- Review
- Open access
- Published:
mTORC1 syndrome (TorS): unifying paradigm for PASC, ME/CFS and PAIS
Journal of Translational Medicine volume 23, Article number: 297 (2025)
Abstract
Post-acute SarS-Cov2 (PASC), Myalgia encephalomyelitis/Chronic fatigue syndrome (ME/CFS) and Post-acute infection syndrome (PAIS) consist of chronic post–acute infectious syndromes, sharing exhaustive fatigue, post exertional malaise, intermittent pain, postural tachycardia and neuro-cognitive-psychiatric dysfunction. However, the concerned shared pathophysiology is still unresolved in terms of upstream drivers and transducers. Also, risk factors which may determine vulnerability/progression to the chronic phase still remain to be defined. In lack of drivers and a cohesive pathophysiology, the concerned syndromes still remain unmet therapeutic needs. ‘mTORC1 Syndrome’ (TorS) implies an exhaustive disease entity driven by sustained hyper-activation of the mammalian target of rapamycin C1 (mTORC1), and resulting in a variety of disease aspects of the Metabolic Syndrome (MetS), non-alcoholic fatty liver disease, chronic obstructive pulmonary disease, some cancers, neurodegeneration and other [Bar-Tana in Trends Endocrinol Metab 34:135–145, 2023]. TorS may offer a cohesive insight of PASC, ME/CFS and PAIS drivers, pathophysiology, vulnerability and treatment options.
Main text
Post-acute sequelae of SARS-CoV-2 infection (PASC), Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), and post-acute infection syndrome (PAIS) represent a spectrum of post-infectious conditions often triggered by viral infection, and characterized by persistent debilitating fatigue, unrefreshing sleep, post-exertional malaise and cognitive dysfunction that may last for months after the acute infection phase. These diseases were hypothesized to reflect long-lasting systemic inflammation due to persistent viral RNA and/or protein, on-going immune response, induction of autoimmunity, mitochondrial dysfunction and/or gut microbiome dysbiosis. However, the pathophysiology of the concerned diseases in terms of shared upstream driver(s) and transduction pathway(s) remain(s) unresolved. Also, predictive risk factors and/or background diseases which may determine vulnerability/progression of respective acute infections to the post-acute/chronic phase remain to be defined. Most importantly, in the absence of a driver and vulnerability profile, the concerned diseases present unmet therapeutic needs. This perspective may offer a cohesive insight of PASC, ME/CFS and PAIS pathophysiology, vulnerability and treatment options.
PASC, ME/CFS and PAIS: unresolved syndromes/unmet treatment needs
Post-acute SARS-Cov2 (PASC)/Long Covid consists of multi-system relapsing–remitting disease, including fatigue, post exertional malaise, intermittent pain, dyspnea, autonomic failure/postural tachycardia syndrome (PoTS) and neuro-psychiatric dysfunction (brain fog, anxiety, depression, peripheral neuropathy) [1]. PASC may follow an initial recovery from acute Covid-19, or the acute illness may persist for more than 12 weeks [2]. PASC inflicts more than 10% of those who had Covid-19 (over 65 million patients worldwide) [3, 4], independently of prior anti-SARS-Cov2 vaccination [5; however see 6] or anti-viral treatment [7]. Females, in particular premenopausal, are twice as affected by PASC compared to males [8], pointing to sex hormones in promoting PASC [9]. PASC is hypothesized to be driven by long-lasting systemic inflammation due to persistent SARS-Cov2 viral RNA and/or protein, on-going immune response, induction of autoimmunity, reactivation of other latent viruses (e.g., EBV, HHV6), and/or gut microbiome dysbiosis [1, 10, 11]. PASC pathophysiology is still unresolved, resulting in unmet therapeutic need [1, 10, 11].
Myalgic encephalomyelitis/Chronic fatigue syndrome (ME/CFS) consists of post-infectious (e.g., EBV, HHV6) multi-system disease that persists for more than 6-months, presenting profound fatigue, post-exertional malaise, unrefreshing sleep, PoTS, intermittent muscle/joint pain, and neuro-cognitive dysfunction (brain fog, depression, peripheral neuropathy) [12, 13]. ME/CFS inflicts nearly 0.9% of worldwide population [14]. Similarly to PASC, ME/CFS affects premenopausal females (3:1 preponderance compared to males) [15]. ME/CFS is hypothesized to be due to persistent pathogen-associated molecular patterns (PAMPs) (e.g., viral, bacterial, parasitic), long-lasting systemic inflammation, mitochondrial dysfunction, on-going immune response, induction of autoimmunity, and/or gut microbiome/virome/mycobiome dysbiosis [16, 17]. ME/CFS pathophysiology and biomarker-based tests are still unresolved, resulting in unmet therapeutic need [17,18,19].
PASC major symptoms, including fatigue, post exertional malaise, myalgia, PoTS and neuro-cognitive-psychiatric dysfunction, overlap with ME/CFS [16]. Also, the pathophysiology proposed for PASC overlaps with that proposed for ME/CFS, including PAMPS-induced long-lasting systemic inflammation, immune activation, autoimmune response against self-antigens, mitochondrial dysfunction, reactivation of latent viruses and gut dysbiosis. The PASC/ME/CFS overlap has been realized by many [16, 20, 21], resulting initially in viewing PASC as a particular case of ME/CFS [22]. Alternatively, PASC and ME/CFS were proposed to present two particular reflections of a Post active phase of infection syndromes (PAPIS) sharing the same pathophysiology [23]. Since the concerned symptoms and pathophysiology are shared as well by a variety of viral (e.g., SARS-CoV2, EBV, SARS, MERS, other) and some non-viral pathogens, the PAPIS view has been further advanced by proposing recently the disease category of Post-acute infection syndromes (PAIS) [24], implying an exhaustive unifying syndrome, including PASC and ME/CFS as particular examples.
The proposed PAIS paradigm still remains ‘Unexplained’ [24] with respect to the following:
-
a.
Driver: The PAIS paradigm lacks an upstream driver which may transduce the complexed pathophysiology and multi-symptoms of PAIS. Specifically, while being central to the PAIS paradigm, mitochondrial stress and its systemic fatigue and neuro-cognitive-psychiatric outcomes, remain to be defined in terms of an upstream mechanistic driver [25, 26]. Similarly, the preponderance of premenopausal females’ progression to PAIS remains to be defined in terms of respective driver(s).
-
b.
Risks: The PAIS paradigm fails to define predictive risk factors/background diseases which may determine vulnerability/progression of respective acute infections to the post-acute PAIS phase.
-
c.
Diagnosis/treatment: In lack of a cohesive pathophysiology, PAIS biomarker(s) remain(s) undefined, resulting in critical deficiency in diagnosing PAIS. Most importantly, in the absence of a driver, PAIS remains an unmet therapeutic need. The mTORC1 Syndrome (TorS) paradigm may offer a cohesive framework for the ‘Unexplained’ aspects of PAIS.
mTORC1 syndrome (TorS)
The mammalian target of rapamycin complex 1 (mTORC1) controls growth and metabolism by affecting a variety of its downstream targets (e.g., S6K1, 4EBP, CRTC2, lipin, ATF4, HIF1a, PPARg, PPARa, ULK1, TFEB and others) [27]. mTORC1 kinase controls G1/S transition and G2/M progression, activates ribosome biogenesis and CAP-dependent mRNA translation, drives purine and pyrimidine biosynthesis, promotes glycolysis and the pentose shunt, and suppresses fatty acid oxidation and ketogenesis [27]. Most importantly, mTORC1 blocks autophagy and mitophagy [28]. mTORC1 kinase activity may range between hyper- and less- active state, as function of genetic, epigenetic, tissue and/or context-dependent factors. These may determine mTORC1 sensitivity to growth factors, nutrients and stress. mTORC1 kinase activity may be hyper-activated by growth factors (e.g., insulin, IGF1), energy/nutrients excess (e.g., glucose, leucine, arginine) and inflammation (e.g., NFkB/IKK), while being suppressed by a variety of metabolic stresses (e.g., hypoxic, hyperosmotic, oxidative, acidic, DNA damage) [27, 29], including caloric restriction [30, 31], carbohydrate restriction [32,33,34], bariatric surgery [35], or sustained physical exercise [36, 37]. When applied, these suppression measures are effective in increasing health span and predicting an increase in life span [38].
‘mTORC1 Syndrome’ (TorS) implies an exhaustive cohesive disease entity driven by sustained hyper-activation of mTORC1 [39]. Hyperactive mTORC1 may drive the glycemic context/beta-cell failure of type 2 diabetes (T2D) as well as a variety of non-glycemic disease aspects of the Metabolic Syndrome (MetS), including obesity, dyslipidemia, hypertension, atherosclerotic cardiovascular disease (ASCVD), MetS/T2D cardiomyopathy, nephropathy and peripheral neuropathy, non-alcoholic fatty liver disease/non-alcoholic steatohepatitis (NAFLD/NASH), chronic obstructive pulmonary disease (COPD), some cancers, neurodegeneration, polycystic ovary syndrome (PCOS), psoriasis and other [39]. The TorS paradigm may offer a unifying view for T2D/MetS pathophysiology and treatment, while solving the apparent dis-concordance between response and resistance to insulin in shaping the exhaustive phenotype of T2D/MetS [39, 40]. Also, mutual interactions between TorS and viral effectors/non-viral pathogens are proposed here to predispose/drive PASC, ME/CFS or PAIS (Fig. 1).
Of note, TorS has been previously proposed to drive the acute COVID-19 disease through interplay of the host innate stress response, plethora of viral measures aimed at subverting the innate stress response, mTORC1-enabled viral proliferation, mTORC1-suppressed autophagy/mitophagy and the impact of the TorS-driven host diseases [41, 42]. Hence, TorS is proposed to predispose/drive both, the acute viral disease as well as the chronic PASC phase of SARS-Cov2 infection.
TorS/PASC
PASC is reported to be predisposed by background diseases, including obesity, T2D, dyslipidemia, hypertension, ischemic cardiovascular disease, cardiomyopathy, chronic kidney disease, restrictive lung disease, thromboembolic disorders, NAFLD, cerebrovascular disease and polyneuropathy [43,44,45,46,47]. These background diseases may all be considered as TorS diseases driven by hyperactive mTORC1 [39, 40]. In line with that, the predisposition of PASC in premenopausal females may be accounted for by mTORC1 activation through estrogen signaling [48, 49]. Of note, in addition to PASC being driven by background TorS diseases, SARS-CoV2 infection may drive new-onset TorS diseases, including T2D, insulin resistance, cardiovascular disease, hypertension, chronic kidney disease, dyslipidemia, autoimmune diseases, systemic inflammation, and neurological and cognitive impairments [50, 51; however see 52]. Hence, hyperactive mTORC1/TorS may act both, as upstream driver of PASC (Fig. 1) as well as downstream target of SARS-CoV2 infection.
In addition to PASC being predisposed by background TorS diseases, hyperactive mTORC1 may further drive the multi disease aspects of PASC. Thus, the most experienced symptoms of PASC include chronic fatigue, post-exertion malaise and brain fog, being presented by more than half of Covid-19 survivors [1, 53], and ascribed to ‘mitochondrial dysfunction’ [54, 55]. Indeed, PASC results in disrupted mitochondrial integrity and mtDNA, decrease in mitochondria membrane potential, oxygen uptake, oxidative phosphorylation and ATP production, with increased mitochondrial ROS production and disrupted ER-mito Ca++ dynamics in heart, liver, kidney and brain [1, 56, 57]. Mitochondrial stress has been primarily ascribed to direct binding and/or transcriptional disruption of mitochondrial components by viral proteins [56, 57]. Alternatively, in line with the TorS paradigm, PASC mitochondrial dysfunction/stress is proposed here to be due to inhibition of mitophagy by hyperactive mTORC1 [28], resulting in disrupted mitochondrial integrity and cell death [58, 59]. Hence, PASC may be predisposed by TorS diseases, while being further driven by disruption of mitophagy due to hyperactive mTORC1. The TorS paradigm may also account for the concomitant increase in glycolysis in PASC patients [60, 61], being driven by mTORC1-driven HIF1alpha [62].
PASC pathophysiology may further be ascribed to mTORC1/STAT3 interplay. Indeed, mTORC1 kinase activates the transcriptional activity of STAT3 [63], while STAT3 may activate mTORC1 kinase activity by suppressing REDD1 [64, 65]. Indeed, STAT3 is reported to drive both, the acute Covid-19 [66, 67] and PASC [68], implying that mTORC1 and STAT3 may synergize each other in promoting the acute and PASC phases of SARS-CoV2 infection (Fig. 2). Indeed, the mTORC1/STAT3 synergism may account for the immune profile of PASC (M1-macrophages activation with decrease in CD4 + and CD8 + effector memory cells [1]) [69,70,71], increase in IL6, CCL11, fibrinogen and D-dimer [1, 72,73,74,75], complement dysregulation [76, 77], NLRP3 inflammasome activation [78, 79], HPA axis dysfunction/decrease in plasma cortisol and serotonin [80, 81], increase in autoimmune antibodies (e.g., anti-ACE2, anti-GPCR) [1, 82], heart failure [83, 84], deep vein thrombosis/pulmonary emboli [85, 86], angiogenesis markers [87, 88], restrictive lung failure/pulmonary fibrosis [89,90,91], autonomic dysfunction/PoTS [92, 93], endothelial dysfunction [94, 95], erectile dysfunction [96, 97], neurocognitive decline, fatigue/post-exertional malaise [1, 28, 53], systemic inflammation, neuro-inflammation/glia activation [98,99,100,101,102] and small nerve fiber loss [103,104,105]. Hence, mTORC1 and IL6/STAT3 may synergize in driving PASC (Fig. 2).
mTORC1/STAT3 synergism. Hyperactive mTORC1 and STAT3 may each drive respective patho aspects of PASC, ME/CFS or PAIS. Moreover, hyperactive mTORC1 and STAT3 are proposed to synergize each other: mTORC1 kinase activates the transcriptional activity of STAT3, while STAT3 may activate mTORC1 kinase activity by suppressing REDD1
TorS/ME/CFS
Similarly to PASC, ME/CFS is reported to be predisposed/driven by background MetS diseases, including obesity, hyperglycemia/T2D, dyslipidemia and hypertension [106,107,108,109]. In line with that, the ME/CFS fatigue score is reported to be linearly correlated with the number of MetS diseases, implying a putative causal linkage [106,107,108,109]. Of note, the concerned MetS diseases may all be considered as diseases driven by hyperactive mTORC1, namely, disease aspects of TorS [39]. Hence, background TorS is proposed to predispose/drive post-infectious ME/CFS (Fig. 1). In line with that, mTORC1 activation through estrogen signaling [48, 49] may account for the increased ME/CFS morbidity of premenopausal females. Moreover, TorS has been previously proposed to predispose/drive the acute disease phase of viral infections, due to mTORC1-enabled viral proliferation [41, 42, 110]. Hence, TorS is proposed to predispose/drive both, the acute viral proliferation phase that precedes ME/CFS, as well as the chronic ME/CFS disease (Fig. 1).
The most experienced symptoms of ME/CFS include extreme chronic fatigue, post-exertion malaise and brain fog, being ascribed to mitochondrial energy stress [111, 112; however see 113]. Indeed, ME/CFS results in disrupted mitochondrial integrity, basal respiration, aerobic respiratory capacity, mito membrane potential, coupling efficiency and ATP production, combined with increased mito ROS production in muscle, brain, immune cells, PBMC and other [25, 26, 114,115,116,117]. However, in contrast to classical mitochondrial diseases which present pronounced fatigue [118], no mtDNA mutations or differences in individual mito complexes activity have been reported in ME/CFS patients [119], pointing to an upstream driver in causing mitochondrial stress [115, 120]. Also, ME/CFS mitochondrial stress is accompanied by increased glycolysis [121,122,123]; however see 124], with concomitant decrease in PDH/TCA cycle activity [125], implying an upstream driver(s) which control(s) non-oxidative metabolism. In line with the TorS/ME/CFS paradigm, ME/CFS mitochondrial stress is proposed to be driven by inhibition of mitophagy/autophagy due to hyperactive mTORC1 [25, 28, 59, 126], resulting in disrupted mitochondrial integrity and cell death. Hyperactive mTORC1 may also account for HIF1alpha activation, resulting in driving glycolysis [62], while suppressing PDH/TCA activity [127]. The TorS/mitochondrial stress paradigm may further account for the immune exhaustion of CD8 + T cells in ME/CFS [25, 128,129,130,131,132,133], meta-inflammation [117, 134,135,136], cardiovascular, endothelial dysfunction, platelet hyper-activation and coagulopathy [21] profiles of ME/CFS. Meta-inflammation and immune exhaustion of ME/CFS may further be driven by mTORC1-activated IL6/STAT3 [132, 133] (Fig. 2). Hence, mTORC1 and STAT3 may synergize in driving ME/CFS.
TorS: unified paradigm for PASC, ME/CFS, PAIS
Viewing PAIS (including PASC and ME/CFS as particular examples) in the context of TorS may answer some of the ‘Unexplained’ [24] aspects of PAIS:
-
a.
Driver: PAIS is proposed to be driven by hyperactive mTORC1/TorS. Hyperactive mTORC1 enables virus proliferation during the acute infectious disease preceding PAIS [41, 42], and further persists throughout the post-acute PAIS phase, being sustained by background TorS diseases. The two concerned effectors of hyperactive mTORC1, namely the acute viral infection and background TorS are proposed to be both required for driving PAIS, while synergizing each other (Fig. 1). Of note, viewing PAIS in the context of hyperactive mTORC1/TorS may account for PAIS mito dysfunction due to suppression of mitophagy [28].
-
b.
Risks: Viewing PAIS in the context of TorS implies that TorS diseases may serve as risk factors in transforming respective acute viral diseases into persistent PAIS, being driven by the mTORC1/STAT3 synergism [63,64,65] (Fig. 2). Also, mTORC1 activation by estrogens may account for the female preponderance in progression to PAIS. Indeed, estrogens may activate mTORC1 through multiple pathways, including the PKC-ERK1/2 pathway, the PI3K/AKT pathway, and/or direct interactions with ERα thereby initiating a signaling cascade involving c-Src and/or PI3K/AKT (48, 49). Activated ERK1, or AKT phosphorylate and inhibit TSC1,2, a negative regulator of mTORC1, thereby promoting mTORC1 activation.
-
c.
Treatment: Prior to the vaccination era, Covid-19 treatments have aimed at inhibiting viral proliferation and/or acute inflammation, using antiviral drugs [137], STAT3 inhibitors [138] and/or glucocorticoids [139]. These treatments became mostly redundant due to the efficacy of Covid-19 vaccinations to prevent and antagonize the acute Covid-19 infection and disease. However, PASC still remains an unmet need, in spite of Covid-19 vaccinations or anti-viral measures [140, 141]. Similarly, ME/CFS still remains a treatment deadlock, resulting in human suffering and major health expenditures [142].
The TorS paradigm, proposed here as driver of PASC/ME/CFS/PAIS pathophysiology, implies that suppression of hyperactive mTORC1, and in particular rescue of mTORC1-suppressed mitophagy, may offer a disease-modifying treatment for PASC, ME/CFS or PAIS (Fig. 3). Indeed, anecdotal reports have indicated prospective efficacy of rapalogs in the treatment of PASC [143] or ME/CFS [144, 145]. In line with that, induction of mitophagy is reported to mitigate meta-inflammation [146,147,148], immune activation [149], and neurological/cognitive dysfunction [150], namely the core presentations of PASC/ME/CFS/PAIS. However, sustained inhibition of mTORC1 activity by rapalogs may result in inhibition of mTORC2 as well, resulting in diabetes [151]. Also, treatment with rapalogs may result in severe side-effects, while trials to bypass side-effects by intermittent treatments still remain to be accomplished. Moreover, suppression of mTORC1 kinase activity by rapalogs results in suppressing some (e.g., S6K1), but not all downstream targets of mTORC1 (e.g., 4EBP) [152], implying a potential resistance to rapalogs in the treatment of TorS-driven PASC, ME/CFS or PAIS.
Alternatively, hyperactive mTORC1 may be indirectly suppressed by a variety of metabolic effectors, including caloric restriction, carbohydrate restriction/ketogenic diets, hyperbaric oxygen or sustained physical exercise [39] (Fig. 3). When applied, these measures are effective in alleviating some of the pleiotropic diseases of TorS [40], including disease aspects of PASC/ME/CFS [44, 153,154,155]. However, the compliance to the concerned dietary and exercise behavioral measures is poor, in particular in PASC/ME/CFS/PAIS patients suffering from gastrointestinal dysbiosis and/or post-exertion malaise. Hence, treatment of TorS-driven PASC, ME/CFS and PAIS calls for alternative measures that may suppress hyperactive mTORC1 while inducing mitophagy.
Indeed, suppression of hyperactive mTORC1, with concomitant rescue of mitophagy, may be effected by mitochondrial energy stress, being transduced to mTORC1 through the integrated stress response (ISR), activated HRI, ATF4 and REDD1 [156, 157]. Mitochondrial energy stress resulting in rescuing mitophagy may be effected by mild uncoupling of mitochondrial oxidative phosphorylation (e.g., DNP, TPP-cations, long-chain fatty acids) [158,159,160,161,162], or mild inhibition of mito complex I (e.g., metformin, phenformin) [163,164,165], or opening of the mito permeability transition pore (PTP) [166], or mild hypoxia [167, 168] (Fig. 3). Of note, long-chain fatty acyl analogs of the MEDICA family are reported to inhibit mito complex I, to open mito PTP, to suppress mTORC1 kinase activity and to suppress STAT3 transcriptional activity [169,170,171], implying prospective double-headed efficacy in the treatment of PAIS (Fig. 4).
Of note, suppression of mitochondrial oxidative phosphorylation by mild uncouplers, complex I inhibitors, PTP openers, hypoxia or MEDICA may apparently sound counterproductive when aiming to rescue ‘mitochondrial dysfunction’ inflicted by hyperactive mTORC1 and suppressed mitophagy. This apparent ambiguity implies that suppression of mitophagy in the PASC, ME/CFS or PAIS context is far more destructive than mitochondrial energy stress due to limited oxidative phosphorylation. Moreover, the concerned ambiguity implies that ‘dysfunction’ due to suppression of mitophagy may be rescued by limited mitochondrial energy stress.
Concluding remarks
PASC, ME/CFS and PAIS consist of chronic post–infectious multi-system syndromes, sharing exhaustive fatigue, post exertional malaise, intermittent pain and neuro-cognitive-psychiatric dysfunction. In lack of a cohesive pathophysiology these syndromes still remain unmet therapeutic needs. TorS may offer an insight of PASC, ME/CFS and PAIS pathophysiology and patient vulnerability. Suppression of hyperactive mTORC1/STAT3 by mitochondrial energy stress may result in disease-modifying treatment for the concerned syndromes. ‘That Which Does Not Kill Us Makes Us Stronger’.
The TorS/PASC/ME/CFS/PAIS paradigm leaves open the following questions:
(a) The proposed paradigm still has to account for the extreme variability of clinical presentations of concerned patients. The phenotypic variability could reflect the featured activity of respective infective agents and/or the multi genetic, epigenetic, and tissue/context-dependent factors that may modulate the mTORC1/STAT3 activity of infected patients. (b) The proposed paradigm still has to define accessible biomarker(s) (e.g., phospho-S6K1, phospho-4EBP1, phospho-STAT3(Y705) [172, 173]) that may point to mTORC1/STAT3 activity over time in candidate patients. (c) The reported anecdotal role of rapamycin in alleviating PAIS [143,144,145] remains to be confirmed in controlled clinical study. d. Mitochondrial energy stress, affected by mild uncoupling of mitochondrial oxidative phosphorylation, or mild inhibition of mitochondrial complex I, or opening of the mitochondrial permeability transition pore, or mild hypoxia may offer a disease-modifying treatment for PASC/ME/CFS/PAIS patients. However, the balance between therapeutic stress and pathological damage might be challenging. e. It still remains to be investigated whether suppression of mTORC1/STAT3 during the preceding acute infectious disease may prevent the chronic disease phase. Animal models may provide early disease correlates and molecular signatures associated with advanced disease development over time, including countermeasure performance before and during the acute infection [174, 175]. f. In view of the similar clinical profile shared by PASC/ME/CFS/PAIS and Fibromyalgia [176,177,178], Fibromyalgia may fit into the proposed TorS paradigm.
Availability of data and materials
All data generated or analyzed during this study are included in this published article.
Abbreviations
- ASCVD:
-
Atherosclerotic cardiovascular disease
- COPD:
-
Chronic obstructive pulmonary disease
- ME/CFS:
-
Myalgia encephalomyelitis/Chronic fatigue syndrome
- MetS:
-
Metabolic syndrome
- mTORC1:
-
Mammalian target of rapamycin C1
- NAFLD:
-
Non-alcoholic fatty liver disease
- NASH:
-
Non-alcoholic steatohepatitis
- PAIS:
-
Post-acute infection syndrome
- PAMPs:
-
Pathogen-associated molecular patterns
- PAPIS:
-
Post active phase of infection syndromes
- PASC:
-
Post-acute SarS-Cov2
- PBMC:
-
Peripheral blood mononuclear cells
- PCOS:
-
Polycystic ovary syndrome
- PDH:
-
Pyruvate dehydrogenase
- PoTS:
-
Postural tachycardia syndrome
- PTP:
-
Permeability transition pore
- SARS-CoV2:
-
Severe acute respiratory syndrome Coronavirus 2
- STAT 3:
-
Signal transducer and activator of transcription 3
- T2D:
-
Type 2 diabetes
- TCA:
-
Tricarboxylic acid cycle
- TorS:
-
MTORC1 syndrome
References
Davis HE, McCorkell L, Vogel JM, Topol EJ. Long COVID: major findings, mechanisms and recommendations. Nat Rev Microbiol. 2023;21(3):133–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41579-022-00846-2.
Soriano JB, Murthy S, Marshall JC, Relan P, Diaz JV. WHO Clinical Case Definition Working Group on Post-COVID-19 Condition. A clinical case definition of post-COVID-19 condition by a Delphi consensus. Lancet Infect Dis. 2022;22(4):e102–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S1473-3099(21)00703-9.
Hastie CE, Lowe DJ, McAuley A, Mills NL, Winter AJ, Black C, Scott JT, O’Donnell CA, Blane DN, Browne S, Ibbotson TR, Pell JP. True prevalence of long-COVID in a nationwide, population cohort study. Nat Commun. 2023;14(1):7892. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-023-43661-w.
Long COVID: 3 years in. Lancet. 2023;401(10379):795. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0140-6736(23)00493-2.
Byambasuren O, Stehlik P, Clark J, Alcorn K, Glasziou P. Effect of covid-19 vaccination on long covid: systematic review. BMJ Med. 2023;2(1): e000385. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/bmjmed-2022-000385.
Lundberg-Morris L, Leach S, Xu Y, Martikainen J, Santosa A, Gisslén M, Li H, Nyberg F, Bygdell M. Covid-19 vaccine effectiveness against post-covid-19 condition among 589 722 individuals in Sweden: population based cohort study. BMJ. 2023;383: e076990. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/bmj-2023-076990.
Nevalainen OPO, Horstia S, Laakkonen S, Rutanen J, Mustonen JMJ, Kalliala IEJ, Ansakorpi H, Kreivi HR, Kuutti P, Paajanen J, Parkkila S, Paukkeri EL, Perola M, Pourjamal N, Renner A, Rosberg T, Rutanen T, Savolainen J, Solidarity Finland Investigators, Haukka JK, Guyatt GH, Tikkinen KAO. Effect of remdesivir post hospitalization for COVID-19 infection from the randomized SOLIDARITY Finland trial. Nat Commun. 2022;13(1):6152. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-022-33825-5.
Sigfrid L, Drake TM, Pauley E, Jesudason EC, Olliaro P, Lim WS, Gillesen A, Berry C, Lowe DJ, McPeake J, Lone N, Munblit D, Cevik M, Casey A, Bannister P, Russell CD, Goodwin L, Ho A, Turtle L, O’Hara ME, Hastie C, Donohue C, Spencer RG, Donegan C, Gummery A, Harrison J, Hardwick HE, Hastie CE, Carson G, Merson L, Baillie JK, Openshaw P, Harrison EM, Docherty AB, Semple MG, Scott JT, ISARIC4C investigators. Long Covid in adults discharged from UK hospitals after Covid-19: A prospective, multicentre cohort study using the ISARIC WHO Clinical Characterisation Protocol. Lancet Reg Health Eur. 2021;8: 100186. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lanepe.2021.100186.
Stewart S, Newson L, Briggs TA, Grammatopoulos D, Young L, Gill P. Long COVID risk—a signal to address sex hormones and women’s health. Lancet Reg Health Eur. 2021;11: 100242. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lanepe.2021.100242.
Bonilla H, Peluso MJ, Rodgers K, Aberg JA, Patterson TF, Tamburro R, Baizer L, Goldman JD, Rouphael N, Deitchman A, Fine J, Fontelo P, Kim AY, Shaw G, Stratford J, Ceger P, Costantine MM, Fisher L, O’Brien L, Maughan C, Quigley JG, Gabbay V, Mohandas S, Williams D, McComsey GA. Therapeutic trials for long COVID-19: A call to action from the interventions taskforce of the RECOVER initiative. Front Immunol. 2023;14:1129459. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1129459.
Sherif ZA, Gomez CR, Connors TJ, Henrich TJ, Reeves WB, RECOVER Mechanistic Pathway Task Force. Pathogenic mechanisms of post-acute sequelae of SARS-CoV-2 infection (PASC). Elife. 2023;12: e86002. https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.86002.
Deumer US, Varesi A, Floris V, Savioli G, Mantovani E, López-Carrasco P, Rosati GM, Prasad S, Ricevuti G. Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): an overview. J Clin Med. 2021;10(20):4786. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/jcm10204786.
Bateman L, Bested AC, Bonilla HF, Chheda BV, Chu L, Curtin JM, Dempsey TT, Dimmock ME, Dowell TG, Felsenstein D, Kaufman DL, Klimas NG, Komaroff AL, Lapp CW, Levine SM, Montoya JG, Natelson BH, Peterson DL, Podell RN, Rey IR, Ruhoy IS, Vera-Nunez MA, Yellman BP. Myalgic encephalomyelitis/chronic fatigue syndrome: essentials of diagnosis and management. Mayo Clin Proc. 2021;96(11):2861–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.mayocp.2021.07.004.
Lim EJ, Ahn YC, Jang ES, Lee SW, Lee SH, Son CG. Systematic review and meta-analysis of the prevalence of chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME). J Transl Med. 2020;18(1):100. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-020-02269-0.
Thomas N, Gurvich C, Huang K, Gooley PR, Armstrong CW. The underlying sex differences in neuroendocrine adaptations relevant to myalgic encephalomyelitis chronic fatigue syndrome. Front Neuroendocrinol. 2022;66: 100995. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.yfrne.2022.100995.
Komaroff AL, Lipkin WI. Insights from myalgic encephalomyelitis/chronic fatigue syndrome may help unravel the pathogenesis of postacute COVID-19 syndrome. Trends Mol Med. 2021;27(9):895–906. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molmed.2021.06.002.
Cortes Rivera M, Mastronardi C, Silva-Aldana CT, Arcos-Burgos M, Lidbury BA. Myalgic encephalomyelitis/chronic fatigue syndrome: a comprehensive review. Diagnostics (Basel). 2019;9(3):91. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/diagnostics9030091.
Missailidis D, Annesley SJ, Fisher PR. Pathological mechanisms underlying myalgic encephalomyelitis/chronic fatigue syndrome. Diagnostics (Basel). 2019;9(3):80. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/diagnostics9030080.
Thoma M, Froehlich L, Hattesohl DBR, Quante S, Jason LA, Scheibenbogen C. Why the psychosomatic view on myalgic encephalomyelitis/chronic fatigue syndrome is inconsistent with current evidence and harmful to patients. Medicina (Kaunas). 2023;60(1):83. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/medicina60010083.
Sukocheva OA, Maksoud R, Beeraka NM, Madhunapantula SV, Sinelnikov M, Nikolenko VN, Neganova ME, Klochkov SG, Amjad Kamal M, Staines DR, Marshall-Gradisnik S. Analysis of post COVID-19 condition and its overlap with myalgic encephalomyelitis/chronic fatigue syndrome. J Adv Res. 2022;40:179–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jare.2021.11.013.
Komaroff AL, Lipkin WI. ME/CFS and Long COVID share similar symptoms and biological abnormalities: road map to the literature. Front Med (Lausanne). 2023;10:1187163. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmed.2023.1187163.
Poenaru S, Abdallah SJ, Corrales-Medina V, Cowan J. COVID-19 and post-infectious myalgic encephalomyelitis/chronic fatigue syndrome: a narrative review. Ther Adv Infect Dis. 2021;8:20499361211009384. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/20499361211009385.
Friedman KJ, Murovska M, Pheby DFH, Zalewski P. Our evolving understanding of ME/CFS. Medicina (Kaunas). 2021;57(3):200. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/medicina57030200.
Choutka J, Jansari V, Hornig M, Iwasaki A. Unexplained post-acute infection syndromes. Nat Med. 2022;28(5):911–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41591-022-01810-6.
Missailidis D, Annesley SJ, Allan CY, Sanislav O, Lidbury BA, Lewis DP, Fisher PR. An isolated complex V inefficiency and dysregulated mitochondrial function in immortalized lymphocytes from ME/CFS patients. Int J Mol Sci. 2020;21(3):1074. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms21031074.
Wood E, Hall KH, Tate W. Role of mitochondria, oxidative stress and the response to antioxidants in myalgic encephalomyelitis/chronic fatigue syndrome: a possible approach to SARS-CoV-2 “long-haulers”? Chronic Dis Transl Med. 2021;7(1):14–26. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cdtm.2020.11.002.
Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell. 2017;168(6):960–76. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2017.02.004.
Bartolomé A, García-Aguilar A, Asahara SI, Kido Y, Guillén C, Pajvani UB, Benito M. MTORC1 regulates both general autophagy and mitophagy induction after oxidative phosphorylation uncoupling. Mol Cell Biol. 2017;37(23):e00441-e517. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/MCB.00441-17.
Demetriades C, Plescher M, Teleman AA. Lysosomal recruitment of TSC2 is a universal response to cellular stress. Nat Commun. 2016;12(7):10662. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ncomms10662.
Madeo F, Carmona-Gutierrez D, Hofer SJ, Kroemer G. Caloric restriction mimetics against age-associated disease: targets, mechanisms, and therapeutic potential. Cell Metab. 2019;29:592–610. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2019.01.018.
Green CL, Lamming DW, Fontana L. Molecular mechanisms of dietary restriction promoting health and longevity. Nat Rev Mol Cell Biol. 2022;23:56–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41580-021-00411-4.
Lin SC, Hardie DG. AMPK: sensing glucose as well as cellular energy status. Cell Metab. 2018;27:299–313. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2017.10.009.
Leprivier G, Rotblat B. How does mTOR sense glucose starvation? AMPK is the usual suspect. Cell Death Discov. 2020;6:27. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41420-020-0260-9.
Westman EC, Tondt J, Maguire E, Yancy WS Jr. Implementing a low-carbohydrate, ketogenic diet to manage type 2 diabetes mellitus. Expert Rev Endocrinol Metab. 2018;13:263–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/17446651.2018.1523713.
Ma N, Ma R, Tang K, Li X, He B. Roux-en-Y gastric bypass in obese diabetic rats promotes autophagy to improve lipid metabolism through mTOR/p70S6K signaling pathway. J Diabetes Res. 2020;2020:4326549. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2020/4326549.
Gordon BS, Steiner JL, Rossetti ML, Qiao S, Ellisen LW, Govindarajan SS, Eroshkin AM, Williamson DL, Coen PM. REDD1 induction regulates the skeletal muscle gene expression signature following acute aerobic exercise. Am J Physiol Endocrinol Metab. 2017;313:E737–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/ajpendo.00120.2017.
Kido K, Sase K, Yokokawa T, Fujita S. Enhanced skeletal muscle insulin sensitivity after acute resistance-type exercise is upregulated by rapamycin-sensitive mTOR complex 1 inhibition. Sci Rep. 2020;10:8509. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-020-65397-z.
Papadopoli D, Boulay K, Kazak L, Pollak M, Mallette F, Topisirovic I, Hulea L. mTOR as a central regulator of lifespan and aging. F1000 Res. 2019;8:998. https://doiorg.publicaciones.saludcastillayleon.es/10.12688/f1000research.17196.1.
Bar-Tana J. mTORC1 syndrome (TorS): unified paradigm for diabetes/metabolic syndrome. Trends Endocrinol Metab. 2023;34(3):135–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tem.2023.01.001.
Bar-Tana J. TorS—reframing a rational for type 2 diabetes treatment. Diabetes Metab Res Rev. 2024;40(1): e3712. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/dmrr.3712.
Bar-Tana J. COVID-19—mTORC1-centric Paradigm. January 2020. SSRN Electron J. https://doiorg.publicaciones.saludcastillayleon.es/10.2139/ssrn.3574559.
Zambalde ÉP, Dias TL, Maktura GC, Amorim MR, Brenha B, Santos LN, Buscaratti L, Elston JGA, Mancini MCS, Pavan ICB, Toledo-Teixeira DA, Bispo-Dos-Santos K, Parise PL, Morelli AP, Silva LGSD, Castro ÍMS, Saccon TD, Mori MA, Granja F, Nakaya HI, Proenca-Modena JL, Marques-Souza H, Simabuco FM. Increased mTOR signaling and impaired autophagic flux are hallmarks of SARS-CoV-2 infection. Curr Issues Mol Biol. 2022;45(1):327–36. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cimb45010023.
Su Y, Yuan D, Chen DG, Ng RH, Wang K, Choi J, Li S, Hong S, Zhang R, Xie J, Kornilov SA, Scherler K, Pavlovitch-Bedzyk AJ, Dong S, Lausted C, Lee I, Fallen S, Dai CL, Baloni P, Smith B, Duvvuri VR, Anderson KG, Li J, Yang F, Duncombe CJ, McCulloch DJ, Rostomily C, Troisch P, Zhou J, Mackay S, DeGottardi Q, May DH, Taniguchi R, Gittelman RM, Klinger M, Snyder TM, Roper R, Wojciechowska G, Murray K, Edmark R, Evans S, Jones L, Zhou Y, Rowen L, Liu R, Chour W, Algren HA, Berrington WR, Wallick JA, Cochran RA, Micikas ME, ISB-Swedish COVID-19 Biobanking Unit, Wrin T, Petropoulos CJ, Cole HR, Fischer TD, Wei W, Hoon DSB, Price ND, Subramanian N, Hill JA, Hadlock J, Magis AT, Ribas A, Lanier LL, Boyd SD, Bluestone JA, Chu H, Hood L, Gottardo R, Greenberg PD, Davis MM, Goldman JD, Heath JR. Multiple early factors anticipate post-acute COVID-19 sequelae. Cell. 2022;185(5):881-895e20. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2022.01.014.
O’Hare AM, Vig EK, Iwashyna TJ, Fox A, Taylor JS, Viglianti EM, Butler CR, Vranas KC, Helfand M, Tuepker A, Nugent SM, Winchell KA, Laundry RJ, Bowling CB, Hynes DM, Maciejewski ML, Bohnert ASB, Locke ER, Boyko EJ, Ioannou GN, VA COVID Observational Research Collaboratory (CORC). Complexity and challenges of the clinical diagnosis and management of long COVID. JAMA Netw Open. 2022;5(11): e2240332. https://doiorg.publicaciones.saludcastillayleon.es/10.1001/jamanetworkopen.2022.40332.
Frere JJ, tenOever BR. Cardiometabolic syndrome—an emergent feature of Long COVID? Nat Rev Immunol. 2022;22(7):399–400. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41577-022-00739-8.
Stefan N, Sippel K, Heni M, Fritsche A, Wagner R, Jakob CEM, Preißl H, von Werder A, Khodamoradi Y, Borgmann S, Rüthrich MM, Hanses F, Haselberger M, Piepel C, Hower M, Vom Dahl J, Wille K, Römmele C, Vehreschild J, Stecher M, Solimena M, Roden M, Schürmann A, Gallwitz B, Hrabe de Angelis M, Ludwig DS, Schulze MB, Jensen BEO, Birkenfeld AL. Obesity and impaired metabolic health increase risk of COVID-19-related mortality in young and middle-aged adults to the level observed in older people: the LEOSS registry. Front Med (Lausanne). 2022;9: 875430. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmed.2022.875430.
Hartmann-Boyce J, Rees K, Onakpoya I, Otunla A, Morris E, Morgan J, Highton P, Suklan J, Curtis F, Goyder C, O’Mahoney L, James O, Sreejith N, Seidu S, Khunti K. An update to the overview of reviews: risks of and from SARS-COV-2 infection and COVID-19 in people with diabetes. Diabetes Care. 2023;46(12):e215–6. https://doiorg.publicaciones.saludcastillayleon.es/10.2337/dc23-1365.
Faltas CL, LeBron KA, Holz MK. Unconventional estrogen signaling in health and disease. Endocrinology. 2020;161(4):bqaa030. https://doiorg.publicaciones.saludcastillayleon.es/10.1210/endocr/bqaa030.
Wang Y, Zhu L, Kuokkanen S, Pollard JW. Activation of protein synthesis in mouse uterine epithelial cells by estradiol-17β is mediated by a PKC-ERK1/2-mTOR signaling pathway. Proc Natl Acad Sci USA. 2015;112(11):E1382–91. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.1418973112.
Lima-Martínez MM, Carrera Boada C, Madera-Silva MD, Marín W, Contreras M. COVID-19 and diabetes: a bidirectional relationship. Clin Investig Arterioscler. 2021;33(3):151–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.arteri.2020.10.001.
Xiong X, Lui DTW, Chung MSH, Au ICH, Lai FTT, Wan EYF, Chui CSL, Li X, Cheng FWT, Cheung CL, Chan EWY, Lee CH, Woo YC, Tan KCB, Wong CKH, Wong ICK. Incidence of diabetes following COVID-19 vaccination and SARS-CoV-2 infection in Hong Kong: a population-based cohort study. PLoS Med. 2023;20(7): e1004274. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pmed.1004274.
Rezel-Potts E, Douiri A, Sun X, Chowienczyk PJ, Shah AM, Gulliford MC. Cardiometabolic outcomes up to 12 months after COVID-19 infection. A matched cohort study in the UK. PLoS Med. 2022; 19(7):e1004052. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pmed.1004052.
Twomey R, DeMars J, Franklin K, Culos-Reed SN, Weatherald J, Wrightson JG. Chronic fatigue and postexertional malaise in people living with long COVID: an observational study. Phys Ther. 2022;102(4):pza005. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/ptj/pzac005.
Bhowal C, Ghosh S, Ghatak D, De R. Pathophysiological involvement of host mitochondria in SARS-CoV-2 infection that causes COVID-19: a comprehensive evidential insight. Mol Cell Biochem. 2023;478(6):1325–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11010-022-04593-z.
Guntur VP, Nemkov T, de Boer E, Mohning MP, Baraghoshi D, Cendali FI, San-Millán I, Petrache I, D’Alessandro A. Signatures of mitochondrial dysfunction and impaired fatty acid metabolism in plasma of patients with post-acute sequelae of COVID-19 (PASC). Metabolites. 2022;12(11):1026. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/metabo12111026.
Guarnieri JW, Angelin A, Murdock DG, Schaefer P, Portluri P, Lie T, Huang J, Wallace DC. SARS-COV-2 viroporins activate the NLRP3-inflammasome by the mitochondrial permeability transition pore. Front Immunol. 2023;14:1064293. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1064293.
Chen TH, Chang CJ, Hung PH. Possible pathogenesis and prevention of long COVID: SARS-CoV-2-induced mitochondrial disorder. Int J Mol Sci. 2023;24(9):8034. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms24098034.
Gordon DE, Jang GM, Bouhaddou M, Xu J, Obernier K, White KM, O’Meara MJ, Rezelj VV, Guo JZ, Swaney DL, Tummino TA, Hüttenhain R, Kaake RM, Richards AL, Tutuncuoglu B, Foussard H, Batra J, Haas K, Modak M, Kim M, Haas P, Polacco BJ, Braberg H, Fabius JM, Eckhardt M, Soucheray M, Bennett MJ, Cakir M, McGregor MJ, Li Q, Meyer B, Roesch F, Vallet T, Mac Kain A, Miorin L, Moreno E, Naing ZZC, Zhou Y, Peng S, Shi Y, Zhang Z, Shen W, Kirby IT, Melnyk JE, Chorba JS, Lou K, Dai SA, Barrio-Hernandez I, Memon D, Hernandez-Armenta C, Lyu J, Mathy CJP, Perica T, Pilla KB, Ganesan SJ, Saltzberg DJ, Rakesh R, Liu X, Rosenthal SB, Calviello L, Venkataramanan S, Liboy-Lugo J, Lin Y, Huang XP, Liu Y, Wankowicz SA, Bohn M, Safari M, Ugur FS, Koh C, Savar NS, Tran QD, Shengjuler D, Fletcher SJ, O’Neal MC, Cai Y, Chang JCJ, Broadhurst DJ, Klippsten S, Sharp PP, Wenzell NA, Kuzuoglu-Ozturk D, Wang HY, Trenker R, Young JM, Cavero DA, Hiatt J, Roth TL, Rathore U, Subramanian A, Noack J, Hubert M, Stroud RM, Frankel AD, Rosenberg OS, Verba KA, Agard DA, Ott M, Emerman M, Jura N, von Zastrow M, Verdin E, Ashworth A, Schwartz O, d’Enfert C, Mukherjee S, Jacobson M, Malik HS, Fujimori DG, Ideker T, Craik CS, Floor SN, Fraser JS, Gross JD, Sali A, Roth BL, Ruggero D, Taunton J, Kortemme T, Beltrao P, Vignuzzi M, García-Sastre A, Shokat KM, Shoichet BK, Krogan NJ. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature. 2020;583(7816):459–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-020-2286-9.
Li S, Zhang J, Liu C, Wang Q, Yan J, Hui L, Jia Q, Shan H, Tao L, Zhang M. The role of mitophagy in regulating cell death. Oxid Med Cell Longev. 2021;2021:6617256. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2021/6617256.
Codo AC, Davanzo GG, Monteiro LB, de Souza GF, Muraro SP, Virgilio-da-Silva JV, Prodonoff JS, Carregari VC, de Biagi Junior CAO, Crunfli F, Jimenez Restrepo JL, Vendramini PH, Reis-de-Oliveira G, Bispo Dos Santos K, Toledo-Teixeira DA, Parise PL, Martini MC, Marques RE, Carmo HR, Borin A, Coimbra LD, Boldrini VO, Brunetti NS, Vieira AS, Mansour E, Ulaf RG, Bernardes AF, Nunes TA, Ribeiro LC, Palma AC, Agrela MV, Moretti ML, Sposito AC, Pereira FB, Velloso LA, Vinolo MAR, Damasio A, Proença-Módena JL, Carvalho RF, Mori MA, Martins-de-Souza D, Nakaya HI, Farias AS, Moraes-Vieira PM. Elevated glucose levels favor SARS-CoV-2 infection and monocyte response through a HIF-1α/glycolysis-dependent axis. Cell Metab. 2020;32(3):437-446e5. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2020.07.007.
Medini H, Zirman A, Mishmar D. Immune system cells from COVID-19 patients display compromised mitochondrial-nuclear expression co-regulation and rewiring toward glycolysis. iScience. 2021;24(12): 103471. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.isci.2021.103471.
Cheng SC, Quintin J, Cramer RA, Shepardson KM, Saeed S, Kumar V, Giamarellos-Bourboulis EJ, Martens JH, Rao NA, Aghajanirefah A, Manjeri GR, Li Y, Ifrim DC, Arts RJ, van der Veer BM, Deen PM, Logie C, O’Neill LA, Willems P, van de Veerdonk FL, van der Meer JW, Ng A, Joosten LA, Wijmenga C, Stunnenberg HG, Xavier RJ, Netea MG. mTOR- and HIF-1α-mediated aerobic glycolysis as metabolic basis for trained immunity. Science. 2014;345(6204):1250684. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.1250684.
Laplante M, Sabatini DM. Regulation of mTORC1 and its impact on gene expression at a glance. J Cell Sci. 2013;126(Pt 8):1713–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1242/jcs.125773.
Pinno J, Bongartz H, Klepsch O, Wundrack N, Poli V, Schaper F, Dittrich A. Interleukin-6 influences stress-signalling by reducing the expression of the mTOR-inhibitor REDD1 in a STAT3-dependent manner. Cell Signal. 2016;28(8):907–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cellsig.2016.04.004.
Köhler N, Wundrack N, Schulz S, Bartonitz F, Schaper F, Dittrich A. Non-canonical STAT3 function reduces REDD1 transcription. FEBS J. 2023;290(7):1765–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/febs.16679.
Jafarzadeh A, Nemati M, Jafarzadeh S. Contribution of STAT3 to the pathogenesis of COVID-19. Microb Pathog. 2021;154: 104836. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.micpath.2021.104836.
Ghosn L, Assi R, Evrenoglou T, Buckley BS, Henschke N, Probyn K, Riveros C, Davidson M, Graña C, Bonnet H, Jarde A, Ávila C, Nejstgaard CH, Menon S, Ferrand G, Kapp P, Breuer C, Schmucker C, Sguassero Y, Boutron I. Interleukin-6 blocking agents for treating COVID-19: a living systematic review. Cochrane Database Syst Rev. 2023;2023(6): CD013881. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/14651858.CD013881.
Writing Committee for the REMAP-CAP Investigators, Higgins AM, Berry LR, Lorenzi E, Murthy S, McQuilten Z, Mouncey PR, Al-Beidh F, Annane D, Arabi YM, Beane A, van Bentum-Puijk W, Bhimani Z, Bonten MJM, Bradbury CA, Brunkhorst FM, Burrell A, Buzgau A, Buxton M, Charles WN, Cove M, Detry MA, Estcourt LJ, Fagbodun EO, Fitzgerald M, Girard TD, Goligher EC, Goossens H, Haniffa R, Hills T, Horvat CM, Huang DT, Ichihara N, Lamontagne F, Marshall JC, McAuley DF, McGlothlin A, McGuinness SP, McVerry BJ, Neal MD, Nichol AD, Parke RL, Parker JC, Parry-Billings K, Peters SEC, Reyes LF, Rowan KM, Saito H, Santos MS, Saunders CT, Serpa-Neto A, Seymour CW, Shankar-Hari M, Stronach LM, Turgeon AF, Turner AM, van de Veerdonk FL, Zarychanski R, Green C, Lewis RJ, Angus DC, McArthur CJ, Berry S, Derde LPG, Gordon AC, Webb SA, Lawler PR. Long-term (180-Day) outcomes in critically ill patients with COVID-19 in the REMAP-CAP randomized clinical trial. JAMA. 2023;329(1):39–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1001/jama.2022.23257.
Phetsouphanh C, Darley DR, Wilson DB, Howe A, Munier CML, Patel SK, Juno JA, Burrell LM, Kent SJ, Dore GJ, Kelleher AD, Matthews GV. Immunological dysfunction persists for 8 months following initial mild-to-moderate SARS-CoV-2 infection. Nat Immunol. 2022;23(2):210–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41590-021-01113-x.
Klein J, Wood J, Jaycox JR, Dhodapkar RM, Lu P, Gehlhausen JR, Tabachnikova A, Greene K, Tabacof L, Malik AA, Silva Monteiro V, Silva J, Kamath K, Zhang M, Dhal A, Ott IM, Valle G, Peña-Hernández M, Mao T, Bhattacharjee B, Takahashi T, Lucas C, Song E, McCarthy D, Breyman E, Tosto-Mancuso J, Dai Y, Perotti E, Akduman K, Tzeng TJ, Xu L, Geraghty AC, Monje M, Yildirim I, Shon J, Medzhitov R, Lutchmansingh D, Possick JD, Kaminski N, Omer SB, Krumholz HM, Guan L, Dela Cruz CS, van Dijk D, Ring AM, Putrino D, Iwasaki A. Distinguishing features of long COVID identified through immune profiling. Nature. 2023;623(7985):139–48. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-023-06651-y.
Altmann DM, Whettlock EM, Liu S, Arachchillage DJ, Boyton RJ. The immunology of long COVID. Nat Rev Immunol. 2023;23(10):618–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41577-023-00904-7.
Taquet M, Skorniewska Z, Hampshire A, Chalmers JD, Ho LP, Horsley A, Marks M, Poinasamy K, Raman B, Leavy OC, Richardson M, Elneima O, McAuley HJC, Shikotra A, Singapuri A, Sereno M, Saunders RM, Harris VC, Houchen-Wolloff L, Greening NJ, Mansoori P, Harrison EM, Docherty AB, Lone NI, Quint J, Sattar N, Brightling CE, Wain LV, Evans RE, Geddes JR, Harrison PJ, PHOSP-COVID Study Collaborative Group. Acute blood biomarker profiles predict cognitive deficits 6 and 12 months after COVID-19 hospitalization. Nat Med. 2023;29(10):2498–508. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41591-023-02525-y.
Holms RD. Long COVID (PASC) is maintained by a self-sustaining pro-inflammatory TLR4/RAGE-Loop of S100A8/A9 > TLR4/RAGE signalling, inducing chronic expression of IL-1b, IL-6 and TNFa: anti-inflammatory ezrin peptides as potential therapy. Immuno. 2022;2:512–33. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/immuno2030033.
Schultheiß C, Willscher E, Paschold L, Gottschick C, Klee B, Henkes SS, Bosurgi L, Dutzmann J, Sedding D, Frese T, Girndt M, Höll JI, Gekle M, Mikolajczyk R, Binder M. The IL-1β, IL-6, and TNF cytokine triad is associated with post-acute sequelae of COVID-19. Cell Rep Med. 2022;3(6): 100663. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.xcrm.2022.100663.
Yin JX, Agbana YL, Sun ZS, Fei SW, Zhao HQ, Zhou XN, Chen JH, Kassegne K. Increased interleukin-6 is associated with long COVID-19: a systematic review and meta-analysis. Infect Dis Poverty. 2023;12(1):43. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40249-023-01086-z.
Cervia-Hasler C, Brüningk SC, Hoch T, Fan B, Muzio G, Thompson RC, Ceglarek L, Meledin R, Westermann P, Emmenegger M, Taeschler P, Zurbuchen Y, Pons M, Menges D, Ballouz T, Cervia-Hasler S, Adamo S, Merad M, Charney AW, Puhan M, Brodin P, Nilsson J, Aguzzi A, Raeber ME, Messner CB, Beckmann ND, Borgwardt K, Boyman O. Persistent complement dysregulation with signs of thromboinflammation in active Long Covid. Science. 2024;383(6680):eadg7942. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.adg7942.
Lindenkamp C, Plümers R, Osterhage MR, Vanakker OM, Van Wynsberghe J, Knabbe C, Hendig D. The activation of JAK/STAT3 signaling and the complement system modulate inflammation in the primary human dermal fibroblasts of PXE patients. Biomedicines. 2023;11(10):2673. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biomedicines11102673.
Dutta D, Liu J, Xiong H. NLRP3 inflammasome activation and SARS-CoV-2-mediated hyperinflammation, cytokine storm and neurological syndromes. Int J Physiol Pathophysiol Pharmacol. 2022;14(3):138–60.
Zhu L, Wang Z, Sun X, Yu J, Li T, Zhao H, Ji Y, Peng B, Du M. STAT3/mitophagy axis coordinates macrophage NLRP3 inflammasome activation and inflammatory bone loss. J Bone Miner Res. 2023;38(2):335–53. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jbmr.4756.
Wong AC, Devason AS, Umana IC, Cox TO, Dohnalová L, Litichevskiy L, Perla J, Lundgren P, Etwebi Z, Izzo LT, Kim J, Tetlak M, Descamps HC, Park SL, Wisser S, McKnight AD, Pardy RD, Kim J, Blank N, Patel S, Thum K, Mason S, Beltra JC, Michieletto MF, Ngiow SF, Miller BM, Liou MJ, Madhu B, Dmitrieva-Posocco O, Huber AS, Hewins P, Petucci C, Chu CP, Baraniecki-Zwil G, Giron LB, Baxter AE, Greenplate AR, Kearns C, Montone K, Litzky LA, Feldman M, Henao-Mejia J, Striepen B, Ramage H, Jurado KA, Wellen KE, O’Doherty U, Abdel-Mohsen M, Landay AL, Keshavarzian A, Henrich TJ, Deeks SG, Peluso MJ, Meyer NJ, Wherry EJ, Abramoff BA, Cherry S, Thaiss CA, Levy M. Serotonin reduction in post-acute sequelae of viral infection. Cell. 2023;186(22):4851-4867.e20. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2023.09.013.
Jensterle M, Herman R, Janež A, Mahmeed WA, Al-Rasadi K, Al-Alawi K, Banach M, Banerjee Y, Ceriello A, Cesur M, Cosentino F, Galia M, Goh SY, Kalra S, Kempler P, Lessan N, Lotufo P, Papanas N, Rizvi AA, Santos RD, Stoian AP, Toth PP, Viswanathan V, Rizzo M. The relationship between COVID-19 and hypothalamic–pituitary–adrenal axis: a large spectrum from glucocorticoid insufficiency to excess—the CAPISCO International Expert Panel. Int J Mol Sci. 2022;23(13):7326. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms23137326.
Tuijnenburg P, AandeKerk DJ, Jansen MH, Morris B, Lieftink C, Beijersbergen RL, van Leeuwen EMM, Kuijpers TW. High-throughput compound screen reveals mTOR inhibitors as potential therapeutics to reduce (auto)antibody production by human plasma cells. Eur J Immunol. 2020;50(1):73–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/eji.201948241.
Puntmann VO, Carerj ML, Wieters I, Fahim M, Arendt C, Hoffmann J, Shchendrygina A, Escher F, Vasa-Nicotera M, Zeiher AM, Vehreschild M, Nagel E. Outcomes of cardiovascular magnetic resonance imaging in patients recently recovered from coronavirus disease 2019 (COVID-19). JAMA Cardiol. 2020;5(11):1265–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1001/jamacardio.2020.3557.
Sciarretta S, Volpe M, Sadoshima J. Mammalian target of rapamycin signaling in cardiac physiology and disease. Circ Res. 2014;114(3):549–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/CIRCRESAHA.114.302022.
Katsoularis I, Fonseca-Rodríguez O, Farrington P, Jerndal H, Lundevaller EH, Sund M, Lindmark K, Fors Connolly AM. Risks of deep vein thrombosis, pulmonary embolism, and bleeding after covid-19: nationwide self-controlled cases series and matched cohort study. BMJ. 2022;377: e069590. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/bmj-2021-069590.
Yang J, Zhou X, Fan X, Xiao M, Yang D, Liang B, Dai M, Shan L, Lu J, Lin Z, Liu R, Liu J, Wang L, Zhong M, Jiang Y, Bai X. mTORC1 promotes aging-related venous thrombosis in mice via elevation of platelet volume and activation. Blood. 2016;128(5):615–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2015-10-672964.
Patel MA, Knauer MJ, Nicholson M, Daley M, Van Nynatten LR, Martin C, Patterson EK, Cepinskas G, Seney SL, Dobretzberger V, Miholits M, Webb B, Fraser DD. Elevated vascular transformation blood biomarkers in Long-COVID indicate angiogenesis as a key pathophysiological mechanism. Mol Med. 2022;28(1):122. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10020-022-00548-8.
Lotfimehr H, Mardi N, Narimani S, Nasrabadi HT, Karimipour M, Sokullu E, Rahbarghazi R. mTOR signalling pathway in stem cell bioactivities and angiogenesis potential. Cell Prolif. 2023;56(12): e13499. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cpr.13499.
Lassan S, Tesar T, Tisonova J, Lassanova M. Pharmacological approaches to pulmonary fibrosis following COVID-19. Front Pharmacol. 2023;14:1143158. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2023.1143158.
Platé M, Guillotin D, Chambers RC. The promise of mTOR as a therapeutic target pathway in idiopathic pulmonary fibrosis. Eur Respir Rev. 2020;29(157): 200269. https://doiorg.publicaciones.saludcastillayleon.es/10.1183/16000617.0269-2020.
Gothe F, Gehrig J, Rapp CK, Knoflach K, Reu-Hofer S, Länger F, Schramm D, Ley-Zaporozhan J, Ehl S, Schwerk N, Faletti L, Griese M. Early-onset, fatal interstitial lung disease in STAT3 gain-of-function patients. Pediatr Pulmonol. 2021;56(12):3934–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ppul.25684.
Mahdi A, Zhao A, Fredengren E, Fedorowski A, Braunschweig F, Nygren-Bonnier M, Runold M, Bruchfeld J, Nickander J, Deng Q, Checa A, Desta L, Pernow J, Ståhlberg M. Dysregulations in hemostasis, metabolism, immune response, and angiogenesis in post-acute COVID-19 syndrome with and without postural orthostatic tachycardia syndrome: a multi-omic profiling study. Sci Rep. 2023;13(1):20230. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-023-47539-1.
Harlan SM, Guo DF, Morgan DA, Fernandes-Santos C, Rahmouni K. Hypothalamic mTORC1 signaling controls sympathetic nerve activity and arterial pressure and mediates leptin effects. Cell Metab. 2013;17(4):599–606. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2013.02.017.
Nicolai L, Kaiser R, Stark K. Thromboinflammation in long COVID-the elusive key to postinfection sequelae? J Thromb Haemost. 2023;21(8):2020–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jtha.2023.04.039.
McConnell MJ, Kawaguchi N, Kondo R, Sonzogni A, Licini L, Valle C, Bonaffini PA, Sironi S, Alessio MG, Previtali G, Seghezzi M, Zhang X, Lee AI, Pine AB, Chun HJ, Zhang X, Fernandez-Hernando C, Qing H, Wang A, Price C, Sun Z, Utsumi T, Hwa J, Strazzabosco M, Iwakiri Y. Liver injury in COVID-19 and IL-6 trans-signaling-induced endotheliopathy. J Hepatol. 2021;75(3):647–58. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jhep.2021.04.050.
Al-Kuraishy HM, Al-Gareeb AI, Alarfaj SJ, Al-Akeel RK, Faidah H, El-Bouseary MM, Sabatier JM, De Waard M, El-Masry TA, Batiha GE. Long COVID and risk of erectile dysfunction in recovered patients from mild to moderate COVID-19. Sci Rep. 2023;13(1):5977. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-023-32211-5.
Yan JF, Huang WJ, Zhao JF, Fu HY, Zhang GY, Huang XJ, Lv BD. The platelet-derived growth factor receptor/STAT3 signaling pathway regulates the phenotypic transition of corpus cavernosum smooth muscle in rats. PLoS ONE. 2017;12(2): e0172191. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0172191.
Leng A, Shah M, Ahmad SA, Premraj L, Wildi K, Li Bassi G, Pardo CA, Choi A, Cho SM. Pathogenesis underlying neurological manifestations of long COVID syndrome and potential therapeutics. Cells. 2023;12(5):816. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells12050816.
Low RN, Low RJ, Akrami A. A review of cytokine-based pathophysiology of long COVID symptoms. Front Med (Lausanne). 2023;10:1011936. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmed.2023.1011936.
Reiss AB, Greene C, Dayaramani C, Rauchman SH, Stecker MM, De Leon J, Pinkhasov A. Long COVID, the brain, nerves, and cognitive function. Neurol Int. 2023;15(3):821–41. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/neurolint15030052.
Keane L, Antignano I, Riechers SP, Zollinger R, Dumas AA, Offermann N, Bernis ME, Russ J, Graelmann F, McCormick PN, Esser J, Tejera D, Nagano A, Wang J, Chelala C, Biederbick Y, Halle A, Salomoni P, Heneka MT, Capasso M. mTOR-dependent translation amplifies microglia priming in aging mice. J Clin Invest. 2021;131(1): e132727. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI132727.
Millot P, San C, Bennana E, Porte B, Vignal N, Hugon J, Paquet C, Hosten B, Mouton-Liger F. STAT3 inhibition protects against neuroinflammation and BACE1 upregulation induced by systemic inflammation. Immunol Lett. 2020;228:129–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.imlet.2020.10.004.
Abrams RMC, Simpson DM, Navis A, Jette N, Zhou L, Shin SC. Small fiber neuropathy associated with SARS-CoV-2 infection. Muscle Nerve. 2022;65(4):440–3. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/mus.27458.
Kim K, Choi S, Cha M, Lee BH. Effects of mTOR inhibitors on neuropathic pain revealed by optical imaging of the insular cortex in rats. Brain Res. 2020;1733: 146720. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.brainres.2020.146720.
Marino Y, Arangia A, Cordaro M, Siracusa R, D’Amico R, Impellizzeri D, Cupi R, Peritore AF, Gugliandolo E, Fusco R, et al. Analysis of the Influence of IL-6 and the activation of the Jak/Stat3 pathway in fibromyalgia. Biomedicines. 2023;11:792. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biomedicines11030792.
Maloney EM, Boneva RS, Lin JM, Reeves WC. Chronic fatigue syndrome is associated with metabolic syndrome: results from a case–control study in Georgia. Metabolism. 2010;59(9):1351–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.metabol.2009.12.019.
Tomic S, Brkic S, Maric D, Mikic AN. Lipid and protein oxidation in female patients with chronic fatigue syndrome. Arch Med Sci. 2012;8(5):886–91. https://doiorg.publicaciones.saludcastillayleon.es/10.5114/aoms.2012.31620.
Flores S, Brown A, Adeoye S, Jason LA, Evans M. Examining the impact of obesity on individuals with chronic fatigue syndrome. Workplace Health Saf. 2013;61(7):299–307. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/216507991306100705.
Kalra S, Sahay R. Diabetes fatigue syndrome. Diabetes Ther. 2018;9(4):1421–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13300-018-0453-x.
Perakakis N, Harb H, Hale BG, Varga Z, Steenblock C, Kanczkowski W, Alexaki VI, Ludwig B, Mirtschink P, Solimena M, Toepfner N, Zeissig S, Gado M, Abela IA, Beuschlein F, Spinas GA, Cavelti-Weder C, Gerber PA, Huber M, Trkola A, Puhan MA, Wong WW, Linkermann A, Mohan V, Lehnert H, Nawroth P, Chavakis T, Mingrone G, Wolfrum C, Zinkernagel AS, Bornstein SR. Mechanisms and clinical relevance of the bidirectional relationship of viral infections with metabolic diseases. Lancet Diabetes Endocrinol. 2023;11(9):675–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S2213-8587(23)00154-7.
Myhill S, Booth NE, McLaren-Howard J. Chronic fatigue syndrome and mitochondrial dysfunction. Int J Clin Exp Med. 2009;2(1):1–16.
Morris G, Maes M. Mitochondrial dysfunctions in myalgic encephalomyelitis/chronic fatigue syndrome explained by activated immuno-inflammatory, oxidative and nitrosative stress pathways. Metab Brain Dis. 2014;29(1):19–36. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11011-013-9435-x.
Holden S, Maksoud R, Eaton-Fitch N, Cabanas H, Staines D, Marshall-Gradisnik S. A systematic review of mitochondrial abnormalities in myalgic encephalomyelitis/chronic fatigue syndrome/systemic exertion intolerance disease. J Transl Med. 2020;18(1):290. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-020-02452-3.
Tomas C, Brown A, Strassheim V, Elson JL, Newton J, Manning P. Cellular bioenergetics is impaired in patients with chronic fatigue syndrome. PLoS ONE. 2017;12(10): e0186802. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0186802.
Tomas C, Newton J. Metabolic abnormalities in chronic fatigue syndrome/myalgic encephalomyelitis: a mini-review. Biochem Soc Trans. 2018;46(3):547–53. https://doiorg.publicaciones.saludcastillayleon.es/10.1042/BST20170503.
Sweetman E, Kleffmann T, Edgar C, de Lange M, Vallings R, Tate W. A SWATH-MS analysis of myalgic encephalomyelitis/chronic fatigue syndrome peripheral blood mononuclear cell proteomes reveals mitochondrial dysfunction. J Transl Med. 2020;18(1):365. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-020-02533-3.
Paul BD, Lemle MD, Komaroff AL, Snyder SH. Redox imbalance links COVID-19 and myalgic encephalomyelitis/chronic fatigue syndrome. Proc Natl Acad Sci USA. 2021;118(34): e2024358118. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.2024358118.
Gorman GS, Elson JL, Newman J, Payne B, McFarland R, Newton JL, Turnbull DM. Perceived fatigue is highly prevalent and debilitating in patients with mitochondrial disease. Neuromuscul Disord. 2015;25(7):563–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.nmd.2015.03.001.
Schoeman EM, Van Der Westhuizen FH, Erasmus E, van Dyk E, Knowles CV, Al-Ali S, Ng WF, Taylor RW, Newton JL, Elson JL. Clinically proven mtDNA mutations are not common in those with chronic fatigue syndrome. BMC Med Genet. 2017;18(1):29. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12881-017-0387-6.
Tomas C. Elson JL The role of mitochondria in ME/CFS: a perspective, fatigue. Biomed Health Behav. 2019;7(1):52–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/21641846.2019.1580855.
Fluge Ø, Mella O, Bruland O, Risa K, Dyrstad SE, Alme K, Rekeland IG, Sapkota D, Røsland GV, Fosså A, Ktoridou-Valen I, Lunde S, Sørland K, Lien K, Herder I, Thürmer H, Gotaas ME, Baranowska KA, Bohnen LM, Schäfer C, McCann A, Sommerfelt K, Helgeland L, Ueland PM, Dahl O, Tronstad KJ. Metabolic profiling indicates impaired pyruvate dehydrogenase function in myalgic encephalopathy/chronic fatigue syndrome. JCI Insight. 2016;1(21): e89376. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci.insight.89376.
Lien K, Johansen B, Veierød MB, Haslestad AS, Bøhn SK, Melsom MN, Kardel KR, Iversen PO. Abnormal blood lactate accumulation during repeated exercise testing in myalgic encephalomyelitis/chronic fatigue syndrome. Physiol Rep. 2019;7(11): e14138. https://doiorg.publicaciones.saludcastillayleon.es/10.14814/phy2.14138.
Mueller C, Lin JC, Sheriff S, Maudsley AA, Younger JW. Evidence of widespread metabolite abnormalities in Myalgic encephalomyelitis/chronic fatigue syndrome: assessment with whole-brain magnetic resonance spectroscopy. Brain Imaging Behav. 2020;14(2):562–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11682-018-0029-4.
Tomas C, Elson JL, Strassheim V, Newton JL, Walker M. The effect of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) severity on cellular bioenergetic function. PLoS ONE. 2020;15(4): e0231136. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0231136.
Ohba T, Domoto S, Tanaka M, Nakamura S, Shimazawa M, Hara H. Myalgic encephalomyelitis/chronic fatigue syndrome induced by repeated forced swimming in mice. Biol Pharm Bull. 2019;42(7):1140–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1248/bpb.b19-00009.
Gottschalk G, Peterson D, Knox K, Maynard M, Whelan RJ. Elevated ATG13 in serum of patients with ME/CFS stimulates oxidative stress response in microglial cells via activation of receptor for advanced glycation end products (RAGE). Mol Cell Neurosci. 2022;120:103731. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.mcn.2022.103731.
Kim JW, Tchernyshyov I, Semenza GL, Dang CV. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 2006;3(3):177–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2006.02.002.
Mandarano AH, Maya J, Giloteaux L, Peterson DL, Maynard M, Gottschalk CG, Hanson MR. Myalgic encephalomyelitis/chronic fatigue syndrome patients exhibit altered T cell metabolism and cytokine associations. J Clin Invest. 2020;130(3):1491–505. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI132185.
Muri J, Kopf M. Redox regulation of immunometabolism. Nat Rev Immunol. 2021;21(6):363–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41577-020-00478-8.
Ando S, Perkins CM, Sajiki Y, Chastain C, Valanparambil RM, Wieland A, Hudson WH, Hashimoto M, Ramalingam SS, Freeman GJ, Ahmed R, Araki K. mTOR regulates T cell exhaustion and PD-1-targeted immunotherapy response during chronic viral infection. J Clin Invest. 2023;133(2): e160025. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI160025.
Chen Y, Xu Z, Sun H, Ouyang X, Han Y, Yu H, Wu N, Xie Y, Su B. Regulation of CD8+ T memory and exhaustion by the mTOR signals. Cell Mol Immunol. 2023;20(9):1023–39. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41423-023-01064-3.
Iu DS, Maya J, Vu LT, Fogarty EA, McNairn AJ, Ahmed F, Franconi CJ, Munn PR, Grenier JK, Hanson MR, Grimson A. Transcriptional reprogramming primes CD8+ T cells toward exhaustion in Myalgic encephalomyelitis/chronic fatigue syndrome. Proc Natl Acad Sci USA. 2024;121(50): e2415119121. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.2415119121.
Eaton-Fitch N, Rudd P, Er T, Hool L, Herrero L, Marshall-Gradisnik S. Immune exhaustion in ME/CFS and long COVID. JCI Insight. 2024;9(20): e183810. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci.insight.183810.
Renz-Polster H, Tremblay ME, Bienzle D, Fischer JE. The pathobiology of myalgic encephalomyelitis/chronic fatigue syndrome: the case for neuroglial failure. Front Cell Neurosci. 2022;16: 888232. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fncel.2022.888232.
Tate W, Walker M, Sweetman E, Helliwell A, Peppercorn K, Edgar C, Blair A, Chatterjee A. Molecular mechanisms of neuroinflammation in ME/CFS and long COVID to sustain disease and promote relapses. Front Neurol. 2022;13: 877772. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fneur.2022.877772.
Tate WP, Walker MOM, Peppercorn K, Blair ALH, Edgar CD. Towards a better understanding of the complexities of myalgic encephalomyelitis/chronic fatigue syndrome and long COVID. Int J Mol Sci. 2023;24(6):5124. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms24065124.
von Delft A, Hall MD, Kwong AD, Purcell LA, Saikatendu KS, Schmitz U, Tallarico JA, Lee AA. Accelerating antiviral drug discovery: lessons from COVID-19. Nat Rev Drug Discov. 2023;22(7):585–603. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41573-023-00692-8.
Ghosn L, Assi R, Evrenoglou T, Buckley BS, Henschke N, Probyn K, Riveros C, Davidson M, Graña C, Bonnet H, Jarde A, Ávila C, Nejstgaard CH, Menon S, Ferrand G, Kapp P, Breuer C, Schmucker C, Sguassero Y, Nguyen TV, Devane D, Meerpohl JJ, Rada G, Hróbjartsson A, Grasselli G, Tovey D, Ravaud P, Chaimani A, Boutron I. Interleukin-6 blocking agents for treating COVID-19: a living systematic review. Cochrane Database Syst Rev. 2023;6(6):CD013881. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/14651858.CD013881.pub2.
RECOVERY Collaborative Group, Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, Linsell L, Staplin N, Brightling C, Ustianowski A, Elmahi E, Prudon B, Green C, Felton T, Chadwick D, Rege K, Fegan C, Chappell LC, Faust SN, Jaki T, Jeffery K, Montgomery A, Rowan K, Juszczak E, Baillie JK, Haynes R, Landray MJ. Dexamethasone in hospitalized patients with Covid-19. N Engl J Med. 2021;384(8):693–704. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa2021436.
McCorkell L, Peluso MJ. Long COVID research risks losing momentum—we need a moonshot. Nature. 2023;622(7983):457–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/d41586-023-03225-w.
Scott A, Ansari W, Chambers R, Reimbaeva M, Mikolajczyk T, Benigno M, Draica F, Atkinson J. Substantial health and economic burden of COVID-19 during the year after acute illness among US adults not at high risk of severe COVID-19. BMC Med. 2024;22(1):47. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12916-023-03235-5.
Jason LA, Mirin AA. Updating the National Academy of Medicine ME/CFS prevalence and economic impact figures to account for population growth and inflation. Fatigue Biomed Health Behav. 2021;9(1):9–13. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/21641846.2021.1878716.
Patocka J, Kuca K, Oleksak P, Nepovimova E, Valis M, Novotny M, Klimova B. Rapamycin: drug repurposing in SARS-CoV-2 infection. Pharmaceuticals (Basel). 2021;14(3):217. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ph14030217.
https://www.healthrising.org/blog/2023/11/14/rapamycin-chronic-fatigue-syndrome-mitochondria-trial/.
https://www.healthrising.org/blog/2022/07/06/apamycin-resurgence-doctor-chronic-fatigue-syndrome/.
Kim MJ, Yoon JH, Ryu JH. Mitophagy: a balance regulator of NLRP3 inflammasome activation. BMB Rep. 2016;49(10):529–35. https://doiorg.publicaciones.saludcastillayleon.es/10.5483/bmbrep.2016.49.10.115.
Sliter DA, Martinez J, Hao L, Chen X, Sun N, Fischer TD, Burman JL, Li Y, Zhang Z, Narendra DP, Cai H, Borsche M, Klein C, Youle RJ. Parkin and PINK1 mitigate STING-induced inflammation. Nature. 2018;561(7722):258–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-018-0448-9.
Harris J, Deen N, Zamani S, Hasnat MA. Mitophagy and the release of inflammatory cytokines. Mitochondrion. 2018;41:2–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.mito.2017.10.009.
Cho DH, Kim JK, Jo EK. Mitophagy and innate immunity in infection. Mol Cells. 2020;43(1):10–22. https://doiorg.publicaciones.saludcastillayleon.es/10.14348/molcells.2020.2329.
Wang Q, Xue H, Yue Y, Hao S, Huang SH, Zhang Z. Role of mitophagy in the neurodegenerative diseases and its pharmacological advances: a review. Front Mol Neurosci. 2022;15:1014251. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnmol.2022.1014251.
Vergès B, Cariou B. mTOR inhibitors and diabetes. Diabetes Res Clin Pract. 2015;110(2):101–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.diabres.2015.09.014.
Choo AY, Yoon SO, Kim SG, Roux PP, Blenis J. Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc Natl Acad Sci USA. 2008;105:17414–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.0809136105.
Palermo A, Li S, Ten Hoeve J, Chellappa A, Morris A, Dillon B, Ma F, Wang Y, Cao E, Shabane B, Acín-Perez R, Petcherski A, Lusis AJ, Hazen S, Shirihai OS, Pellegrini M, Arumugaswami V, Graeber TG, Deb A. A ketogenic diet can mitigate SARS-CoV-2 induced systemic reprogramming and inflammation. Commun Biol. 2023;6(1):1115. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s42003-023-05478-7.
Barrea L, Grant WB, Frias-Toral E, Vetrani C, Verde L, de Alteriis G, Docimo A, Savastano S, Colao A, Muscogiuri G. Dietary recommendations for post-COVID-19 syndrome. Nutrients. 2022;14(6):1305. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/nu14061305.
Zilberman-Itskovich S, Catalogna M, Sasson E, Elman-Shina K, Hadanny A, Lang E, Finci S, Polak N, Fishlev G, Korin C, Shorer R, Parag Y, Sova M, Efrati S. Hyperbaric oxygen therapy improves neurocognitive functions and symptoms of post-COVID condition: randomized controlled trial. Sci Rep. 2022;12(1):11252. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-022-15565-0.
Eckl EM, Ziegemann O, Krumwiede L, Fessler E, Jae LT. Sensing, signaling and surviving mitochondrial stress. Cell Mol Life Sci. 2021;78(16):5925–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00018-021-03887-7.
Winter JM, Yadav T, Rutter J. Stressed to death: mitochondrial stress responses connect respiration and apoptosis in cancer. Mol Cell. 2022;82(18):3321–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molcel.2022.07.012.
Schönfeld P, Bohnensack R. Fatty acid-promoted mitochondrial permeability transition by membrane depolarization and binding to the ADP/ATP carrier. FEBS Lett. 1997;420(2–3):167–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0014-5793(97)01511-1.
Liu D, Zhang Y, Gharavi R, Park HR, Lee J, Siddiqui S, Telljohann R, Nassar MR, Cutler RG, Becker KG, Mattson MP. The mitochondrial uncoupler DNP triggers brain cell mTOR signaling network reprogramming and CREB pathway up-regulation. J Neurochem. 2015;134(4):677–92. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jnc.13176.
Romaschenko VP, Zinovkin RA, Galkin II, Zakharova VV, Panteleeva AA, Tokarchuk AV, Lyamzaev KG, Pletjushkina OY, Chernyak BV, Popova EN. Low concentrations of uncouplers of oxidative phosphorylation prevent inflammatory activation of endothelial cells by tumor necrosis factor. Biochemistry (Mosc). 2015;80(5):610–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1134/S0006297915050144.
Lyamzaev KG, Tokarchuk AV, Panteleeva AA, Mulkidjanian AY, Skulachev VP, Chernyak BV. Induction of autophagy by depolarization of mitochondria. Autophagy. 2018;14(5):921–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/15548627.2018.1436937.
Demine S, Renard P, Arnould T. Mitochondrial uncoupling: a key controller of biological processes in physiology and diseases. Cells. 2019;8(8):795. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells8080795.
Du Y, Zhu YJ, Zhou YX, Ding J, Liu JY. Metformin in therapeutic applications in human diseases: its mechanism of action and clinical study. Mol Biomed. 2022;3(1):41. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s43556-022-00108-w.
Bramante CT, Buse JB, Liebovitz DM, Nicklas JM, Puskarich MA, Cohen K, Belani HK, Anderson BJ, Huling JD, Tignanelli CJ, Thompson JL, Pullen M, Wirtz EL, Siegel LK, Proper JL, Odde DJ, Klatt NR, Sherwood NE, Lindberg SM, Karger AB, Beckman KB, Erickson SM, Fenno SL, Hartman KM, Rose MR, Mehta T, Patel B, Griffiths G, Bhat NS, Murray TA, Boulware DR, COVID-OUT Study Team. Outpatient treatment of COVID-19 and incidence of post-COVID-19 condition over 10 months (COVID-OUT): a multicentre, randomised, quadruple-blind, parallel-group, phase 3 trial. Lancet Infect Dis. 2023;23(10):1119–29. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S1473-3099(23)00299-2.
Howell JJ, Hellberg K, Turner M, Talbott G, Kolar MJ, Ross DS, Hoxhaj G, Saghatelian A, Shaw RJ, Manning BD. Metformin inhibits hepatic mTORC1 signaling via dose-dependent mechanisms involving AMPK and the TSC complex. Cell Metab. 2017;25(2):463–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2016.12.009.
Rottenberg H, Hoek JB. The mitochondrial permeability transition: nexus of aging, disease and longevity. Cells. 2021;10(1):79. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells10010079.
Blagosklonny MV. Hypoxia, MTOR and autophagy: converging on senescence or quiescence. Autophagy. 2013;9(2):260–2. https://doiorg.publicaciones.saludcastillayleon.es/10.4161/auto.22783.
Chun Y, Kim J. AMPK-mTOR signaling and cellular adaptations in hypoxia. Int J Mol Sci. 2021;22(18):9765. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms22189765.
Aisen Y, Gatt ME, Hertz R, Smeir E, Bar-Tana J. Suppression of multiple myeloma by mitochondrial targeting. Sci Rep. 2021;11(1):5862. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-021-83829-2.
Eldad S, Hertz R, Vainer G, Saada A, Bar-Tana J. Treatment of ErbB2 breast cancer by mitochondrial targeting. Cancer Metab. 2020;8:17. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40170-020-00223-8.
Samovski D, Kalderon B, Yehuda-Shnaidman E, Bar-Tana J. Gating of the mitochondrial permeability transition pore by long chain fatty acyl analogs in vivo. J Biol Chem. 2010;285(10):6879–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.M109.080416.
Goul C, Peruzzo R, Zoncu R. The molecular basis of nutrient sensing and signalling by mTORC1 in metabolism regulation and disease. Nat Rev Mol Cell Biol. 2023;24(12):857–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41580-023-00641-8.
Shi D, Tao J, Man S, Zhang N, Ma L, Guo L, Huang L, Gao W. Structure, function, signaling pathways and clinical therapeutics: The translational potential of STAT3 as a target for cancer therapy. Biochim Biophys Acta Rev Cancer. 2024;1879(6): 189207. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbcan.2024.189207.
Schäfer A, Leist SR, Powers JM, Baric RS. Animal models of Long Covid: a hit-and-run disease. Sci Transl Med. 2024;16(773):eado2104. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/scitranslmed.ado2104.
Tamura Y, Yamato M, Kataoka Y. Animal models for neuroinflammation and potential treatment methods. Front Neurol. 2022;27(13): 890217. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fneur.2022.890217.
Clauw DJ, Calabrese L. Rheumatology and long COVID: lessons from the study of fibromyalgia. Ann Rheum Dis. 2024;83(2):136–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/ard-2023-224250.
Ramírez-Morales R, Bermúdez-Benítez E, Martínez-Martínez LA, Martínez-Lavín M. Clinical overlap between fibromyalgia and myalgic encephalomyelitis. A systematic review and meta-analysis. Autoimmun Rev. 2022;21(8): 103129. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.autrev.2022.103129.
Haider S, Janowski AJ, Lesnak JB, Hayashi K, Dailey DL, Chimenti R, Frey-Law LA, Sluka KA, Berardi G. A comparison of pain, fatigue, and function between post-COVID-19 condition, fibromyalgia, and chronic fatigue syndrome: a survey study. Pain. 2023;164(2):385–401. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/j.pain.0000000000002711.
Acknowledgements
Not applicable.
Funding
Funded by Hebrew University Medical School internal grant.
Author information
Authors and Affiliations
Contributions
Not applicable.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Bar-Tana, J. mTORC1 syndrome (TorS): unifying paradigm for PASC, ME/CFS and PAIS. J Transl Med 23, 297 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06220-z
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06220-z