๐  ๐‚๐Ž๐•๐ˆ๐ƒ) ๐ฒ ๐ง๐จ ๐ฅ๐จ๐ ๐ซ๐š๐ง ๐œ๐จ๐ง๐ญ๐ซ๐จ๐ฅ๐š๐ซ๐ฅ๐จ๐ฌ? Pues al final ocurre lo mismo. Al ser dรฉbiles genรฉticamente contra estos patรณgenos acaban tambiรฉn presentando una inmunodeficiencia por la exposiciรณn crรณnica a los antรญgenos, disminuyendo la respuesta efectiva de las cรฉlulas T CD4. Como estas cรฉlulas son las principales que controlan la latencia I del EBV, las cรฉlulas con latencia I acaban evadiendo al sistema inmune y multiplicรกndose, generando los mismos problemas que en el caso de la EM/SFC por EBV.

Por tanto, en cualquier subtipo de paciente con EM/SFC y en el Long COVID, acaban presentando un sรญndrome viral por el EBV. Una vez instaurado el descontrol de las cรฉlulas con latencia I de EBV, permite que cualquier estรญmulo inflamatorio en cualquier tejido reclute leucocitos (entre los cuales se encuentran cรฉlulas con latencia del VEB), lo que en รบltima instancia conduce a la ๐Ÿ๐จ๐ซ๐ฆ๐š๐œ๐ข๐จฬ๐ง ๐๐ž ๐š๐ ๐ซ๐ž๐ ๐š๐๐จ๐ฌ ๐ฅ๐ข๐ง๐Ÿ๐จ๐ข๐๐ž๐ฌ ๐ž๐œ๐ญ๐จฬ๐ฉ๐ข๐œ๐จ๐ฌ ๐ข๐ง๐Ÿ๐ž๐œ๐ญ๐š๐๐š๐ฌ ๐ฉ๐จ๐ซ ๐ž๐ฅ ๐•๐„๐.

Estas formaciones son "ectรณpicas" porque se encuentran fuera de su ubicaciรณn habitual, es decir, no se forman en los tejidos linfoides primarios (como el timo y la mรฉdula รณsea) o secundarios (como los ganglios linfรกticos y el bazo). Se forman de manera transitoria para hacer frente a una infecciรณn o inflamaciรณn y desaparecen cuando se resuelve el estรญmulo.

Las cรฉlulas B con latencia de EBV usan estos agregados linfoides en su beneficio, ya que como hay presentaciรณn de antรญgenos en estas estructuras, lo aprovechan para pasar de formas de latencia a fase lรญtica, generando focos de reactivaciones virales en estos tejidos inflamados. Esto hace que, aunque se haya resuelto el estรญmulo inflamatorio inicial que provocรณ la formaciรณn de estos agregados, permanezcan continuamente estos agregados en estos tejidos al permanecer otro estรญmulo inflamatorio debido tanto a las molรฉculas liberadas por las cรฉlulas con latencia, como las reactivaciones virales. Esto ocurrirรญa sobre todo en las mucosas de nuestro organismo pero puede ocurrir en diferentes tejidos y serรญa la base del ๐๐ž๐ฌ๐š๐ซ๐ซ๐จ๐ฅ๐ฅ๐จ ๐๐ž ๐ฅ๐š๐ฌ ๐ž๐ง๐Ÿ๐ž๐ซ๐ฆ๐ž๐๐š๐๐ž๐ฌ ๐š๐ฎ๐ญ๐จ๐ข๐ง๐ฆ๐ฎ๐ง๐ž๐ฌ.

Estas enfermedades autoinmunes se generan debido a la presentaciรณn de autoantรญgenos celulares de esos tejidos o por la presentaciรณn de EBNA-1 viral a travรฉs de las proteรญnas HLA-II. Los antรญgenos cuando son presentados en las proteรญnas HLA-II sufren una serie de modificaciones que pueden provocar la formaciรณn de nuevos antรญgenos โ€œdiferentesโ€ de los anteriores. Ademรกs, EBNA-1 tiene una secuencia parecida a diferentes proteรญnas de nuestro cuerpo, pudiendo confundir a nuestro sistema inmune y generar una respuesta autoinmune. Aquรญ vuelve a aparecer la implicaciรณn de tener unos haplotipos HLA-II dรฉbiles contra el EBV, ya que la mayorรญa de las enfermedades asociadas a este virus como la esclerosis mรบltiple, lupus, artritis reumatoide, Sjรถgren, etc estรกn asociados a los mismos haplotipos antiguos HLA-II que los dรฉbiles frente al EBV. Por lo que tener estas enfermedades autoinmunes lleva implรญcito que no controlan bien estas cรฉlulas con latencia de EBV y por lo tanto, son las responsables del desarrollo de su autoinmunidad.

ยฟ๐๐จ๐ซ ๐ช๐ฎ๐žฬ ๐ฅ๐š๐ฌ ๐ฆ๐ฎ๐ฃ๐ž๐ซ๐ž๐ฌ ๐ฉ๐ซ๐ž๐ฌ๐ž๐ง๐ญ๐š๐ง ๐ฎ๐ง๐š ๐ฆ๐š๐ฒ๐จ๐ซ ๐ฉ๐ซ๐ž๐ฏ๐š๐ฅ๐ž๐ง๐œ๐ข๐š ๐๐ž ๐„๐Œ/๐’๐…๐‚, ๐‹๐จ๐ง๐  ๐‚๐Ž๐•๐ˆ๐ƒ ๐ฒ ๐ž๐ง๐Ÿ๐ž๐ซ๐ฆ๐ž๐๐š๐๐ž๐ฌ ๐š๐ฎ๐ญ๐จ๐ข๐ง๐ฆ๐ฎ๐ง๐ž๐ฌ?
Las hormonas desempeรฑan un papel crucial. Los estrรณgenos, en particular, afectan la proporciรณn de linfocitos T CD4/CD8 al reducirla, incrementan la longevidad de las cรฉlulas B, potencian la liberaciรณn de anticuerpos y amplifican la expresiรณn de las proteรญnas HLA-II. En condiciones normales, este incremento en los niveles de anticuerpos en las mujeres potencia su resistencia frente a infecciones virales. Sin embargo, bajo circunstancias patolรณgicas, este equilibrio hormonal conduce a una prolongada supervivencia de las cรฉlulas B y a una disminuciรณn en las cรฉlulas T CD4, ademรกs de intensificar la expresiรณn del HLA-II. Esta situaciรณn propicia una mayor vulnerabilidad frente al EBV debido al aumento de la supervivencia de las cรฉlulas B transformadas por el virus y a la mayor expresiรณn del HLA-II, lo que facilita la infecciรณn de un mayor nรบmero de cรฉlulas. Sumado a esto, la elevada expresiรณn de HLA-II puede llevar a una presentaciรณn mรกs robusta de autoantรญgenos celulares o antรญgenos virales que, tras sufrir cambios post-traduccionales, generan neoantรญgenos. Estos pueden activar cรฉlulas autorreactivas. Por tanto, tanto el aumento de la supervivencia de las cรฉlulas B transformadas como el aumento de la presentaciรณn antigรฉnica generada por el aumento de la expresiรณn de HLA-II por el estradiol pueden favorecer un aumento en la presentaciรณn de antรญgenos tanto propios como extraรฑos durante un proceso infeccioso, y aquellas cรฉlulas plasmรกticas anormales que producen autoanticuerpos sobreviven mรกs tiempo, lo que en consecuencia aumenta la probabilidad de que las mujeres desarrollen enfermedades autoinmunes o incluso cรกncer.

Por el lado masculino, la testosterona modula el sistema inmunolรณgico al fomentar la respuesta CD4 Th1 (antiviral) y la activaciรณn de las cรฉlulas T CD8, mientras que inhibe la respuesta de las cรฉlulas NK y la expresiรณn del HLA-II. Dado que las cรฉlulas presentadoras de antรญgenos son esenciales para la diferenciaciรณn de las cรฉlulas T CD4 hacia Th1 o Th2, basรกndose en las citoquinas que liberan, las hormonas sexuales pueden influir en esta diferenciaciรณn. Las mujeres, al tener mayores niveles de anticuerpos (respuesta Th2), muestran una respuesta mรกs eficiente frente a infecciones extracelulares. No obstante, frente a patรณgenos intracelulares, su sistema inmunolรณgico podrรญa no ser tan eficiente como el masculino, que se beneficia de una respuesta celular Th1 mรกs potente para combatir cรฉlulas infectadas por virus.

ยฟ๐„๐ฌ๐ญ๐ž ๐ฆ๐จ๐๐ž๐ฅ๐จ ๐ž๐ฑ๐ฉ๐ฅ๐ข๐œ๐š ๐ฅ๐š ๐ซ๐ž๐ฌ๐ฉ๐ฎ๐ž๐ฌ๐ญ๐š ๐ข๐ง๐ฆ๐ฎ๐ง๐ž ๐ก๐ข๐ฉ๐ž๐ซ๐š๐œ๐ญ๐ข๐ฏ๐š ๐ฒ ๐š ๐ฅ๐š ๐ฏ๐ž๐ณ ๐ง๐จ ๐ž๐Ÿ๐ข๐œ๐š๐ณ?
Si, este modelo describe una situaciรณn en la que el sistema inmunolรณgico del cuerpo estรก trabajando en exceso, pero de manera ineficiente, frente al virus EBV:
Respuesta adaptativa deficiente: Las cรฉlulas T CD4, no estรกn reconociendo y enfrentรกndose correctamente a las cรฉlulas con latencia I del EBV. Esto lleva a que haya mรกs cรฉlulas infectadas circulando y, al mismo tiempo, a una fatiga o agotamiento de las cรฉlulas T, reduciendo su capacidad para luchar contra el virus.

๐’๐จ๐›๐ซ๐ž๐š๐œ๐ญ๐ข๐ฏ๐š๐œ๐ข๐จฬ๐ง ๐๐ž ๐ฅ๐š ๐ข๐ง๐ฆ๐ฎ๐ง๐ข๐๐š๐ ๐ข๐ง๐ง๐š๐ญ๐š: A causa de este fallo en la respuesta adaptativa, otra parte del sistema inmunolรณgico, llamada inmunidad innata, se activa en exceso porque detecta continuamente que hay una infecciรณn pero que no consigue ser resuelta por la inmunidad adaptativa. Esto se traduce en la producciรณn constante de sustancias inflamatorias que, en lugar de ayudar, pueden generar mรกs problemas y mantener una inflamaciรณn crรณnica.

๐ƒ๐ž๐ฌ๐›๐š๐ฅ๐š๐ง๐œ๐ž ๐ž๐ง ๐ฅ๐š๐ฌ ๐ซ๐ž๐ฌ๐ฉ๐ฎ๐ž๐ฌ๐ญ๐š๐ฌ ๐ข๐ง๐ฆ๐ฎ๐ง๐จ๐ฅ๐จฬ๐ ๐ข๐œ๐š๐ฌ: El cuerpo tiende a favorecer una respuesta inmunolรณgica (conocida como Th2) que no es la mรกs adecuada para combatir este tipo de infecciรณn. Esto se debe en parte a la producciรณn de una sustancia llamada IL-6, que redirige la respuesta defensiva del cuerpo hacia la producciรณn de anticuerpos en vez de una respuesta celular antiviral (Th1). El aumento de la respuesta Th2 favorece los ciclos de latencia y lรญtico del EBV al activarse mรกs cรฉlulas B.

ยฟ๐„๐ฌ๐ญ๐ž ๐ฆ๐จ๐๐ž๐ฅ๐จ ๐ž๐ฑ๐ฉ๐ฅ๐ข๐œ๐š ๐ฅ๐š๐ฌ ๐ซ๐ž๐š๐œ๐ญ๐ข๐ฏ๐š๐œ๐ข๐จ๐ง๐ž๐ฌ ๐ฏ๐ข๐ซ๐š๐ฅ๐ž๐ฌ ๐ฒ ๐ฅ๐š ๐๐ž ๐จ๐ญ๐ซ๐จ๐ฌ ๐ฉ๐š๐ญ๐จฬ๐ ๐ž๐ง๐จ๐ฌ ๐ฅ๐š๐ญ๐ž๐ง๐ญ๐ž๐ฌ?
Sรญ, el modelo explica que, debido a la disminuciรณn en la activaciรณn y funciรณn de las cรฉlulas T CD4 citotรณxicas, se produce una pรฉrdida de la vigilancia inmunolรณgica sobre las infecciones latentes de otros patรณgenos. Estas cรฉlulas T CD4 son necesarias para controlar las cรฉlulas con latencia o en fase lรญtica de patรณgenos como el Parvovirus B19, EBV, citomegalovirus y otros herpesvirus. Como resultado, se observarรก un aumento en la reactivaciรณn viral, especialmente en individuos con un grado mayor de inmunodeficiencia.

ยฟ๐„๐ฌ๐ญ๐ž ๐ฆ๐จ๐๐ž๐ฅ๐จ ๐ž๐ฑ๐ฉ๐ฅ๐ข๐œ๐š ๐œ๐จฬ๐ฆ๐จ ๐ฉ๐จ๐๐ซ๐ขฬ๐š ๐ก๐š๐›๐ž๐ซ ๐ฎ๐ง๐š ๐ฆ๐š๐ฒ๐จ๐ซ ๐ข๐ง๐ญ๐จ๐ฑ๐ข๐œ๐š๐œ๐ข๐จฬ๐ง ๐ฉ๐จ๐ซ ๐ฆ๐ž๐ญ๐š๐ฅ๐ž๐ฌ ๐ž๐ง ๐ž๐ฌ๐ญ๐จ๐ฌ ๐ฉ๐š๐œ๐ข๐ž๐ง๐ญ๐ž๐ฌ?
Si hay un aumento en la expresiรณn de MTs debido a niveles elevados de zinc intracelular, como se describe en el modelo, esas MTs estarรกn ocupadas uniendo y regulando el zinc y, potencialmente, el cobre. Como resultado, habrรญa menos MTs disponibles para unirse y desintoxicar los metales pesados que puedan estar presentes. Esto podrรญa llevar a una acumulaciรณn de metales pesados (como el cadmio y el mercurio) no regulados y potencialmente tรณxicos en el cuerpo.

ยฟ๐„๐ฌ๐ญ๐ž ๐ฆ๐จ๐๐ž๐ฅ๐จ ๐ž๐ฑ๐ฉ๐ฅ๐ข๐œ๐š ๐ž๐ฅ ๐š๐ฎ๐ฆ๐ž๐ง๐ญ๐จ ๐๐ž ๐ž๐ฌ๐ญ๐ซ๐žฬ๐ฌ ๐จ๐ฑ๐ข๐๐š๐ญ๐ข๐ฏ๐จ ๐ž๐ง ๐ž๐ฌ๐ญ๐š๐ฌ ๐ž๐ง๐Ÿ๐ž๐ซ๐ฆ๐ž๐๐š๐๐ž๐ฌ?
Sรญ, el modelo explica el aumento de estrรฉs oxidativo. Las estructuras linfoides ectรณpicas infectadas desencadenan respuestas inflamatorias al liberar ciertos componentes virales. Esta activaciรณn induce a la liberaciรณn de citocinas proinflamatorias, las cuales, a su vez, fomentan la producciรณn excesiva de especies reactivas de oxรญgeno, llevando a un marcado estrรฉs oxidativo. Ademรกs, la perturbaciรณn en la homeostasis de ciertos metales contribuye a la disrupciรณn de las respuestas antioxidantes intracelulares.
Especรญficamente, hay evidencia de que una enzima antioxidante (superรณxido dismutasa) es afectada por la alteraciรณn en la concentraciรณn de cobre y zinc. De forma resumida, en el modelo se describe cรณmo la combinaciรณn de respuestas inflamatorias crรณnicas, junto con desequilibrios en la homeostasis de ciertos metales y la liberaciรณn persistente de agentes proinflamatorios, culmina en un aumento significativo del estrรฉs oxidativo y nitrosativo en el cuerpo.

ยฟ๐„๐ฌ๐ญ๐ž ๐ฆ๐จ๐๐ž๐ฅ๐จ ๐ž๐ฑ๐ฉ๐ฅ๐ข๐œ๐š ๐ฅ๐š ๐ข๐ง๐ญ๐จ๐ฅ๐ž๐ซ๐š๐ง๐œ๐ข๐š ๐š๐ฅ ๐ž๐ฃ๐ž๐ซ๐œ๐ข๐œ๐ข๐จ ๐ฒ ๐ž๐ฅ ๐ฆ๐š๐ฅ๐ž๐ฌ๐ญ๐š๐ซ-๐ฉ๐จ๐ฌ๐ญ๐ž๐ฌ๐Ÿ๐ฎ๐ž๐ซ๐ณ๐จ?
Sรญ, este modelo explica la intolerancia al ejercicio y el malestar-postesfuerzo en el contexto de una infecciรณn persistente por EBV y sus interacciones metabรณlicas, inmunolรณgicas y neurofisiolรณgicas. A continuaciรณn, se desglosa cรณmo el modelo aborda este fenรณmeno:

๐€๐ฅ๐ญ๐ž๐ซ๐š๐œ๐ข๐จ๐ง๐ž๐ฌ ๐ฆ๐ž๐ญ๐š๐›๐จฬ๐ฅ๐ข๐œ๐š๐ฌ: El modelo describe cรณmo la infecciรณn por EBV puede llevar a una resistencia a la insulina a travรฉs de la producciรณn elevada de IFN-ฮณ. Esta resistencia, acompaรฑada de una hiperinsulinemia compensatoria, puede conducir a una hipoglucemia transitoria y una disminuciรณn en el metabolismo de los tejidos perifรฉricos. Estos factores contribuyen a la intolerancia al ejercicio, ya que los mรบsculos no pueden obtener la energรญa necesaria de manera eficiente, resultando en fatiga temprana.

๐€๐ฅ๐ญ๐ž๐ซ๐š๐œ๐ข๐จ๐ง๐ž๐ฌ ๐ž๐ง ๐ฅ๐š ๐Ÿ๐ฎ๐ง๐œ๐ข๐จฬ๐ง ๐œ๐š๐ซ๐๐ข๐จ๐ฏ๐š๐ฌ๐œ๐ฎ๐ฅ๐š๐ซ: Los altos niveles de serotonina y la activaciรณn de ciertos receptores, como TLR3 y TLR2, pueden alterar la funciรณn cardiovascular, afectando la distribuciรณn de sangre y la capacidad del cuerpo para satisfacer las demandas de oxรญgeno durante el ejercicio.

๐“๐ž๐ซ๐ฆ๐จ๐ซ๐ซ๐ž๐ ๐ฎ๐ฅ๐š๐œ๐ข๐จฬ๐ง ๐œ๐จ๐ฆ๐ฉ๐ซ๐จ๐ฆ๐ž๐ญ๐ข๐๐š: La capacidad del cuerpo para disipar el calor generado durante el ejercicio puede verse afectada, lo que podrรญa llevar a un sobrecalentamiento.

๐„๐ฌ๐ญ๐ซ๐žฬ๐ฌ ๐จ๐ฑ๐ข๐๐š๐ญ๐ข๐ฏ๐จ: La infecciรณn crรณnica con EBV genera un constante estrรฉs oxidativo, que puede deteriorar la funciรณn mitocondrial y reducir la capacidad del tejido muscular para generar energรญa eficientemente. Este estrรฉs oxidativo exacerbado durante el ejercicio puede llevar a daรฑo celular y fatiga muscular.

๐€๐ฅ๐ญ๐ž๐ซ๐š๐œ๐ข๐จ๐ง๐ž๐ฌ ๐ž๐ง ๐ฅ๐š ๐Ÿ๐ฎ๐ง๐œ๐ข๐จฬ๐ง ๐ซ๐ž๐ฌ๐ฉ๐ข๐ซ๐š๐ญ๐จ๐ซ๐ข๐š: La funciรณn respiratoria puede verse afectada, limitando la oxigenaciรณn adecuada durante el ejercicio y contribuyendo a la fatiga.

๐ˆ๐ง๐Ÿ๐ฅ๐š๐ฆ๐š๐œ๐ข๐จฬ๐ง ๐ฌ๐ข๐ฌ๐ญ๐žฬ๐ฆ๐ข๐œ๐š: La activaciรณn de ciertos receptores, la liberaciรณn de citocinas proinflamatorias y otros mecanismos asociados con la infecciรณn crรณnica pueden generar una inflamaciรณn sistรฉmica. Esta inflamaciรณn puede afectar negativamente la capacidad del cuerpo para recuperarse despuรฉs del ejercicio, contribuyendo al malestar-postesfuerzo.

๐€๐ฅ๐ญ๐ž๐ซ๐š๐œ๐ข๐จ๐ง๐ž๐ฌ ๐ž๐ง ๐ฅ๐š ๐Ÿ๐ฎ๐ง๐œ๐ข๐จฬ๐ง ๐ง๐ž๐ฎ๐ซ๐จ๐ฅ๐จฬ๐ ๐ข๐œ๐š: Los cambios metabรณlicos y la inflamaciรณn sistรฉmica pueden tener un impacto en el sistema nervioso central. La reducciรณn en la disponibilidad de serotonina y otras alteraciones pueden contribuir a la sensaciรณn de fatiga y letargo.

ยฟ๐„๐ฌ๐ญ๐ž ๐ฆ๐จ๐๐ž๐ฅ๐จ ๐ž๐ฑ๐ฉ๐ฅ๐ข๐œ๐š ๐ฅ๐š ๐๐ข๐ฌ๐š๐ฎ๐ญ๐จ๐ง๐จ๐ฆ๐ขฬ๐š ๐ฉ๐ซ๐ž๐ฌ๐ž๐ง๐ญ๐ž ๐ž๐ง ๐ž๐ฌ๐ญ๐จ๐ฌ ๐ฉ๐š๐œ๐ข๐ž๐ง๐ญ๐ž๐ฌ?
Si, a continuaciรณn, desgloso cรณmo el modelo puede generar disautonomรญa:

- ๐ˆ๐ง๐Ÿ๐ž๐œ๐œ๐ข๐จฬ๐ง ๐ฉ๐จ๐ซ ๐„๐๐•: La incapacidad para controlar adecuadamente las cรฉlulas de latencia I del EBV podrรญa resultar en respuestas inflamatorias crรณnicas, lo que perturba la homeostasis del sistema inmunolรณgico y, por extensiรณn, afecta al sistema nervioso autรณnomo (SNA).

-๐‘๐ž๐ฌ๐ฉ๐ฎ๐ž๐ฌ๐ญ๐š๐ฌ ๐ข๐ง๐Ÿ๐ฅ๐š๐ฆ๐š๐ญ๐จ๐ซ๐ข๐š๐ฌ: Las citocinas proinflamatorias liberadas en respuesta al EBV, como IL-1ฮฒ, IL-6 y TNF-ฮฑ, pueden actuar en el cerebro y otros รณrganos, alterando la funciรณn del SNA, lo que lleva a sรญntomas de disautonomรญa.

-๐€๐ฅ๐ญ๐ž๐ซ๐š๐œ๐ข๐จ๐ง๐ž๐ฌ ๐ฆ๐ž๐ญ๐š๐›๐จฬ๐ฅ๐ข๐œ๐š๐ฌ: La hipozincemia y las alteraciones en el transporte de cobre pueden desequilibrar la funciรณn de enzimas claves, como la DAO. Una funciรณn deficiente de la DAO lleva a una acumulaciรณn de histamina, un mediador que puede causar sรญntomas de disautonomรญa, como vasodilataciรณn y arritmias.

- ๐€๐ฅ๐ญ๐ž๐ซ๐š๐œ๐ข๐จ๐ง๐ž๐ฌ ๐ ๐š๐ฌ๐ญ๐ซ๐จ๐ข๐ง๐ญ๐ž๐ฌ๐ญ๐ข๐ง๐š๐ฅ๐ž๐ฌ: La acumulaciรณn de serotonina en el intestino puede estimular al nervio vago, una conexiรณn primordial entre el intestino y el cerebro. La sobreestimulaciรณn del nervio vago puede desencadenar sรญntomas de disautonomรญa, como bradicardia.

-๐€๐ฅ๐ญ๐ž๐ซ๐š๐œ๐ข๐จ๐ง๐ž๐ฌ ๐ง๐ž๐ฎ๐ซ๐จ๐ฅ๐จฬ๐ ๐ข๐œ๐š๐ฌ: Una disminuciรณn en la serotonina extracelular cerebral y un aumento en metabolitos neurotรณxicos del kynurenina pueden alterar la funciรณn neuronal y del SNA, lo que podrรญa manifestarse como fatiga, intolerancia al ejercicio y otros sรญntomas de disautonomรญa.

- ๐€๐ฅ๐ญ๐ž๐ซ๐š๐œ๐ข๐จ๐ง๐ž๐ฌ ๐ฏ๐š๐ฌ๐œ๐ฎ๐ฅ๐š๐ซ๐ž๐ฌ: La formaciรณn de microcoรกgulos puede afectar la perfusiรณn sanguรญnea adecuada en รณrganos y tejidos, incluyendo el cerebro. Una perfusiรณn inadecuada puede resultar en sรญntomas neurolรณgicos y autonรณmicos.

-๐€๐ฅ๐ญ๐ž๐ซ๐š๐œ๐ข๐จ๐ง๐ž๐ฌ ๐ž๐ง๐๐จ๐œ๐ซ๐ข๐ง๐š๐ฌ: La hiperinsulinemia y la posible reducciรณn en la secreciรณn de cortisol pueden afectar el equilibrio del SNA. Por ejemplo, la hipoglucemia puede desencadenar una respuesta simpรกtica aguda, mientras que la disminuciรณn del cortisol puede afectar la capacidad del cuerpo para manejar el estrรฉs.

ยฟ๐„๐ฌ๐ญ๐ž ๐ฆ๐จ๐๐ž๐ฅ๐จ ๐ž๐ฑ๐ฉ๐ฅ๐ข๐œ๐š ๐ฅ๐š ๐ง๐ž๐ฎ๐ซ๐จ๐ข๐ง๐Ÿ๐ฅ๐š๐ฆ๐š๐œ๐ข๐จฬ๐ง, ๐ฅ๐š ๐ง๐ข๐ž๐›๐ฅ๐š ๐ฆ๐ž๐ง๐ญ๐š๐ฅ ๐ฒ ๐ž๐ฅ ๐๐ž๐ญ๐ž๐ซ๐ข๐จ๐ซ๐จ ๐œ๐จ๐ ๐ง๐ข๐ญ๐ข๐ฏ๐จ?
Sรญ, este modelo presentado podrรญa explicar la neuroinflamaciรณn, la niebla mental y el deterioro cognitivo de la siguiente manera:

1๏ธโƒฃ๐๐ž๐ฎ๐ซ๐จ๐ข๐ง๐Ÿ๐ฅ๐š๐ฆ๐š๐œ๐ข๐จฬ๐ง: En pacientes con ME/CFS y LC, hay evidencia de infecciรณn viral crรณnica o reactivaciรณn de virus, especialmente EBV. Cuando hay presencia de material genรฉtico viral, especialmente EBERs del EBV, se activan los receptores TLR3 en la microglรญa (cรฉlulas inmunolรณgicas del sistema nervioso central). Esta activaciรณn resulta en la liberaciรณn de citocinas proinflamatorias como IL-1ฮฒ y TNF-ฮฑ. Estas citocinas pueden contribuir a la inflamaciรณn crรณnica del sistema nervioso central, caracterizando la neuroinflamaciรณn.

2๏ธโƒฃ๐๐ข๐ž๐›๐ฅ๐š ๐ฆ๐ž๐ง๐ญ๐š๐ฅ ๐ฒ ๐๐ž๐ญ๐ž๐ซ๐ข๐จ๐ซ๐จ ๐œ๐จ๐ ๐ง๐ข๐ญ๐ข๐ฏ๐จ: Varias vรญas mencionadas en el modelo pueden contribuir a estos sรญntomas. Por ejemplo:
-La infecciรณn viral puede causar daรฑo a la barrera hematoencefรกlica, permitiendo la entrada de material genรฉtico viral que podrรญa activar aรบn mรกs la microglรญa y contribuir a la neuroinflamaciรณn.

-El aumento de la actividad de IDO y la disminuciรณn de los niveles de triptรณfano llevan a una reducciรณn en la sรญntesis de serotonina (5-HT) y melatonina. Dado que la serotonina juega un papel en la regulaciรณn del estado de รกnimo, la cogniciรณn y la alerta, su reducciรณn podrรญa contribuir a la niebla mental.

-El รกcido quinolรญnico, un metabolito del triptรณfano, tiene propiedades neurotรณxicas al unirse al receptor NMDA, lo que puede aumentar el estrรฉs nitrosativo y contribuir al deterioro cognitivo.

-El aumento del estrรฉs oxidativo y nitrosativo en cรฉlulas infectadas por EBV, junto con la neuroinflamaciรณn, puede interferir con el correcto funcionamiento neuronal y contribuir al deterioro cognitivo.

-Las alteraciones en el sistema serotonรฉrgico tambiรฉn pueden afectar directamente la funciรณn cognitiva y la percepciรณn de fatiga.

๐‚๐จ๐ง๐ญ๐ข๐ง๐ฎฬ๐š ๐ž๐ฅ ๐Ÿงต๐Ÿ‘‡๐Ÿฝ

๐‹๐ข๐ง๐ค ๐„๐ฌ๐ญ๐ฎ๐๐ข๐จ:

#VirusEpsteinBarr #EBV #LongCovid #EMsfc #EncefalomielitisMialgica #Salud #investigacion #Noticias #microE2324 #Medicina #Virus #Microbiologรญa #EnfermedadesCronicas
@drmiguelmarcos @DoctorCasado @MasTwitts
Image Este modelo explica la apariciรณn de problemas digestivos, intolerancias alimentarias y alteraciones en la microbiota en estos pacientes?

Sรญ. A continuaciรณn, te resumo los puntos clave:
1๏ธโƒฃProblemas digestivos:
-La acumulaciรณn de 5-HT (serotonina) en la mucosa intestinal provoca inflamaciรณn crรณnica.
-Esta inflamaciรณn intestinal lleva a una disminuciรณn en la secreciรณn รกcida del estรณmago y en la expresiรณn de enzimas necesarias para digerir carbohidratos. Como resultado, mรกs carbohidratos llegan a la microbiota intestinal sin ser digeridos o absorbidos, lo que puede causar problemas digestivos como distensiรณn, gas y diarrea.
La alteraciรณn serotonรฉrgica, junto con el aumento de citocinas proinflamatorias, provoca una alteraciรณn en la secreciรณn รกcida, descomposiciรณn de la barrera intestinal y disminuciรณn en la expresiรณn de enzimas necesarias para digerir carbohidratos.
Sep 26, 2023 โ€ข 21 tweets โ€ข 4 min read
๐Ÿ”ฌ ๐„๐ฑ๐œ๐ข๐ญ๐ข๐ง๐  ๐ฎ๐ฉ๐๐š๐ญ๐ž! Findings from the @VirusesImmunity study support our review article on the relationship between EBV and symptoms in #LongCovid and #MECFS . I've detailed this connection below. ๐Ÿงฌ #ResearchUpdate #EBVLink. 1/ Many wonder why EBV is the main trigger for ME/CFS and Long COVID symptoms, over other factors such as other infections and metal poisoning. Let's dive into it, summarizing our recent review article. Link: link.springer.com/article/10.118โ€ฆ
Sep 18, 2023 โ€ข 48 tweets โ€ข 8 min read
๐Ÿ”ฅ๐Ÿงฌ NEW PAPER PUBLISHED! Is there a link between EBV (Epstein-Barr Virus) and conditions such as myalgic encephalomyelitis and Long COVID? ๐Ÿค”๐ŸŒ Dive into this thread and discover how EBV could be behind the symptoms of both pathologies and what therapeutic strategies exist: โฌ‡๏ธ๐Ÿ” Image 1/ ๐Ÿค” ๐ˆ๐ง๐ญ๐ซ๐จ๐๐ฎ๐œ๐ญ๐ข๐จ๐ง: ME/CFS and Long COVID are two pathologies that, although arising from different causes, present astonishing similarities in how they affect the body and their symptoms. #MyalgicEncephalomyelitis #COVIDSurvivors

Paper link: link.springer.com/article/10.118โ€ฆ
Aug 16, 2022 โ€ข 15 tweets โ€ข 9 min read
Thread ๐Ÿงต๐Ÿ‘‡:
After so many years of work and as a post Epstein-Barr virus infection #MECFS sufferer it makes me really happy that they have put our article on the cover of the August issue of Pathogens journal ๐Ÿ‘‰:mdpi.com/2076-0817/11/8
(1/) Until recently nobody talked about post-viral syndromes (actually they should be called chronic infectious syndromes) and they left all of us patients isolated both at a health and social level. Unfortunately it took another virus like #SARSCoV2 to show us how viruses are ...(2/)
Jul 26, 2022 โ€ข 23 tweets โ€ข 7 min read
A long thread ๐Ÿ‘‡๐Ÿผ:
I would like to share with you my latest review article. The Epstein-Barr virus (EBV) has long been known to be behind the development of autoimmune diseases, cancer and is even suspected to be behind the development of Chronic Fatigue Syndrome/โ€ฆ (1) โ€ฆMyalgic Encephalomyelitis and Long COVID. But it is still unclear what pathways it uses.

In this review I describe how the possession of certain ancestral HLA-II alleles (a system used by our immune system to recognize which proteins are foreign,โ€ฆ (2)
Jul 25, 2022 โ€ข 10 tweets โ€ข 2 min read
Te respondo con otro hilo ๐Ÿ˜…

La cuantificaciรณn del virus de Epstein-Barr utilizando la tรฉcnica PCR en sangre normalmente sale negativo en estos pacientes, puesto que el problema son las cรฉlulas con latencia del virus. (1) Hay que realizar la prueba en lugares donde haya acumulaciรณn de estas cรฉlulas, es decir, en tejido, por ejemplo, en la mucosa intestinal. (2)
Jul 25, 2022 โ€ข 25 tweets โ€ข 7 min read
Abro hilo largo๐Ÿ‘‡
Me gustarรญa compartir con vosotros mi รบltimo artรญculo de revisiรณn. Desde hace mucho tiempo se sabe que el virus de Epstein-Barr (EBV) estรก detrรกs del desarrollo enfermedades autoinmunes, cรกncer e incluso se sospecha que estรก detrรกs... (1) ...del desarrollo de Sรญndrome de Fatiga Crรณnica/Encefalomielitis Miรกlgica y del Long COVID. Pero todavรญa no estรก claro las vรญas que utiliza. En esta revisiรณn describo como la posesiรณn de ciertos alelos ancestrales de HLA-II... (2)