tirsdag den 1. september 2020

ZFR suppresses the antiviral response, but not in ME? Is macroH2A1/H2AFY involved?

ME peripheral blood mononuclear cell (PBMC) proteomes reveal decreased level of zinc finger RNA-binding protein (ZFR), foldchange = 0,52, p-value = 0,00047 (1).

ZFR suppresses interferon-beta induction and the antiviral response. ZFR controls interferon signaling by preventing aberrant splicing and nonsense-mediated decay (NMD) of histone variant macroH2A1/H2AFY mRNAs (2).

Serum from ME patients contains an activity which induces a powerful antiviral state, but it seems like the interferon response has been ruled out in the ME pathomechanism (3, 4).

However, macroH2A1 has also been shown to bind and repress interferon-stimulated genes (ISG) promoters, raising the possibility that the constitutive ISG induction in ZFR-depleted cells could be partially due to loss of direct action of macroH2A1 on these genes. Taking the available data together, macroH2A1 appears to both suppress the initiation of type I interferon signaling by inhibiting IFNB1 transcription and by modulating the consequences of IFNB1 transcription by directly repressing ISG promoters (2).

The gene macroH2A1/H2AFY is differentially methylated in PBMC from ME patients. Read the blogpost:

H2AFY and ME
https://followmeindenmark.blogspot.com/2018/10/h2afy-and-me.html

Is dysregulated macroH2A1/H2AFY involved in the ME pathomechanism?

MacroH2A1 can buffer transcriptional noise associated with the antiviral response

Knockdown of macroH2A causes a dramatic change in the antiviral gene expression program. Genes that normally do not respond to virus infection get activated or repressed (5).



References

preprint 1) Eiren Sweetman, Torsten Kleffmann, Christina Edgar, Michel de Lange, Rosamund Vallings Howick Warren Tate: A SWATH-MS analysis of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome peripheral blood mononuclear cell proteomes reveals mitochondrial dysfunction
https://assets.researchsquare.com/files/rs-52172/v1/6b319d19-80fb-46e3-b56e-3687ae1c7203.pdf

2) Haque N, Ouda R, Chen C, Ozato K, Hogg JR. ZFR coordinates crosstalk between RNA decay and transcription in innate immunity. Nat Commun. 2018;9(1):1145. Published 2018 Mar 20. doi:10.1038/s41467-018-03326-5
https://www.nature.com/articles/s41467-018-03326-5

3) Schreiner P, Harrer T, Scheibenbogen C, et al. Human Herpesvirus-6 Reactivation, Mitochondrial Fragmentation, and the Coordination of Antiviral and Metabolic Phenotypes in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Immunohorizons. 2020;4(4):201-215. Published 2020 Apr 23. doi:10.4049/immunohorizons.2000006
https://pubmed.ncbi.nlm.nih.gov/32327453/

4) Corbitt M, Eaton-Fitch N, Staines D, Cabanas H, Marshall-Gradisnik S. A systematic review of cytokines in chronic fatigue syndrome/myalgic encephalomyelitis/systemic exertion intolerance disease (CFS/ME/SEID). BMC Neurol. 2019;19(1):207. Published 2019 Aug 24. doi:10.1186/s12883-019-1433-0

5) Lavigne MD, Vatsellas G, Polyzos A, et al. Composite macroH2A/NRF-1 Nucleosomes Suppress Noise and Generate Robustness in Gene Expression. Cell Rep. 2015;11(7):1090-1101. doi:10.1016/j.celrep.2015.04.022

fredag den 28. august 2020

Atrogin 1 mRNA is elevated in muscle biopsies of ME patients

 Quote reference 1):
"Levels of Atrogin 1 mRNA were significantly elevated in muscle biopsies of patients with ME/CFS compared with HCs 
(Healthy Controls) suggesting an increase in protein degradation processes in muscles of patients with ME/CFS compared with HCs. Atrogin-1 binds to polyubiquitinated proteins to direct them for subsequent degradation by the 26S proteasome and as such is an important regulator of ubiquitin-mediated protein degradation in skeletal muscle (69). Increased levels of Atrogin-1 mRNA are associated with reduced muscle mass (69) and in this study Atrogin-1 was associated with a significant reduction in muscle fibre size, although a detailed examination of muscle protein degradation was not undertaken."


Atrogin 1, also known as muscle atrophy F-box (MAFbx), is encoded by FBXO32. Atrogin 1 degrades various muscle proteins, such as titin, nebulin, troponin, myosin-binding protein C, myosin light chains 1 and 2 and myosin heavy chains (2).

Increased degradation of myosin light chain proteins may lead to increased plasma N,N,N-trimethyl-L-alanyl-L-proline betaine (TMAP) (3). 


Read the blogpost:
Increased plasma N,N,N-trimethyl-L-alanyl-L-proline betaine in ME patients
http://followmeindenmark.blogspot.com/2020/02/


References:
preprint: 1) Arief Gusnanto, Kate Elizabeth Earl, George K Sakellariou, Daniel J Owens, Adam Lightfoot, Sandra A Fawcett, Euan Owen, Caroline A Staunton, Tu Shu, Fiona C Croden, Manuel Fenech, Melanie A Sinclair, Libuse Ratcliffe, Kasia A Whysall, Rebecca I Haynes, Nicola M Wells, Malcolm J Jackson, Graeme L Close, Clare L Lawton, Michael BJ Beadsworth, Louise Dye, Anne MCARDLE:
Discriminatory cytokine profiles predict muscle function, fatigue and cognitive function in patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS)
doi: https://doi.org/10.1101/2020.08.17.20164715
https://www.medrxiv.org/content/10.1101/2020.08.17.20164715v1

2) Steinbacher and Eckl: Impact of oxidative stress on exercising skeletal muscle. Biomolecules  2015 Apr 10;5(2):356-77.
doi: 10.3390/biom5020356.
https://pubmed.ncbi.nlm.nih.gov/25866921/

3) Velenosi, T.J., Thomson, B.K.A., Tonial, N.C. et al. Untargeted metabolomics reveals N, N, N-trimethyl-L-alanyl-L-proline betaine (TMAP) as a novel biomarker of kidney function. Sci Rep 9, 6831 (2019). https://www.nature.com/articles/s41598-019-42992-3
https://doi.org/10.1038/s41598-019-42992-3 PMID: 31048706 PMCID: PMC6497643

fredag den 31. juli 2020

Mutationer i muciner kan måske forklare øget immun aktivering og kemikalie intolerance hos ME patienter

Muciner er en gruppe glykoproteiner, som findes i slimhinderne i luftveje, tarm, skede urinrør og øjne.

Muciner sørger for at slimhinde-laget bliver tykt og højviskøst, så slimhinden bliver svær gennemtrængelig for virus, bakterier, skimmel og toxiner. Samtidig virker mucinerne som lokkemad for mikroorganismerne, der klistrer sig fast til sukkermolekylerne på mucinerne. Herved kan mikroorganismerne udstødes fra kroppen sammen med den slim, som kontinuerlig udskilles fra slimhinderne.

Under slimhindelaget findes et lag epitel med immunceller, som aktiveres hvis mikroorganismer eller toxiner alligevel slipper igennem slimhindelaget. Mutationer i muciner kan påvirke slimhindernes beskaffenhed, således at der sker en større indtrængning af mikroorganismer og toxiner til epitelet. Herved vil immunforsvaret hyppigt aktiveres (1).

Muciner og ME
Der er fundet mutationer i muciner hos ME patienter, og det danner baggrund for en ny hypotese: 

Mutations in ME/CFS nasal mucins result in compromised protective barriere

  • an altered nasal microbiome
  • a highly sensitive and irritated epithelial layer
  • chronic low grade non-allergic inflammatory response
  • exacerbation of ME/CFS symptomatology


Hypotesen er fremlagt i et webinar. Indslaget begynder til tiden cirka 1 time og 55 minutter:
INIM Webinar - Understanding ME/CFS Today: A Clinical & Research Approach

https://www.youtube.com/watch?v=QfrCF2atQxI&feature=youtu.be&t=8591

Forskerne bag studiet oplyser, at en mutation i mucin19 (MUC19) har særlig betydning for en beskadiget slimhinden i næse/øvre luftveje. Dette kan medføre øget følsomhed over for de kemikalier, som vi indånder. Det betyder at mutiple chemical sensitivity (MCS) kan forklares med hypotesen. MUC19 mutationen er nævnt i reference 2. 


Den nye mucin hypotese er forenelig med anden ME forskning: 

Der er tidligere fundet mutationer i IDO2 genet hos meget syge ME patienter, og det danner baggrund for IDO ME hypotesen (3). IDO har betydning for nasal tolerance (4).
Læs blog: IDO-ME hypotesen er forenelig med AHR-MCS hypotesen
http://followmeindenmark.blogspot.com/2019/06/ido-me-hypotesen-er-forenelig-med-ahr.html

Det er tidligere vist, at ME patienter har nedsat sygdomtolerance, og nasal stimulation kan afhjælpe ME symptomer (5).
Læs blog: Det inflammatoriske respons og induktion af sygdomstolerance er dysreguleret hos ME patienter
http://followmeindenmark.blogspot.com/2020/03/det-inflammatoriske-respons-og.html

Proteinet cathelicidin antimicrobial peptide (CAMP) kan påvises ved inflammatorisk respons. Det er f.eks. påvist i tarmens slimhinde hos patienter med inflammatory bowel disease (IBD). Plasma niveauet af CAMP er for forhøjet hos ME patienter (6).

Mere viden om CAMP:
Topical cathelicidin (LL-37) an innate immune peptide induces acute olfactory epithelium inflammation in a mouse model https://pubmed.ncbi.nlm.nih.gov/28837619/


Læs også om mucin studiet på Health Rising bloggen:
Surprise Gene Finding Could Give Pathogens and Toxins a Leg Up in ME/CFS

https://www.healthrising.org/blog/2020/07/11/mucosal-genes-chronic-fatigue-syndrome/


Referencer:
1) S K Linden, P Sutton, N G Karlsson, V Korolik, M A McGuckin: Mucins in the mucosal barrier to infection. Mucosal Immunol 2008 May;1(3):183-97. doi: 10.1038/mi.2008.5. Epub 2008 Mar 5. https://pubmed.ncbi.nlm.nih.gov/19079178/

2) Melanie Perez et al: Genetic Predisposition for Immune System, Hormone, and Metabolic Dysfunction in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Pilot Study Front Pediatr. 2019 May 24;7:206. doi: 10.3389/fped.2019.00206. eCollection 2019. https://pubmed.ncbi.nlm.nih.gov/31179255/


3) Kashi AA Davis RW and, Phair RD: The IDO Metabolic Trap Hypothesis for the Etiology of ME/CFS. Diagnostics (Basel). 2019 Jul 26;9(3). pii: E82. doi: 10.3390/diagnostics9030082. https://www.mdpi.com/2075-4418/9/3/82

4) van der Marel AP1, Samsom JN, Greuter M, van Berkel LA, O'Toole T, Kraal G, Mebius RE. Blockade of IDO inhibits nasal tolerance induction. J Immunol. 2007 Jul 15;179(2):894-900.
https://www.ncbi.nlm.nih.gov/pubmed/17617580

5) Lucie S.T. Rodriguez, Christian Pou, Tadepally Lakshmikanth, Jingdian Zhang, Constantin Habimana Mugabo, Jun Wang, Jaromir Mikes, Axel Olin, Yang Chen, Joanna Rorbach, Jan-Erik Juto, Tie Qiang Li, Per Julin, Petter Brodin: Achieving symptom relief in patients with Myalgic encephalomyelitis by targeting the neuro-immune interface and inducing disease tolerance
doi: https://doi.org/10.1101/2020.02.20.958249
https://www.biorxiv.org/content/10.1101/2020.02.20.958249v1.abstract


6) Milica Milivojevic et al: Plasma proteomic profiling suggests an association between antigen driven clonal B cell expansion and ME/CFS. PLoS One . 2020 Jul 21;15(7):e0236148. doi: 10.1371/journal.pone.0236148. eCollection 2020. https://pubmed.ncbi.nlm.nih.gov/32692761/

fredag den 29. maj 2020

Kynurenine skal afprøves til behandling af ME patienter

Open Medicine Foundation finansierer et nyt placebo-kontrolleret studie, hvor ME patienter skal behandles med kynurenine:

Kynurenine Clinical Trial for ME/CFS
https://omfcanada.ngo/kynurenine-clinical-trial-for-me-cfs/


Jonas Bergquist, MD, PhD, direktør for det svenske ME/CFS forskningscenter i Uppsala, skal stå for studiet.

Baggrunden for studiet er Dr. Robert Phair's ME- hypotese "den metaboliske fælde", der beskriver, at ME patienter ikke omsætter aminosyren tryptofan i tilstrækkelig omfang i nogle af vores celletyper (1). Blogindlæg om den metaboliske fælde:

ME hypotese: The Metabolic Trap - den metaboliske fælde
https://followmeindenmark.blogspot.com/2019/06/me-hypotese-metabolic-trap-den.html

Kynurenine metabolitter opstår ved omsætning af tryptofan, og disse metabolitter er vigtige for normal funktion af bl.a. hjerne og immunforsvar. Dette er vist i dette blogindlæg: 


Tryptofan metabolitten kynureninsyre har immunmodulerende egenskaber
https://followmeindenmark.blogspot.com/2019/06/tryptofan-metabolitten-kynureninsyre.html


Yderligere blogindlæg om kynurenine og den metaboliske fælde:

Is the ME hypothesis "the metabolic trap" able to explain endothelial dysfunction?
https://followmeindenmark.blogspot.com/2019/06/is-me-hypothesis-metabolic-trap-able-to.html

Kynurenine metabolisme påvirkes af motion
https://followmeindenmark.blogspot.com/2019/06/kynurenine-metabolisme-pavirkes-af.html

Omsætning af tryptofan og forgrenede aminosyrer hos ME patienter under motion
https://followmeindenmark.blogspot.com/2019/06/omstning-af-tryptofan-og-forgrenede.html

Is the kynurenic acid responsive Gpr35 involved in the ME pathomechanism?
https://followmeindenmark.blogspot.com/2019/06/is-kynurenic-acid-responsive-gpr35.html

IDO-ME hypotesen er forenelig med AHR-MCS hypotesen
https://followmeindenmark.blogspot.com/2019/06/ido-me-hypotesen-er-forenelig-med-ahr.html

Mutations in the IDO2 gene and DNA methylations in genes in the NAD/NADP synthesis pathway in ME
https://followmeindenmark.blogspot.com/2019/07/mutations-in-ido2-gene-and-dna.html


Reference: 
1) Kashi AA Davis RW and, Phair RD: The IDO Metabolic Trap Hypothesis for the Etiology of ME/CFS. Diagnostics (Basel). 2019 Jul 26;9(3). pii: E82. doi: 10.3390/diagnostics9030082. https://www.mdpi.com/2075-4418/9/3/82


mandag den 4. maj 2020

Serum fra ME patienter udviser antiviral aktivitet

Et nyt studie af Schreiner et al har vist, at serum fra ME patienter udviser antiviral aktivitet mod både DNA- og RNA-virus i en cellekultur. Samtidig blev det observeret, at serummet inducerer et fragmenteret mitokondriel netværk og nedsat cellulær ATP produktion (1).

Citat fra artiklen (1), side 213: 
"... we showed that serum from ME/CFS patients contained an activity that produced mitochondrial fragmentation, decreased mitochondrial ATP production, and induced a powerful antiviral state."

Det er ukendt, hvad det er i serum, som udløser og udøver den antivirale aktivitet. Schreiner et al har en hypotese om, at en delvis aktivering af herpesvirus (HHV-6) kan være årsag til den kroniske immunaktivering hos ME patienter. Forskerne understøtter hypotesen med et forsøg, hvor en cellekultur udsat for delvis aktivering af HHV-6 udskilder et stof som har samme effekt som serum fra ME patienter (1). 

Citat fra artiklen (1), side 206:
"...These results showed that cells containing latent HHV-6A DNA that had been transactivated by TSA (trichostatin-A) secreted a potent activity that could be adoptively transferred and induce mitochondrial fragmentation and a proinflammatory CDR (cell danger response) in naive responder cells, conferring strong protection from both DNA and RNA virus infections."

ME er tidligere sat i forbindelse med kronisk herpes infektion (2).

Schreiner et al foreslår, at nye forsøg skal vise om ME patienter har en produktion af HHV-6 proteiner (ikke hele, levende HHV-6).

Citat fra artiklen (1), side 213:
"Larger multicohort studies involving ME/CFS patients from different age groups should be carried out in the future and should include methods for detecting and quantifying both productive and nonproductive (incomplete) viral reactivation events. Furthermore, potential factors affecting mitochondrial dynamics in ME/CFS patients should be systematically evaluated for their ability to induce a powerful antiviral state."


Læs også:

For ME/CFS Patients, Viral Immunities Come at a Devastating, Lifelong Cost
https://health.ucsd.edu/news/releases/Pages/2020-04-27-for-me-cfs-patients-viral-immunities-come-at-lifelong-cost.aspx

Explaining ME/CFS? Prusty / Naviaux Study Ties Infections to Energy Breakdowns
https://www.healthrising.org/blog/2020/04/26/explaining-chronic-fatigue-syndrome-naviaux-prusty/

Viden om fragmenteret mitokondriel netværk:
Mitochondria in Innate Immune Responses
https://pubmed.ncbi.nlm.nih.gov/21597473/
Figur: https://pubmed.ncbi.nlm.nih.gov/21597473/#&gid=article-figures&pid=figure-2-uid-1


Referencer:

1) Philipp Schreiner, Thomas Harrer, Carmen Scheibenbogen, Stephanie Lamer, Andreas Schlosser, Robert K. Naviaux and Bhupesh K. Prusty
Human Herpesvirus-6 Reactivation, Mitochondrial Fragmentation, and the Coordination of Antiviral and Metabolic Phenotypes in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome
Immunohorizons. 2020 Apr 23;4(4):201-215.
DOI: 10.4049/immunohorizons.2000006
PMID: 32327453 

2) Nuno Sepúlveda, Jorge Carneiro,  Eliana Lacerda, Luis Nacul Myalgic Encephalomyelitis/Chronic Fatigue Syndrome as a Hyper-Regulated Immune System Driven by an Interplay Between Regulatory T Cells and Chronic Human Herpesvirus Infections
Front Immunol 2019 Nov 21;10:2684. doi: 10.3389/fimmu.2019.02684. eCollection 2019.
https://www.frontiersin.org/articles/10.3389/fimmu.2019.02684/full

mandag den 20. april 2020

Kan POTS patienter være mindre sårbare over for COVID-19 pga mindre ACE2 aktivitet?

Coronavirus anvender et af menneskets enzymer til at komme ind i cellerne. Enzymet hedder angiotensin converting enzyme 2 (ACE2).

ACE-hæmmere er lægemidler, der anvendes til behandling af for højt blodtryk. 

Der er fremsat en hypotese om at ACE-hæmmere kan øge ACE2 og hermed fremme indtrængning af coronavirus i cellerne. Læs om emnet her: 

ACE-hæmmere øger muligvis risikoen for at dø af COVID-19
Danske forskere vil undersøge årsag til visse patienters coronadød
https://sundhedspolitisktidsskrift.dk/nyheder/3250-er-patienter-i-bestemt-behandling-i-storre-risiko-for-at-blive-alvorligt-syge-af-coronavirus.html 


COVID-19, ACE2, and the Cardiovascular Consequences
https://pubmed.ncbi.nlm.nih.gov/32228252/

Renin-Angiotensin-Aldosterone System: Double-Edged Sword in COVID-19 Infection
https://www.preprints.org/manuscript/202003.0365/v1

Der er også fremsat en hypotese om, at lupus patienter måske har øget sårbarhed over for COVID-19 på grund af mulig øget ekspression af ACE2:
"...we provide evidence in lupus to suggest hypomethylation and overexpression of ACE2..."  
Citat fra: Epigenetic Dysregulation of ACE2 and Interferon-Regulated Genes Might Suggest Increased COVID-19 Susceptibility and Severity in Lupus Patients
https://pubmed.ncbi.nlm.nih.gov/32276140/


Der er så vidt jeg ved IKKE fremsat hypotese om COVID-19  og patienter med Postural Ortostatisk Takykardi Syndrom (POTS). Men jeg vil henlede opmærksomheden på viden fra to artikler, der omhandler POTS og ACE2:

Defects in Cutaneous Angiotensin-Converting Enzyme 2 and angiotensin-(1-7) Production in Postural Tachycardia Syndrome
https://pubmed.ncbi.nlm.nih.gov/19289653/

Abnormalities of Angiotensin Regulation in Postural Tachycardia Syndrome
https://pubmed.ncbi.nlm.nih.gov/21266211/

Citat fra sidstnævnte artikel:
"Estimated angiotensin-converting enzyme-2 (ACE2) activity was significantly lower in POTS patients than in controls (0.25 ± 0.02 vs 0.33 ± 0.03, P = .038)."

Kan POTS patienter være mindre sårbare over for COVID-19 pga mindre ACE2 aktivitet? Eller vejer ulempen ved at have en kronisk sygdom tungere på vægtskålen?

torsdag den 5. marts 2020

ME - a failure of inducing exercise tolerance?

ME hypothesis from Karolinska Institutet and Karolinska University Hospital in Sweden (1):

"ME - a failure of inducing disease tolerance upon chronic immune activation"

My question:
Is ME also a failure of inducing exercise tolerance?

At cellular level disease and inflammation result in increased reactive oxygen species (ROS) production. Stress-response pathways are countermeasures to ROS. Exercise also leads to increased ROS levels. The antioxidant enzyme superoxide dismutase 2 (SOD2) is one the primary mechanisms against ROS generated during exercise.

Quote from a review "Impact of oxidative stress on exercising skeletal muscle" (2):
"It is well established that muscle contractions during exercise lead to elevated levels of reactive oxygen species (ROS) in skeletal muscle. These highly reactive molecules have many deleterious effects, such as a reduction of force generation and increased muscle atrophy. Since the discovery of exercise-induced oxidative stress several decades ago, evidence has accumulated that ROS produced during exercise also have positive effects by influencing cellular processes that lead to increased expression of antioxidants. These molecules are particularly elevated in regularly exercising muscle to prevent the negative effects of ROS by neutralizing the free radicals. In addition, ROS also seem to be involved in the exercise-induced adaptation of the muscle phenotype.

Chronic oxidative stress is associated with an increase in protein loss and muscle atrophy. High ROS levels cause a sustained activation of NF-κB and of FoxO which then activate two muscle-specific E3 ubiquitin ligases, atrogin-1 or muscle atrophy F-box (MAFbx) and muscle RING (Really Interesting New Gene)-finger protein 1 (MuRF-1) [52]. MAFbx and MuRF-1 then degrade various proteins, such as titin, nebulin, troponin, myosin-binding protein C, myosin light chains 1 and 2 and myosin heavy chain [53,54]. Recently, it was demonstrated that excessive oxidative stress also enhances the transcription factor C/EBP homology protein (CHOP). This transcription factor also enhances expression of MuRF1, which again results in increased protein degradation [35]."

It seems like ME patients have increased muscle protein degradation:

Increased serum and urine 3-methylhistidine in ME patients
http://followmeindenmark.blogspot.com/2020/03/increased-serum-and-urine-3.html

Increased plasma N,N,N-trimethyl-L-alanyl-L-proline betaine in ME patients
https://followmeindenmark.blogspot.com/2020/02/increased-plasma-nnn-trimethyl-l-alanyl.html

Proline, P5C and 4-hydroxyglutamate in ME
http://followmeindenmark.blogspot.com/2020/02/proline-p5c-and-4-hydroxyglutamate-in-me.html

And do remember the transcription profile analysis of skeletal muscle from ME patients (3), quote:
"In an effort to establish which pathways might be involved in the onset and development of muscle symptoms, we used global transcriptome analysis to identify genes that were differentially expressed in the vastus lateralis muscle of female and male CFS patients. We found that the expression of genes that play key roles in mitochondrial function and oxidative balance, including superoxide dismutase 2, were altered, as were genes involved in energy production, muscular trophism and fiber phenotype determination. Importantly, the expression of a gene encoding a component of the nicotinic cholinergic receptor binding site was reduced, suggesting impaired neuromuscular transmission. We argue that these major biological processes could be involved in and/or responsible for the muscle symptoms of CFS."


Are the inflammatory reponse to disease and the adaptive response to exercise dysregulated in ME through the same pathways?

Reference

1) Lucie S.T. Rodriguez, Christian Pou, Tadepally Lakshmikanth, Jingdian Zhang, Constantin Habimana Mugabo, Jun Wang, Jaromir Mikes, Axel Olin, Yang Chen, Joanna Rorbach, Jan-Erik Juto, Tie Qiang Li, Per Julin, Petter Brodin: Achieving symptom relief in patients with Myalgic encephalomyelitis by targeting the neuro-immune interface and inducing disease tolerance
doi: https://doi.org/10.1101/2020.02.20.958249
https://www.biorxiv.org/content/10.1101/2020.02.20.958249v1.abstract

2) Peter Steinbacher, Peter Eckl:
Impact of Oxidative Stress on Exercising Skeletal Muscle

tirsdag den 3. marts 2020

Det inflammatoriske respons og induktion af sygdomstolerance er dysreguleret hos ME patienter

Forskere fra Karolinska Instituttet og Karolinska Universitetshospital har beskrevet en ME hypotese (1):

"ME - a failure of inducing disease tolerance upon chronic immune activation"

Sygdomstolerance er en overordnet betegnelse for en række stress-respons stiveje. Disse skal sørge for at begrænse skade fra patogene mikroorganismer og fra kroppens inflammationsproces.

Vagusnerve og hjernestamme regulerer systemisk inflammation ved at skrue op eller ned for den inflammatoriske refleks. Nerver der fører fra kroppen (afferent vagus nerve endings) til hjernestamme sender besked om immunstatus i kroppen. Nerverne kan f.eks. sende besked om aktivitet fra patogene mikroorganismer i tarmen. Nerver der fører fra hjernestamme (efferente) til kroppen  sender signaler via acetylcholin om at dæmpe inflammationen. 

Det er kendt viden, at påvirkning af vagusnerven kan dæmpe inflammatoriske tilstande.

Forskerne fra Karolinska afprøvede Intranasal Mekanisk Stimulation (INMEST) på en gruppe ME patienter. INMEST udstyret består af et tyndt plasticrør, som placeres i næsen. Udstyret vibrerer og danner en turbulent luftstrøm. Dette inducerer en nervereflex, som påvirker vagusnerven. Behandlingen dæmpede nogle ME symptomer (men ikke fatigue), og blodprøver fra patienterne viste normalisering af inflammatoriske tilstande (1).

En analyse af sygdomstoleranceprogrammer viste, at INMEST behandling opregulerede disse programmer.

Forskerene påpegede, at årsagen til den manglende induktion af tolerance hos ME patienter skal klarlægges. Samtidig henviser de til forskningen i IDO2 enzymet, som netop er involveret i regulering af sygdomstolerance.



Mere viden om emnet kan findes her:

Neural Reflexes in Inflammation and Immunity
https://pubmed.ncbi.nlm.nih.gov/22665702-neural-reflexes-in-inflammation-and-immunity/

The Vagus Nerve and the Inflammatory Reflex--Linking Immunity and Metabolism
https://pubmed.ncbi.nlm.nih.gov/23169440-the-vagus-nerve-and-the-inflammatory-reflex-linking-immunity-and-metabolism/

Er der behandlingsmuligheder i lægemidler, der øger aktivitet i acetylcholin-stiveje?:

Central and peripheral anti-inflammatory effects of acetylcholinesterase inhibitors
https://www.sciencedirect.com/science/article/abs/pii/S0028390820300861

Aryl Hydrocarbon Receptor Control of a Disease Tolerance Defence Pathway
https://pubmed.ncbi.nlm.nih.gov/24930766-aryl-hydrocarbon-receptor-control-of-a-disease-tolerance-defence-pathway/



Relevante blogindlæg:

Indånding af skimmel er noget, der kan gøre en ME/MCS patient ganske syg. IDO er med til at inducere tolerance i næsens slimhinder, og mutationer i IDO-generne har betydning for kroppens reaktion på skimmel infektion (AHR = Arylhydrocarbon receptor, MCS = multiple chemical sensitivity): 

IDO-ME hypotesen er forenelig med AHR-MCS hypotesen

Tryptofan metabolitten kynureninsyre har immunmodulerende egenskaber



Reference

1) Lucie S.T. Rodriguez, Christian Pou, Tadepally Lakshmikanth, Jingdian Zhang, Constantin Habimana Mugabo, Jun Wang, Jaromir Mikes, Axel Olin, Yang Chen, Joanna Rorbach, Jan-Erik Juto, Tie Qiang Li, Per Julin, Petter Brodin: Achieving symptom relief in patients with Myalgic encephalomyelitis by targeting the neuro-immune interface and inducing disease tolerance
doi: https://doi.org/10.1101/2020.02.20.958249
https://www.biorxiv.org/content/10.1101/2020.02.20.958249v1.abstract

mandag den 2. marts 2020

Increased serum and urine 3-methylhistidine in ME patients

3-metylhistidine is a breakdown product of muscle contractile proteins. Serum or urine 3-methylhistidine is used as a biomarker of muscle protein degradation (1).

Fluge et al found increased serum 3-metylhistidine in male ME patients (2).

McGregor et al found increased urine 3-metylhistidine in post-exertional malaise (PEM) ME patients (1).

Quote from McGregor et al (1):
"The findings that the PEM is associated with a loss of metabolites, reduction in acetylation, deregulation of purine metabolism, increased contractile protein breakdown and bacteremia associated with exercise suggest that treatments such as graded exercise may be more detrimental than beneficial as claimed in some studies [39,40]. Until such time as these biological changes can be further investigated, the use of graded exercise as a therapy for those with severe forms of ME/CFS should be considered potentially harmful. In support of this, the use of graded exercise therapy has caused significant protest by ME/CFS sufferers as they see it as harmful [41,42]."


Further reading about possible breakdown of skeletal muscles in ME patients :

Proline, P5C and 4-hydroxyglutamate in ME
http://followmeindenmark.blogspot.com/2020/02/proline-p5c-and-4-hydroxyglutamate-in-me.html

Increased plasma N,N,N-trimethyl-L-alanyl-L-proline betaine in ME patients
https://followmeindenmark.blogspot.com/2020/02/increased-plasma-nnn-trimethyl-l-alanyl.html




Referencer
1) McGregor et al: Post-Exertional Malaise Is Associated with Hypermetabolism, Hypoacetylation and Purine Metabolism Deregulation in ME/CFS Cases Diagnostics (Basel). 2019 Jul 4;9(3). pii: E70. doi: 10.3390/diagnostics9030070. https://www.ncbi.nlm.nih.gov/pubmed/31277442

2) Fluge et al: Metabolic profiling indicates impaired pyruvate dehydrogenase function in myalgic encephalopathy / chronic fatigue syndrome. JCI Insight. 2016; 1(21):e89376. Doi 10.1172/jci.insight.89276

onsdag den 19. februar 2020

Increased plasma N,N,N-trimethyl-L-alanyl-L-proline betaine in ME patients

N,N,N-trimethyl-L-alanyl-L-proline betaine (TMAP) is a plasma biomarker of reduced kidney function (1).

Quote from ref 1:
"TMAP was the most consistently cleared metabolite by all hemodialysis modalities in our untargeted metabolomics analysis. Although the biological origin of TMAP has not been identified, we suggest that TMAP may be produced from degradation of myosin light chain (MYL) proteins. N,N,N-trimethylalanine is mainly found in myosin light chain (MYL) proteins and in each of the four MYL isoforms (MYL1, MYL2, MYL3, and MYL4), the c-terminus of N,N,N-trimethylalanine forms a peptide bond with proline (26). Therefore, MYL protein degradation may be responsible for the release of TMAP. Further study is necessary to determine the biological origin and potential physiological effects of TMAP."

Myosin light chains are components of macromcular myosin complexes. Fx. myosin II is the myosin type responsible for producing muscle contractions in muscle cells. Myosin II contains two heavy chains and four ligtht chains. ( Wikipedia. Myosin )

Plasma TMAP was increased in ME patients in Germain et al's metabolomic study (2020) (2).

As far as I know, ME patients have normal kidney function, so why is TMAP increased? Is the turnover of myosin light chains increased? And/or do the increased TMAP level reflect the breakdown of skeletal muscles?


Further reading about possible breakdown of skeletal muscles in ME patients :

Proline, P5C and 4-hydroxyglutamate in ME



References:
1) Velenosi, T.J., Thomson, B.K.A., Tonial, N.C. et al. Untargeted metabolomics reveals N, N, N-trimethyl-L-alanyl-L-proline betaine (TMAP) as a novel biomarker of kidney function. Sci Rep 9, 6831 (2019). https://www.nature.com/articles/s41598-019-42992-3
https://doi.org/10.1038/s41598-019-42992-3 PMID: 31048706  PMCID: PMC6497643

2Arnaud Germain , Dinesh K Barupal , Susan M Levine , Maureen R HansonComprehensive Circulatory Metabolomics in ME/CFS Reveals Disrupted Metabolism of Acyl Lipids and Steroids. Metabolites 2020, 10(1), 4; https://doi.org/10.3390/metabo10010034
https://www.mdpi.com/2218-1989/10/1/34

onsdag den 5. februar 2020

Proline, P5C and 4-hydroxyglutamate in ME

What do

  • Pre-eclampsia
  • The inborn error of metabolism: Primary Hyperoxaluria Type 3
  • Myalgic encephalomyelitis (ME)

have in common?

Increased levels of 4-hydroxyglutamate (1, 2, 3).

Germain et al's (2020) new metabolomic study found increased level of plasma 4-hydroxyglutamate  in ME patients compared to controls (3).

From figure S1 in reference 3. 4-hydroxyglutamate. Box plot distribution of logged values from table 2 in reference 3. Controls are shown in red and ME patients in blue. The yellow diamond represents the mean. https://www.mdpi.com/2218-1989/10/1/34


Proline and its metabolite hydroxyproline (OH-proline) are amino acids. They constitute one-third of the amino acids in collagen proteins.

High levels of 4-hydroxyglutamate could result from increased collagen turnover and the release of proline and 4-hydroxyproline. The latter is metabolized to 4-hydroxyglutamate. Proline is re-used and 4-hydroxyproline is broken down and excreted in this pathway (4):



Figure 1 from Riedel et al, reference 4: Metabolism of 4-hydroxyproline and glyoxylate.
Four mitochondrial enzymes are responsible for 4-hydroxproline (4-Hyp) breakdown: hydroxyproline oxidase (HPOX), Δ1-pyrroline-5-carboxylate dehydrogenase (1P5CDH), aspartate aminotransferase (AspAT), and 4-hydroxy-2-oxoglutarate aldolase (HOGA). The terminal HOGA reaction cleaves 4-hydroxy-2-oxoglutarate (HOG) into pyruvate and glyoxylate. Glyoxylate is metabolized either to glycolate by glyoxylate reductase (GR) in the mitochondria and cytoplasm or to glycine by peroxisomal alanine-glyoxylate aminotransferase (AGT). AGT and GR are mutated within primary hyperoxaluria (type 1 and 2, respectively) patients resulting in the buildup of glyoxylate and its conversion by lactate dehydrogenase (LDH) to oxalate, a key component of kidney stones.
https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0026021

Naviaux et al showed increased plasma level of hydroxyproline in female ME patients (5).

Primary hyperoxaluria type 3 with increased urine 4-hydroxyglutamate is caused by mutations in the HOGA1 gene (4, 6).

Increasd levels of 4-hydroxyglutamate could also result from decreased activity of glutamic-oxaloacetic transaminase 2 (GOT2 = AspAT in figure 1 from ref 4) (1).

Schutzer et al showed decreased level of GOT2 precursor in cerebrospinal fluid from ME patients. Number of unique peptides in cerebrospinal fluid (table S1 in ref. 7): 
1) Controls: 6
2) ME patients: 1
3) Post treatment Lyme patients: 8

Why is 4-hydroxyglutamate increased in ME?


Is the proline-P5C cycle dysregulated in ME?

Naviaux et al showed increased plasma level of 1-pyrroline-5-carboxylic acid  (P5C) in ME patients (5).

A unique aspect of proline metabolism is the cycling of proline and P5C to maintain redox homeostasis between the cytosol and mitochondria. Proline biosynthesis, catabolism, and cycling, known as "the proline-P5C cycle" have been implicated as metabolic pathways selectively altered in cancer cells providing ATP, macromolecules, and redox cofactors (8).

The proline metabolism plays an important role in metabolic reprogramming, not only in cancer but also in related fields such as aging, senescence, and development (9).


The proline-P5C cycle: 


Figure 3 from reference 9. Hypothesis for proline cycle revised.
The cycle has been revised according to locations of the enzymes. The colored areas are for emphasis and do not represent specific locations. The dotted arrows represent putative shuttle systems, for example, malate/aspartate shuttle. PYCR1/2/L, pyrroline-5-carboxylate reductase 1/2/L.

Quote from reference 9: 
"The enzyme that oxidizes proline to P5C is tightly bound to mitochondrial inner membranes (30, 37, 38) and is linked to site II of the mitochondrial electron transport chain (30, 61) with a flavine adenine dinucleotide at the active site, which transfers electrons from proline to coenzyme Q (30, 95); at site III, proline-derived electrons have two dispositions. They can be transferred to cytochrome c, which is oxidized at complex IV with electrons transferred to O2 to form H2O. On the contrary, proline-derived electrons can directly reduce dissolved oxygen at complex III to form superoxide (23, 30). Since complex III has access to both the matrix space and the intermembrane space, ROS can evolve in the mitochondrial matrix or in the intermembrane space to be transferred into the cytosol as a putative redox signal. "

Is the collagen being broken down in ME patients to provide electrons for ATP synthesis?

If so, is AMPK involved in the process? And when/if AMPK "gives up", do ME cells go into senescence?


An external file that holds a picture, illustration, etc.
Object name is fig-2.jpg

Figure 2 from reference 9. PRODH/POX-mediated signaling. 
AMPK, AMP-activated protein kinase; ETC, electron transport chain; MYC, myelocytomatosis oncogene cellular homologue; PPARγ, peroxisome proliferator-activated receptor gamma. PRODH/POX, proline dehydrogenase/proline oxidase; ROS, reactive oxygen species.
https://www.liebertpub.com/doi/10.1089/ars.2017.7350


A concern for a dysregulated proline-P5C cycle is the potential for cells to accumulate P5C, which was recently listed among the top 30 damage-prone endogenous metabolites. (8, 10).



References: 

1) Sovio et al: 4-Hydroxyglutamate Is a Novel Predictor of Pre-Eclampsia.
PMID: 31098639 DOI: 10.1093/ije/dyz098

2) Pitt et al: 4-hydroxyglutamate Is a Biomarker for Primary Hyperoxaluria Type 3
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4270872/
PMID: 24563386 PMCID: PMC4270872 DOI: 10.1007/8904_2013_291

3) Arnaud Germain , Dinesh K Barupal , Susan M Levine , Maureen R Hanson:
Comprehensive Circulatory Metabolomics in ME/CFS Reveals Disrupted Metabolism of Acyl Lipids and Steroids. Metabolites 2020, 10(1), 4; https://doi.org/10.3390/metabo10010034
https://www.mdpi.com/2218-1989/10/1/34

4) Travis J Riedel , Lynnette C Johnson, John Knight, Roy R Hantgan, Ross P Holmes, W Todd Lowther: Structural and Biochemical Studies of Human 4-hydroxy-2-oxoglutarate Aldolase: Implications for Hydroxyproline Metabolism in Primary Hyperoxaluria.
Plos One https://doi.org/10.1371/journal.pone.0026021https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0026021

5) Naviaux RK, Naviaux JC, Li K, Bright AT, Alaynick WA, Wang L, Baxter A, Nathan N et al (2016) Metabolic features of chronic fatigue syndrome. Proc Natl Acad Sci U S A 113:E5472–E5480. https://doi.org/10.1073/pnas.1607571113

6) Greed, L., Willis, F., Johnstone, L. et al. Metabolite diagnosis of primary hyperoxaluria type 3. Pediatr Nephrol 33, 1443–1446 (2018). https://doi.org/10.1007/s00467-018-3967-6
https://link.springer.com/article/10.1007/s00467-018-3967-6

7) Schutzer et al: Distinct Cerebrospinal Fluid Proteomes Differentiate Post- Treatment Lyme Disease from Chronic Fatigue Syndrome. PLOS One February 2011, volume 6, Issuehttps://journals.plos.org/plosone/article?id=10.1371/journal.pone.0017287

8) John J Tanner, Sarah-Maria Fendt , Donald F Becker , John J Tanner, Sarah-Maria Fendt , Donald F Becker: The Proline Cycle As a Potential Cancer Therapy Target
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6026536/
https://pubs.acs.org/doi/10.1021/acs.biochem.8b00215

9) Phang: Proline Metabolism in Cell Regulation and Cancer Biology: Recent Advances and Hypotheses. Antioxid Redox Signal, 30 (4), 635-649 2019 Feb 1
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6338564/

10) Claudia Lerma-Ortiz et al: Nothing of Chemistry Disappears in Biology': The Top 30 Damage-Prone Endogenous Metabolites.
PMID: 27284066 DOI: 10.1042/BST20160073 Biochem Soc Trans (2016) 44 (3): 961–971.
https://doi.org/10.1042/BST20160073

fredag den 17. januar 2020

Naltrexone til behandling af ME

NK (Natural Killer) celler er en del af immunforsvaret. Det er gentagne gange påvist, at NK celler ikke fungerer optimalt hos ME patienter (1).

NK celler reguleres af en lang række celle signaler, - herunder calcium ion signalering. Ionkanalen TRPM3 bidrager væstentligt til calcium ion signalering i NK celler. 

Der er påvist signifikant reduktion af TRPM3 ekspression på overfladen af NK celler fra ME patienter (1).

Den G-protein-koblede receptor mu-opoid receptor (μOR) interagerer med TRPM3. Når μOR stimuleres inhiberes TRPM3 (1).

μOR kan hæmmes af lægemidlet Naltrexone, som er et lægemiddel målrettet til opioid-afhængige mennesker (2). Naltrexone i lav dosis anvendes også off-label til at modulere immunforsvaret i en række forskellige sygdomme.

Et forsøg med NK celler isoleret fra ME patienter viste at ved tilsætning af Naltrexone til NK cellerne, blev TRPM3 aktiviteten normaliseret (1).

Spørgsmålet er om Naltrexone kan være en behandlingsmulighed til ME? 


Læs også:
Low-dose naltrexone as a treatment for chronic fatigue syndrome
https://casereports.bmj.com/content/13/1/e232502.long


Finally Found – A Natural Killer Cell Enhancer for ME/CFS?
https://www.healthrising.org/blog/2019/12/19/50190/


Referencer:

1) Helene Cabanas, Katsuhiko Muraki, Donald Staines1 and Sonya Marshall-Gradisnik: Naltrexone Restores Impaired Transient Receptor Potential Melastatin 3 Ion Channel Function in Natural Killer Cells From Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Patients. Front. Immunol., 31 October 2019 | https://doi.org/10.3389/fimmu.2019.02545
https://www.frontiersin.org/articles/10.3389/fimmu.2019.02545/full



2) Naltrexone
http://pro.medicin.dk/Medicin/Praeparater/7045




torsdag den 2. januar 2020

Trådløs teknologi påvirker mitokondrierne, når cellerne er stressede

Forskere fra et universitet i Schweiz har undersøgt, hvordan nerveceller påvirkes af radiofrekvente elektromagnetiske felter (RF-EMF). Forskerne udsatte en cellekultur af nerveceller for 935 MHz, 4 W/kg i 24 timer. Forsøget viste ikke påvirkning af cellerne, når næring og metabolisme var under normale forhold. Men når glucosen blev fjernet og cellerne blev stressede, registrerede man forringet mitokondrie funktion ved den følgende maximale respiration (1):

"These findings indicate that RF-EMF might lead to an impairment of mitochondrial function that is only manifest at maximal respiration and additional stressors such as glucose deprivation."


I et andet studie af RF-EMF udsætter man immunceller for 1,8 GHz og 200 V/m i 20 timer. Forsøget viste øget mitokondriel respiration, som var relateret til mitokondrie complex V aktivitet (2). Complex V danner energi-molekylerne ATP og kaldes derfor også for ATP syntase.

En del patienter med Myalgisk Encephalomyelitis (ME) udvikler elektromagnetisk hypersensitivitet (EHS). Man kan således formode, at ME sygdomsmekanismen er relateret til en RF-EMF følsom celleproces.

En undersøgelse af en lymfoblast cellekultur fra ME patienter viste (3): 

  • det stress-registrerende protein TORC1 var opreguleret
  • mitokondrie complex V fungerede ineffektivt
  • den mitokondrielle respiratoriske kapacitet var opreguleret (sandsynligvis for at kompensere for complex V ineffektivitet)


Cellestress, øget mitokondriel respiration og complex V påvirkning er således fællesnævnere for RF-EMF påvirkning og for ME.



Læs også:

Complex V is down in ME - does it also explain Electromagnetic Hypersensitivity? https://followmeindenmark.blogspot.com/2019/07/complex-v-is-down-in-me-does-it-also.html



Are rises in Electro-Magnetic Field in the human environment, interacting with multiple environmental pollutions, the tripping point for increases in neurological deaths in the Western World? https://www.ncbi.nlm.nih.gov/pubmed/31088653


Referencer
1) Niederhäusern et al: Effects of radiofrequency electromagnetic field exposure on neuronal differentiation and mitochondrial function in SH-SY5Y cells Toxicol In Vitro. 2019 Dec;61:104609. doi: 10.1016/j.tiv.2019.104609. Epub 2019 Jul 24.
https://www.ncbi.nlm.nih.gov/pubmed/31351122


2) Lassaivia et al: Exposure to 1.8 GHz electromagnetic fields affects morphology, DNA-related Raman spectra and mitochondrial functions in human lympho-monocytes. PLOS ONE. February 20, 2018.https://doi.org/10.1371/journal.pone.0192894

3) Missailidis, D.; Annesley, S.; Allan, C.; Sanislav, O.; Lidbury, B.; Lewis, D.; Fisher, P. An Isolated Complex V Inefficiency and Dysregulated Mitochondrial Function in Immortalized Lymphocytes from ME/CFS Patients. Preprints 2019, 2019090043 (doi: 10.20944/preprints201909.0043.v1). https://www.preprints.org/manuscript/201909.0043/v1