1
|
Parak M, Asgari A, Hasani Nourian Y, Ghanei M. A review of poisoning with various types of biotoxins and its common clinical symptoms. Toxicon 2024; 240:107629. [PMID: 38336277 DOI: 10.1016/j.toxicon.2024.107629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 01/01/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024]
Abstract
INTRODUCTION Biotoxins are toxic substances that originate from living organisms and are harmful to humans. Therefore, we need to know the symptoms of biotoxins poisoning to manage the damage. The purpose of this study is to establish a practical diagnostic protocol for dealing with poisoned patients exposed to biotoxins. MATERIALS AND METHODS The present study is a review study. Our studied community is articles and books matching the title of the project and relevant keywords. First, by searching the key words sign, symptom, biotoxins, relevant articles were extracted and studied from valid databases. By reviewing the studies based on the search strategy, four groups of biotoxins that were studied the most were identified. These four groups are marine biotoxins, bacterial biotoxins, fungal biotoxins and plant biotoxins. In each of these biotoxin groups, important toxins were selected and studied. RESULTS A total of 1864 articles were initially identified from the databases searched in present study. After screening titles and abstracts, 26 articles were included in the systematic review. Specifically, 7 articles were included for bacterial toxins, 9 articles for marine toxins, 5 articles for plant toxins and 5 articles for fungal toxins. CONCLUSION The symptoms of plant biotoxins poisoning may include cardiovascular, hematologic, neurologic, respiratory, renal, and gastrointestinal symptoms, while the symptoms of fungal biotoxins poisoning may include hepatic, renal, gastrointestinal, musculoskeletal, metabolic, respiratory, neurological, and cardiovascular symptoms. marine biotoxins poisoning presents with gastrointestinal and neurological symptoms, with varying incubation periods and recovery times. bacterial biotoxins exposure can lead to a wide range of clinical symptoms, with diarrhea, vomiting, and abdominal pain being the most common, and hemoglobinuria or hematuria being a sensitive and specific clinical manifestation for diagnosing ongoing HUS in children.
Collapse
Affiliation(s)
- Mohammadreza Parak
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Alireza Asgari
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Yazdan Hasani Nourian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Sumala S, Ekalaksananan T, Pientong C, Buddhisa S, Passorn S, Duangjit S, Janyakhantikul S, Suktus A, Bumrungthai S. The Association of HHV-6 and the TNF-α (-308G/A) Promotor with Major Depressive Disorder Patients and Healthy Controls in Thailand. Viruses 2023; 15:1898. [PMID: 37766304 PMCID: PMC10535374 DOI: 10.3390/v15091898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Major depressive disorder (MDD) is a silent global health problem that can lead to suicide. MDD development is suggested to result from numerous risk factors, including genetic factors. A precise tool for MDD diagnosis is currently not available. Recently, inflammatory processes have been identified as being strongly involved in MDD development and the reactivation of human herpesvirus type 6 (HHV-6), upregulating cytokines such as TNF-α, which are associated with MDD. Therefore, this study aimed to determine the association of HHV-6 with genetic factors, especially TNF-α mutation, in MDD patients and their relatives compared to healthy controls. The Patient Health Questionnaire (PHQ-9) was used to evaluate MDD status, and 471 oral buccal samples were investigated for HHV-6 infection and viral copy number by qPCR. TNF-α (-308G/A) gene mutation and the cytokines TNF-α, IL-6, and IL-10 were analyzed by high-resolution melting (HRM) analysis and enzyme-linked immunosorbent assay (ELISA). Whole-exome sequencing of buccal samples was performed to analyze for genetic factors. The results showed significantly higher HHV-6 positivities and viral loads in MDD patients (15/59 (25.67%) and 14,473 ± 16,948 copies/µL DNA) and their relatives (blood relatives 17/36 (47.22%) and 8146 ± 5656 copies/µL DNA); non-blood relatives 7/16 (43.75%) and 20,721 ± 12,458 copies/µL DNA) compared to the healthy population (51/360 (14.17%) and 6303 ± 5791 copies/µL DNA). The TNF-α (-308G/A) mutation showed no significant difference. Surprisingly, 12/26 (46.15%) participants with the TNF-α (-308G/A) mutation showed HHV-6 positivities at higher rates than those with wild-type TNF-α (-308G) (70/267 (26.22%)). HHV-6-positive participants with TNF-α (-308G/A) showed higher levels of TNF-α, IL-6, and IL-10 than those of negative control. Exome analysis revealed that common mutations in immune genes were associated with depression. Therefore, this study unveiled the novel association of inflammatory gene TNF-α (-308G/A) mutations with HHV-6 reactivation, which could represent a combined risk factor for MDD. This result could induce further research on MDD development and clinical applications.
Collapse
Affiliation(s)
- Sasiwimon Sumala
- Division of Biotechnology, School of Agriculture and Natural resources, University of Phayao, Phayao 56000, Thailand
| | - Tipaya Ekalaksananan
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chamsai Pientong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Surachat Buddhisa
- Department of Medical Technology, Faculty of Allied Health Sciences, Burapha University, Chonburi 20131, Thailand
| | - Supaporn Passorn
- Division of Biotechnology, School of Agriculture and Natural resources, University of Phayao, Phayao 56000, Thailand
| | - Sureewan Duangjit
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmaceutical Sciences, Ubon Ratchathani University, Ubon Ratchathani 34190, Thailand
| | - Somwang Janyakhantikul
- Division of Biopharmacy, Faculty of Pharmaceutical Sciences, Ubon Ratchathani University, Ubon Ratchathani 34190, Thailand
| | - Areeya Suktus
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sureewan Bumrungthai
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
- Division of Biopharmacy, Faculty of Pharmaceutical Sciences, Ubon Ratchathani University, Ubon Ratchathani 34190, Thailand
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| |
Collapse
|
3
|
Harding CF, Liao D, Persaud R, DeStefano RA, Page KG, Stalbow LL, Roa T, Ford JC, Goman KD, Pytte CL. Differential effects of exposure to toxic or nontoxic mold spores on brain inflammation and Morris water maze performance. Behav Brain Res 2023; 442:114294. [PMID: 36638914 PMCID: PMC10460635 DOI: 10.1016/j.bbr.2023.114294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/02/2023] [Accepted: 01/08/2023] [Indexed: 01/12/2023]
Abstract
People who live or work in moldy buildings often complain of "brain fog" that interferes with cognitive performance. Until recently, there was no published research on the effects of controlled exposure to mold stimuli on cognitive function or an obvious mechanism of action, fueling controversy over these claims. The constellation of health problems reported by mold-exposed individuals (respiratory issues, fatigue, pain, anxiety, depression, and cognitive deficits) correspond to those caused by innate immune activation following exposure to bacterial or viral stimuli. To determine if mold-induced innate immune activation might cause cognitive issues, we quantified the effects of both toxic and nontoxic mold on brain immune activation and spatial memory in the Morris water maze. We intranasally administered either 1) intact, toxic Stachybotrys chartarum spores; 2) ethanol-extracted, nontoxic Stachybotrys chartarum spores; or 3) control saline vehicle to mice. Inhalation of nontoxic spores caused significant deficits in the test of long-term memory of platform location, while not affecting short-term memory. Inhalation of toxic spores increased motivation to reach the platform. Interestingly, in both groups of mold-exposed males, numbers of interleukin-1β-immunoreactive cells in many areas of the hippocampus significantly correlated with latency to find the platform, path length, and swimming speed during training, but not during testing for long-term memory. These data add to our prior evidence that mold inhalation can interfere with cognitive processing in different ways depending on the task, and that brain inflammation is significantly correlated with changes in behavior.
Collapse
Affiliation(s)
- Cheryl F Harding
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA; Behavioral and Cognitive Neuroscience Doctoral Program, The Graduate Center, CUNY, 365 Fifth Avenue, New York, NY 10016, USA.
| | - David Liao
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA; Macaulay Honors College, CUNY, 35 West 67th Street, New York, NY 10023, USA
| | - Ramona Persaud
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA; Macaulay Honors College, CUNY, 35 West 67th Street, New York, NY 10023, USA; Chemistry Department, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA
| | - Richard A DeStefano
- Macaulay Honors College, CUNY, 35 West 67th Street, New York, NY 10023, USA; Chemistry Department, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA
| | - Kimberly G Page
- Behavioral and Cognitive Neuroscience Doctoral Program, The Graduate Center, CUNY, 365 Fifth Avenue, New York, NY 10016, USA
| | - Lauren L Stalbow
- Macaulay Honors College, CUNY, 35 West 67th Street, New York, NY 10023, USA; Psychology Department, Queens College, CUNY, 65-30 Kissena Boulevard, Flushing 11367, NY, USA
| | - Tina Roa
- Biological Sciences, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA
| | - Jordan C Ford
- Biological Sciences, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA
| | - Ksenia D Goman
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA
| | - Carolyn L Pytte
- Behavioral and Cognitive Neuroscience Doctoral Program, The Graduate Center, CUNY, 365 Fifth Avenue, New York, NY 10016, USA; Psychology Department, Queens College, CUNY, 65-30 Kissena Boulevard, Flushing 11367, NY, USA
| |
Collapse
|
4
|
Ostojic SM. Diagnostic and Pharmacological Potency of Creatine in Post-Viral Fatigue Syndrome. Nutrients 2021; 13:nu13020503. [PMID: 33557013 PMCID: PMC7913646 DOI: 10.3390/nu13020503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
Post-viral fatigue syndrome (PVFS) is a widespread chronic neurological disease with no definite etiological factor(s), no actual diagnostic test, and no approved pharmacological treatment, therapy, or cure. Among other features, PVFS could be accompanied by various irregularities in creatine metabolism, perturbing either tissue levels of creatine in the brain, the rates of phosphocreatine resynthesis in the skeletal muscle, or the concentrations of the enzyme creatine kinase in the blood. Furthermore, supplemental creatine and related guanidino compounds appear to impact both patient- and clinician-reported outcomes in syndromes and maladies with chronic fatigue. This paper critically overviews the most common disturbances in creatine metabolism in various PVFS populations, summarizes human trials on dietary creatine and creatine analogs in the syndrome, and discusses new frontiers and open questions for using creatine in a post-COVID-19 world.
Collapse
Affiliation(s)
- Sergej M. Ostojic
- FSPE Applied Bioenergetics Lab, University of Novi Sad, 21000 Novi Sad, Serbia;
- Faculty of Health Sciences, University of Pecs, H-7621 Pecs, Hungary
| |
Collapse
|
5
|
Mold inhalation causes innate immune activation, neural, cognitive and emotional dysfunction. Brain Behav Immun 2020; 87:218-228. [PMID: 31751617 PMCID: PMC7231651 DOI: 10.1016/j.bbi.2019.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 11/07/2019] [Accepted: 11/17/2019] [Indexed: 01/01/2023] Open
Abstract
Individuals living or working in moldy buildings complain of a variety of health problems including pain, fatigue, increased anxiety, depression, and cognitive deficits. The ability of mold to cause such symptoms is controversial since no published research has examined the effects of controlled mold exposure on brain function or proposed a plausible mechanism of action. Patient symptoms following mold exposure are indistinguishable from those caused by innate immune activation following bacterial or viral exposure. We tested the hypothesis that repeated, quantified doses of both toxic and nontoxic mold stimuli would cause innate immune activation with concomitant neural effects and cognitive, emotional, and behavioral symptoms. We intranasally administered either 1) intact, toxic Stachybotrys spores; 2) extracted, nontoxic Stachybotrys spores; or 3) saline vehicle to mice. As predicted, intact spores increased interleukin-1β immunoreactivity in the hippocampus. Both spore types decreased neurogenesis and caused striking contextual memory deficits in young mice, while decreasing pain thresholds and enhancing auditory-cued memory in older mice. Nontoxic spores also increased anxiety-like behavior. Levels of hippocampal immune activation correlated with decreased neurogenesis, contextual memory deficits, and/or enhanced auditory-cued fear memory. Innate-immune activation may explain how both toxic mold and nontoxic mold skeletal elements caused cognitive and emotional dysfunction.
Collapse
|
6
|
Bansal RA, Tadros S, Bansal AS. The presence of overlapping quality of life symptoms in primary antibody deficiency (PAD) and chronic fatigue syndrome (CFS). Allergy Asthma Clin Immunol 2020; 16:21. [PMID: 32256619 PMCID: PMC7106611 DOI: 10.1186/s13223-020-0417-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 03/04/2020] [Indexed: 01/26/2023] Open
Abstract
Background Fatigue, sleep disturbance and altered mood are frequently reported in patients with primary antibody deficiency syndrome (PADS) on adequate immunoglobulin replacement therapy. This study aimed to determine the frequency of symptoms compatible with chronic fatigue syndrome (CFS) in patients with PADS. Methods The study involved the distribution of 682 self-completed postal questionnaires to ascertain the presence and frequency of symptoms compatible with CFS in patients with PADS. The reporting of symptoms for each patient were scored against the CFS diagnostic criteria used within our own South London Chronic Fatigue service. Results The frequency of symptoms compatible with CFS were evident in 26 of the 188 patients (16.25%) returning adequately completed questionnaires. We considered a bias in the return of questionnaires amongst PADS patients with fatigue to be likely. As such we estimated the minimum frequency of CFS in patients with PADS to be 4% based on the 682 PAD patients to whom the questionnaire was distributed. This was significantly higher than the 0.5% estimate of the prevalence of CFS in the community in western populations. While the presence of significant fatigue correlated with the presence of anxiety and depression, there was no association with self-reported lung damage. Sleep disturbance affected 60% of the PAD patients returning satisfactory questionnaires and as expected the CFS score was higher in those with greater physical limitation. Conclusions We conclude that patients with PADS have a high frequency of fatigue, low mood and anxiety. We suggest routine questioning for the symptoms of fatigue, disturbed sleep and altered mood in patients with PADS. The use of several treatment strategies in CFS may prove beneficial in improving the quality of life of patients with PAD.
Collapse
Affiliation(s)
- Rhea A Bansal
- Frimley Health NHS Trust, Portsmouth Road, Camberley, Surrey GU16 7UJ UK
| | - Susan Tadros
- 2The Royal Free Hospital, Pond Street, Hampstead, London, NW3 2QG UK
| | | |
Collapse
|
7
|
Shoenfeld Y, Ryabkova VA, Scheibenbogen C, Brinth L, Martinez-Lavin M, Ikeda S, Heidecke H, Watad A, Bragazzi NL, Chapman J, Churilov LP, Amital H. Complex syndromes of chronic pain, fatigue and cognitive impairment linked to autoimmune dysautonomia and small fiber neuropathy. Clin Immunol 2020; 214:108384. [PMID: 32171889 DOI: 10.1016/j.clim.2020.108384] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/21/2022]
Abstract
Chronic fatigue syndrome, postural orthostatic tachycardia syndrome, complex regional pain syndrome and silicone implant incompatibility syndrome are a subject of debate among clinicians and researchers. Both the pathogenesis and treatment of these disorders require further study. In this paper we summarize the evidence regarding the role of autoimmunity in these four syndromes with respect to immunogenetics, autoimmune co-morbidities, alteration in immune cell subsets, production of autoantibodies and presentation in animal models. These syndromes could be incorporated in a new concept of autoimmune neurosensory dysautonomia with the common denominators of autoantibodies against G-protein coupled receptors and small fiber neuropathy. Sjogren's syndrome, which is a classical autoimmune disease, could serve as a disease model, illustrating the concept. Development of this concept aims to identify an apparently autoimmune subgroup of the disputable disorders, addressed in the review, which may most benefit from the immunotherapy.
Collapse
Affiliation(s)
- Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Aviv, Israel; Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Russia.
| | - Varvara A Ryabkova
- Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Russia
| | - Carmen Scheibenbogen
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Louise Brinth
- Department of Nuclear Medicine, Herlev Gentofte Hospital, Gentofte Hospitalsvej 1, 2900 Hellerup, Denmark
| | - Manuel Martinez-Lavin
- Rheumatology Department, National Institute of Cardiology, Juan Badiano 1, 14080 Mexico City, Mexico
| | - Shuichi Ikeda
- Intractable Disease Care Center, Shinshu University Hospital, Matsumoto 390-0802, Japan
| | | | - Abdulla Watad
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Aviv, Israel; Section of Musculoskeletal Disease, Leeds Institute of Molecular Medicine, University of Leeds, NIHR Leeds Musculoskeletal Biomedical Research Unit, Chapel Allerton Hospital, Leeds, UK
| | - Nicola L Bragazzi
- Postgraduate School of Public Health, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Joab Chapman
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Aviv, Israel
| | - Leonid P Churilov
- Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Russia
| | - Howard Amital
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Aviv, Israel
| |
Collapse
|
8
|
de Groot MHM, Castorena CM, Cox KH, Kumar V, Mohawk JA, Ahmed NI, Takahashi JS. A novel mutation in Slc2a4 as a mouse model of fatigue. GENES BRAIN AND BEHAVIOR 2019; 18:e12578. [PMID: 31059591 DOI: 10.1111/gbb.12578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 11/28/2022]
Abstract
Chronic fatigue is a debilitating disorder with widespread consequences, but effective treatment strategies are lacking. Novel genetic mouse models of fatigue may prove invaluable for studying its underlying physiological mechanisms and for testing treatments and interventions. In a screen of voluntary wheel-running behavior in N-ethyl-N-nitrosourea mutagenized C57BL/6J mice, we discovered two lines with low body weights and aberrant wheel-running patterns suggestive of a fatigue phenotype. Affected progeny from these lines had lower daily activity levels and exhibited low amplitude circadian rhythm alterations. Their aberrant behavior was characterized by frequent interruptions and periods of inactivity throughout the dark phase of the light-dark cycle and increased levels of activity during the rest or light phase. Expression of the behavioral phenotypes in offspring of strategic crosses was consistent with a recessive inheritance pattern. Mapping of phenotypic abnormalities showed linkage with a single locus on chromosome 1, and whole exome sequencing identified a single point mutation in the Slc2a4 gene encoding the GLUT4 insulin-responsive glucose transporter. The single nucleotide change (A-T, which we named "twiggy") was in the distal end of exon 10 and resulted in a premature stop (Y440*). Additional metabolic phenotyping confirmed that these mice recapitulate phenotypes found in GLUT4 knockout mice. However, to the best of our knowledge, this is the first time a mutation in this gene has been shown to result in extensive changes in general behavioral patterns. These findings suggest that GLUT4 may be involved in circadian behavioral abnormalities and could provide insights into fatigue in humans.
Collapse
Affiliation(s)
- Marleen H M de Groot
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Carlos M Castorena
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kimberly H Cox
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Vivek Kumar
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jennifer A Mohawk
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Newaz I Ahmed
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Joseph S Takahashi
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
9
|
Myalgia and chronic fatigue syndrome following immunization: macrophagic myofasciitis and animal studies support linkage to aluminum adjuvant persistency and diffusion in the immune system. Autoimmun Rev 2019; 18:691-705. [PMID: 31059838 DOI: 10.1016/j.autrev.2019.05.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 01/31/2019] [Indexed: 01/04/2023]
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a multifactorial and poorly undersood disabling disease. We present epidemiological, clinical and experimental evidence that ME/CFS constitutes a major type of adverse effect of vaccines, especially those containing poorly degradable particulate aluminum adjuvants. Evidence has emerged very slowly due to the multiplicity, lack of specificity, delayed onset, and frequent medical underestimation of ME/CFS symptoms. It was supported by an epidemiological study comparing vaccinated vs unvaccinated militaries that remained undeployed during Gulf War II. Affected patients suffer from cognitive dysfunction affecting attention, memory and inter-hemispheric connexions, well correlated to brain perfusion defects and associated with a stereotyped and distinctive pattern of cerebral glucose hypometabolism. Deltoid muscle biopsy performed to investigate myalgia typically yields macrophagic myofasciitis (MMF), a histological biomarker assessing longstanding persistency of aluminum agglomerates within innate immune cells at site of previous immunization. MMF is seemingly linked to altered mineral particle detoxification by the xeno/autophagy machinery. Comparing toxicology of different forms of aluminum and different types of exposure is misleading and inadequate and small animal experiments have turned old dogma upside down. Instead of being rapidly solubilized in the extracellular space, injected aluminum particles are quickly captured by immune cells and transported to distant organs and the brain where they elicit an inflammatory response and exert selective low dose long-term neurotoxicity. Clinical observations and experiments in sheep, a large animal like humans, confirmed both systemic diffusion and neurotoxic effects of aluminum adjuvants. Post-immunization ME/CFS represents the core manifestation of "autoimmune/inflammatory syndrome induced by adjuvants" (ASIA).
Collapse
|
10
|
Cliff JM, King EC, Lee JS, Sepúlveda N, Wolf AS, Kingdon C, Bowman E, Dockrell HM, Nacul L, Lacerda E, Riley EM. Cellular Immune Function in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). Front Immunol 2019; 10:796. [PMID: 31057538 PMCID: PMC6477089 DOI: 10.3389/fimmu.2019.00796] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/26/2019] [Indexed: 12/16/2022] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating condition with unknown aetiology, Myalgic encephalomyelitis unclear pathophysiology and with no diagnostic test or biomarker available. Many patients report their ME/CFS began after an acute infection, and subsequent increased frequency of infections, such as colds or influenza, is common. These factors imply an altered immunological status exists in ME/CFS, in at least a proportion of patients, yet previous studies of peripheral immunity have been discrepant and inconclusive. The UK ME/CFS Biobank, which has collected blood samples from nearly 300 clinically-confirmed ME/CFS patients, enables large-scale studies of immunological function in phenotypically well-characterised participants. In this study, herpes virus serological status and T cell, B cell, NK cell and monocyte populations were investigated in 251 ME/CFS patients, including 54 who were severely affected, and compared with those from 107 healthy participants and with 46 patients with Multiple Sclerosis. There were no differences in seroprevalence for six human herpes viruses between ME/CFS and healthy controls, although seroprevalence for the Epstein-Barr virus was higher in multiple sclerosis patients. Contrary to previous reports, no significant differences were observed in NK cell numbers, subtype proportions or in vitro responsiveness between ME/CFS patients and healthy control participants. In contrast, the T cell compartment was altered in ME/CFS, with increased proportions of effector memory CD8+ T cells and decreased proportions of terminally differentiated effector CD8+ T cells. Conversely, there was a significantly increased proportion of mucosal associated invariant T cells (MAIT) cells, especially in severely affected ME/CFS patients. These abnormalities demonstrate that an altered immunological state does exist in ME/CFS, particularly in severely affected people. This may simply reflect ongoing or recent infection, or may indicate future increased susceptibility to infection. Longitudinal studies of ME/CFS patients are needed to help to determine cause and effect and thus any potential benefits of immuno-modulatory treatments for ME/CFS.
Collapse
Affiliation(s)
- Jacqueline M. Cliff
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Elizabeth C. King
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Ji-Sook Lee
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Nuno Sepúlveda
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Centre of Statistics and Applications, University of Lisbon, Lisbon, Portugal
| | - Asia-Sophia Wolf
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Caroline Kingdon
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Erinna Bowman
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Hazel M. Dockrell
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Luis Nacul
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Eliana Lacerda
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Eleanor M. Riley
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
11
|
Morris G, Maes M, Berk M, Puri BK. Myalgic encephalomyelitis or chronic fatigue syndrome: how could the illness develop? Metab Brain Dis 2019; 34:385-415. [PMID: 30758706 PMCID: PMC6428797 DOI: 10.1007/s11011-019-0388-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 01/23/2019] [Indexed: 12/19/2022]
Abstract
A model of the development and progression of chronic fatigue syndrome (myalgic encephalomyelitis), the aetiology of which is currently unknown, is put forward, starting with a consideration of the post-infection role of damage-associated molecular patterns and the development of chronic inflammatory, oxidative and nitrosative stress in genetically predisposed individuals. The consequences are detailed, including the role of increased intestinal permeability and the translocation of commensal antigens into the circulation, and the development of dysautonomia, neuroinflammation, and neurocognitive and neuroimaging abnormalities. Increasing levels of such stress and the switch to immune and metabolic downregulation are detailed next in relation to the advent of hypernitrosylation, impaired mitochondrial performance, immune suppression, cellular hibernation, endotoxin tolerance and sirtuin 1 activation. The role of chronic stress and the development of endotoxin tolerance via indoleamine 2,3-dioxygenase upregulation and the characteristics of neutrophils, monocytes, macrophages and T cells, including regulatory T cells, in endotoxin tolerance are detailed next. Finally, it is shown how the immune and metabolic abnormalities of chronic fatigue syndrome can be explained by endotoxin tolerance, thus completing the model.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
- Department of Psychiatry, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Victoria, Australia
| | - Basant K Puri
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, England, W12 0HS, UK.
| |
Collapse
|
12
|
Richman S, Morris MC, Broderick G, Craddock TJA, Klimas NG, Fletcher MA. Pharmaceutical Interventions in Chronic Fatigue Syndrome: A Literature-based Commentary. Clin Ther 2019; 41:798-805. [PMID: 30871727 DOI: 10.1016/j.clinthera.2019.02.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 02/11/2019] [Indexed: 12/22/2022]
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating disorder characterized by prolonged periods of fatigue, chronic pain, depression, and a complex constellation of other symptoms. Currently, ME/CFS has no known cause, nor are the mechanisms of illness well understood. Therefore, with few exceptions, attempts to treat ME/CFS have been directed mainly toward symptom management. These treatments include antivirals, pain relievers, antidepressants, and oncologic agents as well as other single-intervention treatments. Results of these trials have been largely inconclusive and, in some cases, contradictory. Contributing factors include a lack of well-designed and -executed studies and the highly heterogeneous nature of ME/CFS, which has made a single etiology difficult to define. Because the majority of single-intervention treatments have shown little efficacy, it may instead be beneficial to explore broader-acting combination therapies in which a more focused precision-medicine approach is supported by a systems-level analysis of endocrine and immune co-regulation.
Collapse
Affiliation(s)
- Spencer Richman
- Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, USA; Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY, USA
| | - Matthew C Morris
- Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, USA; Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY, USA
| | - Gordon Broderick
- Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, USA; Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY, USA; Institute for Neuro Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA; Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA.
| | - Travis J A Craddock
- Institute for Neuro Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA; Departments of Psychology and Neuroscience, Computer Science, Nova Southeastern University, Fort Lauderdale, FL, USA; Department of Clinical Immunology, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Nancy G Klimas
- Institute for Neuro Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA; Department of Clinical Immunology, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Mary Ann Fletcher
- Institute for Neuro Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA; Department of Clinical Immunology, Nova Southeastern University, Fort Lauderdale, FL, USA
| |
Collapse
|
13
|
Dampness and mold hypersensitivity syndrome and vaccination as risk factors for chronic fatigue syndrome. Autoimmun Rev 2018; 18:107-108. [PMID: 30408578 DOI: 10.1016/j.autrev.2018.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 01/28/2023]
|
14
|
Valtonen V. Clinical Diagnosis of the Dampness and Mold Hypersensitivity Syndrome: Review of the Literature and Suggested Diagnostic Criteria. Front Immunol 2017; 8:951. [PMID: 28848553 PMCID: PMC5554125 DOI: 10.3389/fimmu.2017.00951] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 07/25/2017] [Indexed: 11/13/2022] Open
Abstract
A great variety of non-specific symptoms may occur in patients living or working in moisture-damaged buildings. In the beginning, these symptoms are usually reversible, mild, and present irritation of mucosa and increased morbidity due to respiratory tract infections and asthma-like symptoms. Later, the disease may become chronic and a patient is referred to a doctor where the assessment of dampness and mold hypersensitivity syndrome (DMHS) often presents diagnostic challenges. Currently, unanimously accepted laboratory tests are not yet available. Therefore, the diagnosis of DMHS is clinical and is based on the patient’s history and careful examination. In this publication, I reviewed contemporary knowledge on clinical presentations, laboratory methods, and clinical assessment of DMHS. From the literature, I have not found any proposed diagnostic clinical criteria. Therefore, I propose five clinical criteria to diagnose DMHS: (1) the history of mold exposure in water-damaged buildings, (2) increased morbidity to due infections, (3) sick building syndrome, (4) multiple chemical sensitivity, and (5) enhanced scent sensitivity. If all the five criteria are met, the patient has a very probable DMHS. To resolve the current problems in assigning correct DMHS diagnosis, we also need novel assays to estimate potential risks of developing DMHS.
Collapse
Affiliation(s)
- Ville Valtonen
- Division of Infectious Diseases, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
15
|
Somppi TL. Non-Thyroidal Illness Syndrome in Patients Exposed to Indoor Air Dampness Microbiota Treated Successfully with Triiodothyronine. Front Immunol 2017; 8:919. [PMID: 28824644 PMCID: PMC5545575 DOI: 10.3389/fimmu.2017.00919] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 07/20/2017] [Indexed: 01/21/2023] Open
Abstract
Long-term exposure to dampness microbiota induces multi-organ morbidity. One of the symptoms related to this disorder is non-thyroidal illness syndrome (NTIS). A retrospective study was carried out in nine patients with a history of mold exposure, experiencing chronic fatigue, cognitive disorder, and different kinds of hypothyroid symptoms despite provision of levothyroxine (3,5,3',5'-tetraiodothyronine, LT4) monotherapy. Exposure to volatile organic compounds present in water-damaged buildings including metabolic products of toxigenic fungi and mold-derived inflammatory agents can lead to a deficiency or imbalance of many hormones, such as active T3 hormone. Since the 1970s, the synthetic prohormone, levothyroxine (LT4), has been the most commonly prescribed thyroid hormone in replacement monotherapy. It has been presumed that the peripheral conversion of T4 (3,5,3',5'-tetraiodothyronine) into T3 (3,5,3'-triiodothyronine) is sufficient to satisfy the overall tissue requirements. However, evidence is presented that this not the case for all patients, especially those exposed to indoor air molds. This retrospective study describes the successful treatment of nine patients in whom NTIS was treated with T3-based thyroid hormone. The treatment was based on careful interview, clinical monitoring, and laboratory analysis of serum free T3 (FT3), reverse T3 (rT3) and thyroid-stimulating hormone, free T4, cortisol, and dehydroepiandrosterone (DHEA) values. The ratio of FT3/rT3 was calculated. In addition, some patients received adrenal support with hydrocortisone and DHEA. All patients received nutritional supplementation and dietary instructions. During the therapy, all nine patients reported improvements in all of the symptom groups. Those who had residual symptoms during T3-based therapy remained exposed to indoor air molds in their work places. Four patients were unable to work and had been on disability leave for a long time during LT4 monotherapy. However, during the T3-based and supportive therapy, all patients returned to work in so-called "healthy" buildings. The importance of avoiding mycotoxin exposure via the diet is underlined as DIO2 genetic polymorphism and dysfunction of DIO2 play an important role in the development of symptoms that can be treated successfully with T3 therapy.
Collapse
|
16
|
Nguyen CB, Alsøe L, Lindvall JM, Sulheim D, Fagermoen E, Winger A, Kaarbø M, Nilsen H, Wyller VB. Whole blood gene expression in adolescent chronic fatigue syndrome: an exploratory cross-sectional study suggesting altered B cell differentiation and survival. J Transl Med 2017; 15:102. [PMID: 28494812 PMCID: PMC5426002 DOI: 10.1186/s12967-017-1201-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Chronic fatigue syndrome (CFS) is a prevalent and disabling condition affecting adolescents. The pathophysiology is poorly understood, but immune alterations might be an important component. This study compared whole blood gene expression in adolescent CFS patients and healthy controls, and explored associations between gene expression and neuroendocrine markers, immune markers and clinical markers within the CFS group. METHODS CFS patients (12-18 years old) were recruited nation-wide to a single referral center as part of the NorCAPITAL project. A broad case definition of CFS was applied, requiring 3 months of unexplained, disabling chronic/relapsing fatigue of new onset, whereas no accompanying symptoms were necessary. Healthy controls having comparable distribution of gender and age were recruited from local schools. Whole blood samples were subjected to RNA sequencing. Immune markers were blood leukocyte counts, plasma cytokines, serum C-reactive protein and immunoglobulins. Neuroendocrine markers encompassed plasma and urine levels of catecholamines and cortisol, as well as heart rate variability indices. Clinical markers consisted of questionnaire scores for symptoms of post-exertional malaise, inflammation, fatigue, depression and trait anxiety, as well as activity recordings. RESULTS A total of 29 CFS patients and 18 healthy controls were included. We identified 176 genes as differentially expressed in patients compared to controls, adjusting for age and gender factors. Gene set enrichment analyses suggested impairment of B cell differentiation and survival, as well as enhancement of innate antiviral responses and inflammation in the CFS group. A pattern of co-expression could be identified, and this pattern, as well as single gene transcripts, was significantly associated with indices of autonomic nervous activity, plasma cortisol, and blood monocyte and eosinophil counts. Also, an association with symptoms of post-exertional malaise was demonstrated. CONCLUSION Adolescent CFS is characterized by differential gene expression pattern in whole blood suggestive of impaired B cell differentiation and survival, and enhanced innate antiviral responses and inflammation. This expression pattern is associated with neuroendocrine markers of altered HPA axis and autonomic nervous activity, and with symptoms of post-exertional malaise. Trial registration Clinical Trials NCT01040429.
Collapse
Affiliation(s)
- Chinh Bkrong Nguyen
- Department of Paediatrics and Adolescent Health, Akershus University Hospital, 1478 Lørenskog, Norway
- Division of Medicine and Laboratory Sciences, Medical Faculty, University of Oslo, Oslo, Norway
| | - Lene Alsøe
- Institute of Clinical Medicine, Department of Clinical Molecular Biology, University of Oslo, and Akershus University Hospital, Lørenskog, Norway
| | - Jessica M. Lindvall
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Dag Sulheim
- Department of Paediatrics, Lillehammer County Hospital, Lillehammer, Norway
| | - Even Fagermoen
- Department of Anesthesiology and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Anette Winger
- Institute of Nursing Sciences, Oslo and Akershus University College of Applied Sciences, Oslo, Norway
| | - Mari Kaarbø
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Hilde Nilsen
- Institute of Clinical Medicine, Department of Clinical Molecular Biology, University of Oslo, and Akershus University Hospital, Lørenskog, Norway
| | - Vegard Bruun Wyller
- Department of Paediatrics and Adolescent Health, Akershus University Hospital, 1478 Lørenskog, Norway
- Division of Medicine and Laboratory Sciences, Medical Faculty, University of Oslo, Oslo, Norway
| |
Collapse
|
17
|
Eriksen W. The spread of EBV to ectopic lymphoid aggregates may be the final common pathway in the pathogenesis of ME/CFS. Med Hypotheses 2017; 102:8-15. [PMID: 28478837 DOI: 10.1016/j.mehy.2017.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/02/2017] [Accepted: 02/26/2017] [Indexed: 12/22/2022]
Abstract
According to the hypothesis presented here, myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) develops over 3 steps: Step 1 is characterized by the aggregation of lymphoid cells in dorsal root ganglia or other nervous structures. The cause of this formation of ectopic lymphoid aggregates may be an acute infection, asymptomatic reactivations of a common neurotropic virus, exposure to a neurotoxin, or physical injury to peripheral nerves. In step 2, Epstein-Barr virus (EBV)-infected lymphocytes or monocytes bring EBV from the circulation to one or several of these lymphoid aggregates, whereupon cell-to-cell transmission of EBV and proliferation of latently EBV-infected lymphocytes lead to the presence of many EBV-infected cells in the lymphoid aggregates. The EBV-infected cells in the aggregates ignite an inflammation in the surrounding nervous tissue. This local inflammation elicits, in turn, a wave of glial cell activation that spreads from the EBV-infected area to parts of the nervous system that are not EBV-infected, disturbing the neuron-glial interaction in both the peripheral - and central nervous system. In step 3, immune cell exhaustion contributes to a consolidation of the pathological processes. There might be a cure: Infusions of autologous EBV-specific T-lymphocytes can perhaps remove the EBV-infected cells from the nervous system.
Collapse
Affiliation(s)
- Willy Eriksen
- Domain for Mental and Physical Health, Norwegian Institute of Public Health, Box 4404 Nydalen, 0403 Oslo, Norway.
| |
Collapse
|
18
|
Moos WH, Pinkert CA, Irwin MH, Faller DV, Kodukula K, Glavas IP, Steliou K. Epigenetic Treatment of Persistent Viral Infections. Drug Dev Res 2016; 78:24-36. [PMID: 27761936 DOI: 10.1002/ddr.21366] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Preclinical Research Approximately 2,500 years ago, Hippocrates used the word herpes as a medical term to describe lesions that appeared to creep or crawl on the skin, advocating heat as a possible treatment. During the last 50 years, pharmaceutical research has made great strides, and therapeutic options have expanded to include small molecule antiviral agents, protease inhibitors, preventive vaccines for a handful of the papillomaviruses, and even cures for hepatitis C virus infections. However, effective treatments for persistent and recurrent viral infections, particularly the highly prevalent herpesviruses, continue to represent a significant unmet medical need, affecting the majority of the world's population. Exploring the population diversity of the human microbiome and the effects its compositional variances have on the immune system, health, and disease are the subjects of intense investigational research and study. Among the collection of viruses, bacteria, fungi, and single-cell eukaryotes that comprise the human microbiome, the virome has been grossly understudied relative to the influence it exerts on human pathophysiology, much as mitochondria have until recently failed to receive the attention they deserve, given their critical biomedical importance. Fortunately, cellular epigenetic machinery offers a wealth of druggable targets for therapeutic intervention in numerous disease indications, including those outlined above. With advances in synthetic biology, engineering our body's commensal microorganisms to seek out and destroy pathogenic species is clearly on the horizon. This is especially the case given recent breakthroughs in genetic manipulation with tools such as the CRISPR/Cas (clustered regularly interspaced short palindromic repeats/CRISPR-associated) gene-editing platforms. Tying these concepts together with our previous work on the microbiome and neurodegenerative and neuropsychiatric diseases, we suggest that, because mammalian cells respond to a viral infection by triggering a cascade of antiviral innate immune responses governed substantially by the cell's mitochondria, small molecule carnitinoids represent a new class of therapeutics with potential widespread utility against many infectious insults. Drug Dev Res 78 : 24-36, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Walter H Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, California
| | - Carl A Pinkert
- Department of Biological Sciences, College of Arts and Sciences, The University of Alabama, Tuscaloosa, Alabama
| | - Michael H Irwin
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Douglas V Faller
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts.,Boston University School of Medicine, Cancer Research Center, Boston, Massachusetts
| | | | - Ioannis P Glavas
- Department of Ophthalmology, New York University School of Medicine, New York
| | - Kosta Steliou
- Boston University School of Medicine, Cancer Research Center, Boston, Massachusetts.,PhenoMatriX, Boston, Massachusetts
| |
Collapse
|