64
Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=irab20 Download by: [UNIVERSITY OF ADELAIDE LIBRARIES] Date: 04 January 2018, At: 07:03 International Journal of Radiation Biology ISSN: 0955-3002 (Print) 1362-3095 (Online) Journal homepage: http://www.tandfonline.com/loi/irab20 Chronic Fatigue and Immune Deficiency Syndrome (CFIDS), cellular metabolism, and ionizing radiation: A review of contemporary scientific literature and suggested directions for future research Andrej Rusin, Colin Seymour & Carmel Mothersill To cite this article: Andrej Rusin, Colin Seymour & Carmel Mothersill (2018): Chronic Fatigue and Immune Deficiency Syndrome (CFIDS), cellular metabolism, and ionizing radiation: A review of contemporary scientific literature and suggested directions for future research, International Journal of Radiation Biology, DOI: 10.1080/09553002.2018.1422871 To link to this article: https://doi.org/10.1080/09553002.2018.1422871 View supplementary material Accepted author version posted online: 03 Jan 2018. Submit your article to this journal View related articles View Crossmark data

Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

  • Upload
    others

  • View
    9

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Full Terms & Conditions of access and use can be found athttp://www.tandfonline.com/action/journalInformation?journalCode=irab20

Download by: [UNIVERSITY OF ADELAIDE LIBRARIES] Date: 04 January 2018, At: 07:03

International Journal of Radiation Biology

ISSN: 0955-3002 (Print) 1362-3095 (Online) Journal homepage: http://www.tandfonline.com/loi/irab20

Chronic Fatigue and Immune Deficiency Syndrome(CFIDS), cellular metabolism, and ionizingradiation: A review of contemporary scientificliterature and suggested directions for futureresearch

Andrej Rusin, Colin Seymour & Carmel Mothersill

To cite this article: Andrej Rusin, Colin Seymour & Carmel Mothersill (2018): Chronic Fatigue andImmune Deficiency Syndrome (CFIDS), cellular metabolism, and ionizing radiation: A review ofcontemporary scientific literature and suggested directions for future research, International Journalof Radiation Biology, DOI: 10.1080/09553002.2018.1422871

To link to this article: https://doi.org/10.1080/09553002.2018.1422871

View supplementary material

Accepted author version posted online: 03Jan 2018.

Submit your article to this journal

View related articles

View Crossmark data

Page 2: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Short Title: Ionizing radiation effects on CFIDS, cell metabolism

REVIEW

Chronic Fatigue and Immune Deficiency Syndrome (CFIDS), cellular metabolism, and ionizing

radiation: A review of contemporary scientific literature and suggested directions for future

research

Andrej Rusin

McMaster University, Biology, Hamilton, Ontario, Canada

Colin Seymour

McMaster University, Medical Physics and Applied Radiation Sciences, Hamilton, Ontario,

Canada

Carmel Mothersill

McMaster University, Biology, 1280 Main street West, Hamilton, Ontario, Canada

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 3: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

CONTACT Andrej Rusin Email: [email protected] McMaster University, Biology, 1280 Main

Street West, Hamilton, Ontario, L8S 4L8 Canada

Supplemental Material for this article can be accessed on the publisher’s website.

Abstract

Purpose: To investigate biochemical pathways known to be involved in radiation response and

in CFIDS to determine if there might be common underlying mechanisms leading to symptoms

experienced by those accidentally or deliberately exposed to radiation and those suffering from

CFIDS. If such a link were established, to suggest testable hypotheses to investigate the

mechanisms with the aim of identifying new therapeutic targets.

Conclusions: Evidence for involvement of the alpha-synuclein, cytochrome c oxidiase, αB-

crystallin, RNase L, and lactate dehydrogenase/STAT1 pathways is strong and suggests a

common underlying mechanism involving mitochondrial dysfunction mediated by ROS and

disruption of ATP production. The downstream effect of this is compromised energy

production. Testable hypotheses are suggested to investigate the involvement of these

pathways further.

Key Words: Chronic Fatigue Syndrome, Bystander effects of radiation, Reactive oxygen species

(ROS), Post-Radiation Syndrome, Atomic Veterans

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 4: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Introduction

Atomic Veterans and Chronic Fatigue

Victims of radiation accidents such as Chernobyl or Fukushima, and those exposed deliberately

in the Atomic Bombs, the Cold War nuclear detonations, and the more recent Gulf War

complain of diffuse symptoms many years later (Loganovsky 2000; Milano 2000; Durakoviæ

2003; Bertell 2006; Broinowski 2013). Generally, their contention that ionizing radiation

exposure led to their symptoms manifesting up to 60 years later are dismissed as non-causal

due to the allegedly low doses they received and the lack of sufficient numbers to establish

good epidemiology (McCauley et al. 2002; Coughlin et al. 2013; Mothersill and Seymour 2016).

Many of the symptoms and disease manifestations are similar to those seen in patients

diagnosed with Chronic Fatigue and Immune Deficiency Syndrome (CFIDS).

The onset of symptoms following acute radiation exposure has been termed Post-Radiation

Syndrome (PRS), an illness studied mainly in atomic bomb and Chernobyl survivors. A few

studies have noted that the pathology of PRS is very similar to a family of multisystem illnesses

including CFIDS, fibromyalgia, and multiple chemical sensitivity, according to Pall (2008). This

connection was mainly made due to a similar onset of the disease after a short-term stressor,

the same chemical markers appearing in both illnesses, and similar symptoms (Loganovsky

2000; Pall and Satterlee 2001; Pall 2007, 2008, 2009). According to the review published by Pall

(2008), while the pathways thought to be involved in PRS were mostly studied in patients

exposed to very high doses of radiation, other studies have demonstrated that much lower

doses can result in activation of the same pathways (Mohan and Meltz 1994; Prasad et al. 1994;

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 5: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Yin et al. 2003; Mitra et al. 2005; Rithidech et al. 2005). These include doses within the range

experienced by the Chernobyl Liquidators (Kumerova et al. 2000). The same study discussing

Chernobyl Liquidators indicated that the workers experienced symptoms consistent with CFIDS,

although it did not mention the disease by name. There have been many studies that have

linked low-dose radiation exposure to CFIDS directly, primarily due to neurophysiologic and

metabolic changes that occur in some patients; studies include those conducted on Chernobyl

survivors and Gulf War veterans. Among these are studies showing effects in patients receiving

total biological doses of 0-70 mSv (Pflugbeil et al. 2006; Pall 2008; Loganovsky 2009; Bazyka et

al. 2010; Loganovsky et al. 2016).

While no clinical study has directly linked CFIDS with radiation exposure, some symptoms

documented in victims of radiation accidents and in radiotherapy patients were shown to be

consistent with symptoms of CFIDS, including fatigue, immune system dysfunction, and

cognitive dysfunction (Pastel et al. 2002; Pflugbel et al. 2006; Pall 2008; Loganovsky 2016;

Mothersill and Seymour 2016). Such symptoms are often dismissed as inconsequent to

radiation exposure or as radiophobia mainly on the grounds that they do not appear to be dose

dependent and are reported by people who have been exposed to doses considered too low to

cause biological effects (Pastel 2002; Mothersill and Seymour 2016). What these authors are

ignoring is that non-targeted effects of radiation (NTE) have an exceedingly low threshold for

induction in the region of 2-5mGy (Liu Z et al. 2007; Schettino et al. 2005) and once turned on

they continue to be expressed over time, including over generations (Nagasawa and Little 1992;

Mothersill and Seymour 1997; Wright 1998; Limoli et al. 2000; Mothersill et al. 2000; Morgan

2003; Smith et al. 2007; Lorimore et al. 2008; reviewed in Nugent et al. 2010; Shi et al. 2016;

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 6: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Smith et al. 2016). The second point about NTE is that the effects saturate at relatively low

doses, being fully expressed after a 0.5Gy acute dose of gamma radiation (Seymour and

Mothersill 2000; Liu Z et al 2007). Therefore, it is not surprising that there is no correlation of

these symptoms with dose; this idea has been discussed and reviewed in Mothersill and

Seymour 2016 cited above.

Chronic Fatigue and Immune Deficiency Syndrome

CFIDS is a disease without a definitive cause or mechanism. CFIDS patients claim that low dose

exposure to ionizing radiation may be a common factor involved in their disease (Loganovsky

2000; Pastel 2002). While investigation into the disease has yielded many findings over the last

few decades, the full nature of the disease remains unresolved. The purpose of this review is to

examine the literature concerning a few core biochemical pathways, such as mitochondrial

dysfunction, the cellular oxidative stress response, and inflammatory processes, where

radiation exposure is known to cause perturbations and where there is evidence that the

pathway is aberrant in patients with CFIDS. By drawing the two largely independent fields of

CFIDS and radiation research together, it is hoped that common ground can be found which

could inform new testable hypotheses in the area.

CFIDS is a disease primarily characterized by persistent or recurring long-term fatigue and

susceptibility to infections (Holmes et al. 1988). Patients present a combination of other

symptoms in addition to the ubiquitous lassitude, including headache, muscle aches,

impairment of neurocognitive functions, and reduced quality of sleep (Evengård et al. 1999),

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 7: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

symptoms usually encountered in patients following acute radiation exposure as well

(Kumerova et al. 2000; Pastel 2002). Considering that the disease has affected millions since it

was discovered, it is surprising that researchers are still uncertain about the exact cause of

CFIDS and the mechanisms behind its manifestation. Despite some stigma towards the pursuit

of research concerning CIFDS outside of a psychopathology (Anderson et al. 2013), recent

studies have shown that CFIDS almost certainly has a physiological basis. While it is important

to distinguish between the putative immunologic, physiologic, and neurologic models of CFIDS,

it is also important to understand, as eloquently argued by Whiteside & Friberg (1998), the

potential for interconnectedness among the three and work towards developing a

comprehensive model that explains all facets of the illness. Among the potential immunologic

and neurologic factors that may contribute to the onset and maintenance of CFIDS pathology,

one of the most exciting areas involves the investigation of the metabolic symptoms of the

disease.

CFIDS, Mitochondria, and Metabolism

A recent comprehensive study of the metabolomics of patients with CFIDS carried out by Dr.

Robert Naviaux et al. (2016) effectively showed that the disease can be characterized as a

“highly concerted hypometabolic response” to environmental stress, and one that can be easily

traced to mitochondria. Specifically, the study showed that the metabolic response was

homogeneous to heterogeneous stressors by examining a variety of biochemical pathways that

were consistently altered or abnormal in patients with CFIDS. The study also suggested that

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 8: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

every metabolic abnormality observed in CFIDS patients was either regulated directly by redox

reactions or the availability of NADPH. In addition to this, they found that many of the NADPH-

dependent enzymes identified in the study had mitochondrial isoforms that were known to be

upregulated during periods of environmental stress. They describe NADPH as a “global

barometer of cellular fuel status” by interacting with both mitochondrial NADH consumption

and the availability of similar reducing agents in the cytosol. Considering the findings of this

paper, it is not only appropriate to propose that CFIDS may be intrinsically linked to metabolic

dysfunction, but also that it may be consequently inextricably linked to the function, or rather

the dysfunction, of mitochondria.

The idea that CFIDS is correlated to the improper functioning of mitochondria is not a radically

new one; in fact, it has been described by many authors in the past. When taking into account

the symptoms of CFIDS alone, they all appear to stand out as a hallmark of mitochondrial

dysfunction in terms of energy production and deficit in the cell (Filler et al. 2014). The same

review by Filler et al. (2014) proposed that significant downregulation of genes associated with

antioxidant mechanisms, aerobic energy production, and metabolism in peripheral blood

mononuclear cells of CFIDS patients when compared to control groups. The article also

described proper mitochondrial function as negatively correlated with fatigue symptoms and

another study determining the partial blockage of an ADP-ATP translocator in the mitochondria

in patients with CFIDS (Booth et al. 2012). Oxidative stress, which can be caused by the

presence of compounds such as radical oxygen species (ROS) and radical nitrogen species (RNS)

in a cell, is another feature indicative of mitochondrial dysfunction (Lin & Beal 2006) which is

also observed in patients with CFIDS, according to the literature. One study (R Kurup & P Kurup

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 9: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

2003) showed that antioxidants, which include compounds such as superoxide dismutase that

are produced by cells to combat oxidative stress, were decreased in patients with CFIDS; in

addition to this, they suggested that, due to decreased free-radical scavenging, that ROS and

RNS were increased in plasma samples of CFIDS patients when compared to control groups

potentially leading to mitochondrial dysfunction. They proposed that free-radical generation

could be implicated in the pathogenesis of CFIDS, accounting for muscle pain and fatigability in

patients. ROS have been shown to be involved in a variety of diseases and therefore their

presence generally results in a greatly diminished quality of life if improperly regulated in

human cells; high concentrations of ROS in a cell can cause oxidative damage to integral

metabolic components (Panieri & Santoro 2016), DNA damage (Tada-Oikawa et al. 2000), and

can eventually induce apoptosis (Armeni et al. 2004). Interestingly, one clinical study (Fulle et

al. 2000) noted specific oxidative modifications and reduced oxidative metabolism in the

myocytes of patients with CFIDS. Another study (Wawrzyniak et al. 2016) showed impaired

biogenesis signalling and abnormal mitochondrial content in the myocytes of older individuals

with idiopathic, or rapid-onset, chronic fatigue. When taken as a whole, these findings suggest

that oxidative muscle damage and impaired mitochondrial function could be culprits in the

onset of CFIDS. Due to the presence of mitochondrial dysfunction in patients with CFIDS and

organelle’s tendency to produce damaging radical chemical species (RCS) if perturbed, the

mechanisms that contribute to oxidative stress and their relationship to the disease should be

of particular interest to researchers.

According to a study by Myhill et al. (2009), there is considerable evidence that mitochondrial

dysfunction is present in some sufferers of CFIDS. Biopsies of human muscles in CFIDS patients

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 10: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

have shown abnormal mitochondrial degeneration. (Byrne et al. 1985; Behan et al. 1991;

Vecchiet et al. 1996). Further studies have shown that CFIDS patients contain deletions in

mtDNA genes associated with energy production, specifically in skeletal muscle (Zhang et al.

1995; Vecchiet et al. 1996). Increased activity of antioxidant enzymes and RCS-linked oxidative

damage has been observed in the muscles of CFIDS patients (Fulle et al. 2000). Decreased levels

of chemicals essential in metabolic reactions in mitochondria have also been shown to be

deceased in CFIDS patients. (Kuratsune et al. 1994; Plioplys and Plioplys 1995). Reduced

oxidative metabolism has also been observed in patients with CFIDS along with higher levels of

lactate and pyruvate (Arnold et al. 1984; Buist 1989; Wong et al. 1992; McCully et al. 1996; Lane

et al. 1998). Two studies showed that lipid replacement and antioxidant nutritional therapy

administered through a dietary regimen can help restore mitochondrial function and alleviate

fatigue symptoms (Nicolson 2003; Nicolson and Ellithorpe 2006).

The Evidence for Potential Involvement of Radiation

Many publications have linked ionizing radiation to the mitochondria-dependent generation of

ROS and RNS, primarily linking it to radiation-induced bystander effects and genomic instability

(reviewed in Szumiel 2015). According to Azzam et al. 2012, short and long-term radiation

induced ROS/RNS could result to damage to mitochondrial DNA in genes coding for electron-

transport subunits and their associated proteins. Cells without a functioning electron transport

chain do not experience ROS/RNS production (Leach et al. 2001). Cells with ROS/RNS induced

mtDNA damage could produce dysfunctional proteins associated with the electron transport

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 11: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

chain, resulting in pro-oxidant chemical formation, which can eventually lead to de-novo

oxidative damage to biological structures long after radiation exposure with the mtDNA

potentially being passed on to daughter cells after division contributing to genomic instability

(Bogenhagen and Clayton 1977; Spitz et al. 2004; Gaziev and Shaĭkhaev 2007; Azzam et al.

2012). An in-vivo study using mice exposed to 0.2 Gy of x-rays found significant changes in

pyruvate metabolism and structural proteins persisting four weeks after exposure in heart cells.

Higher doses resulted in respiratory complex I and III partial deactivation as well (Barjaktarovic

et al. 2011). A few studies have shown that mitochondrial protein import could be used as a

marker for the long-term effects of ionizing radiation exposure, even at small doses, as this

could be the result of damage to components in translocational machinery or changes to

membrane potential (Pandey et al. 2006; Azzam et al. 2012).

After reviewing the literature, it is plausible to suggest that the production of ROS due an

endogenous or exogenous cause can contribute to the dysfunction of mitochondria in patients

suffering from CFIDS, among other potential causes. It is generally accepted that the primary

target for radiation effects in biological systems is DNA. However, in addition to the targeted

effects of ionizing radiation on DNA, the radiolysis of water and other compounds a human cell

result in the formation of ROS which can directly alter DNA through oxidative damage (Dandona

et al. 1996). Many RCS are known to damage many biological components in cells other than

DNA, including membranes that are essential to the homeostasis of the cell (Bonnekoh et al.

1990; Kryston et al. 2011; Martin et al. 2011). A high concentration of ROS in the cell can

critically compromise the mitochondrial membrane and affect the mitochondrial membrane

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 12: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

potential, which is crucial for the production of the majority of aerobic energy in cells (Tada-

Oikawa et al. 2000). As reviewed earlier, many studies have linked the production of RCS to

mitochondrial dysfunction and to CFIDS, suggesting involvement of factors such as lipid

peroxidation, mtDNA damage, pyruvate metabolism, and protein import. Other studies outside

of the field have observed very similar events in cells exposed to ionizing radiation, both at high

and low doses. Considering the broader metabolic abnormalities in individuals suffering from

CFIDS and the link to ATP production (Myhill et al. 2009), there is some compelling evidence in

contemporary scientific literature to warrant the association of CFIDS and mitochondrial

dysfunction with ionizing radiation exposure and general RCS production. If these connections

are juxtaposed with the strong evidence for a biophoton bystander effect as described by Le et

al. in Radiation Research (2015) and Le et al. in Physics in Medicine and Biology (2015), it is

reasonable to suggest that CFIDS may be aggravated or even caused by low level radiation

exposure through a novel signalling cascade.

It is difficult to estimate how many different pathways are linked to mitochondria that might be

affected by ionizing radiation either directly or indirectly by bystander signaling. The approach

in this paper is to focus on pathways and proteins involved in low dose radiation response that

are also suggested as being involved in CFIDS. The particular focus is on involvement in the

phenomena of cellular oxidative stress and altered cellular energy metabolism and production. Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 13: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Biochemical Pathways and Proteins of Interest

Alpha-synuclein

Alpha-synuclein (α-syn) is, by virtue of scientific debate, either an intrinsically unfolded protein

(Riedel et al. 2011) or a protein that might undergo folding with the help of heat shock proteins

(HSPs) to assume its native conformation in some circumstances (Bruinsma et al. 2011). In

humans, α-syn has no known function other than its deleterious role in disease (Riedel et al.

2011). α-syn is known to readily oligomerize, fibrillate, and aggregate, and is the major

component of inclusion bodies in cells of patients with Parkinson’s, Multiple System Atrophy,

and certain forms of Dementia (Bruinsma et al. 2011). Specifically, the same paper showed that

α-syn is known to interact with many HSPs, including αB-crystallin and sHspB8, with different

mutations altering the ability of HSPs to prevent fibrilization.

α-syn is thought to have two major forms: a membrane-bound and nonmembrane bound form.

This is believed due to the finding that α-syn has the capability to interact with poly-

unsaturated fats, which, in turn, may help promote the oligomerization process (Riedel et al.

2011). Following this finding, it was postulated that α-syn exists in its nucleating form when

bound to membranes and its monomeric form when in the cytosol (Riedel et al. 2011). Because

unsaturated fats are known to easily undergo lipid peroxidation after exposure to ionizing

radiation (Hammer & Wills 1979), it was also proposed that subsequent α-syn oxidative

modification by lipid peroxides and their radical intermediates generated from proximal

membrane material could promote its aggregation (Thomas et al. 2007; Riedel et al. 2011).

Studies concerning the tendency for α-syn to aggregate independent of cellular components

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 14: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

after overexpression and ROS introduction respectively have also been conducted

(Andrekopoulos et al. 2004; Uversky et al. 2005). Aggregation of α-syn consequently promotes

further oxidative stress in the cell (Flower et al. 2005) and could lead to the compromising of

broader cellular metabolism. Indeed, increased α-syn levels have already been linked to

compromised mitochondrial function and altered cellular metabolism (Hu et al. 2009; Brown et

al. 2016a). A recent article (Brown et al. 2016a) demonstrated that α-syn binds to TOM20, a

mitochondrial outer membrane translocase receptor, causing deficient mitochondrial

respiration, increased ROS production, and loss of membrane potential. Other studies have

shown that mitochondrial dysfunction and ER stress can lead to α-syn-mediated cell death

(Smith et al. 2005). Another study (Booth et al. 2012) found that blockage of mitochondrial

membrane proteins is a potential biomarker of CFIDS. Therefore α-syn should be further

examined with regards to its potential role in CFIDS and other diseases.

One study (Saligan et al. 2013) also showed statistically significant correlations between α-syn

induction, ionizing radiation, and fatigue symptoms. The extensive study showed that cancer

patients exposed to gamma rays by external beam radiotherapy (EBRT) showed elevated α-syn

expression after the therapy. The paper also found that the fatigue experienced by patients

following the treatment was significantly correlated to α-syn plasma concentration, which

indicates that the EBRT produced the same effects on α-syn expression as it did on the fatigue

experienced by the patients. While this study alone does not conclusively link α-syn to fatigue

and radiation exposure, it raises some important questions concerning its role in cellular

metabolism and inflammation in relation to fatigue symptoms. The underlying mechanisms of

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 15: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

this relationship could prove very insightful in the study of CFIDS and its association with

ionizing radiation.

In conclusion, α-syn could be a protein that is highly involved in the development or

aggravation of CFIDS in many ways. Its link to membranes and lipid peroxidation, mitochondria

and cellular metabolism, and radiation treatment-related fatigue symptoms warrant further

investigation into its role and relationship to ionizing radiation in the context of CFIDS (Figure

1).

Lactate Dehydrogenase and STAT1

Lactate Dehydrogenase (LDH) is an enzyme that is involved in the reaction that converts lactate

to pyruvic acid and back using NAD+ and NADH in the cell. Because of its abundance in the cell

and because it is mainly released following tissue damage, it is an important marker for

measuring alterations to the cell membrane (Bonnekoh et al. 1990; Armeni et al. 2004).

Presence of excess lactate has been linked to plasma acidosis and muscle fatigue (Lemire et al.

2008). Studies conducted in the past few years have shown that the importance of LDH may be

greater than previously thought, as lactate was identified as a major energy source for human

neurons (Hall et al. 2009; Boumezbeur et al. 2011; Wyss et al. 2011). Lactate concentrations are

positively correlated with radioresistance, which could indicate intrinsic antioxidant properties

(Hirschhaeuser et al. 2011).

LDH subtype A (LDH-A) is an isozyme that is highly expressed in various kinds of malignant

tumors and is probably heavily involved in the Warburg Effect (Fantin et al. 2006). The Warburg

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 16: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Effect is primarily used to describe the metabolism of tumor cells, wherein a switch is made in

generating the majority of energy between oxidative phosphorylation and aerobic glycolysis

(Vander Heiden et al. 2009). Reduction of oxidative phosphorylation efficiency has been

established in CFIDS patients in neutrophils, with a switch to glycolysis postulated after blood

samples were taken from CFIDS patients and assayed (Booth et al 2012). When taking into

consideration the fact that mitochondrial oxidative phosphorylation produces ROS, it is thought

that this effect limits the production of ROS in normal cells (Vander Heiden et al. 2009). LDH-A

is also considered a potential target of cancer suppression. A recent study (Kim et al. 2014)

showed that inhibition of LDH-A activity decreases the conversion rate from pyruvate to lactate,

and consequently mitochondrial membrane potentials and cellular ATP levels as well; this

ultimately leads to oxidative stress in the cell. Reduction of the production of ATP is one of the

many biomarkers that have recently been discovered in CFIDS patients’ blood and has been

tested extensively (Myhill et al. 2009). A few studies have further linked LDH to mitochondria by

indicating that LDH can exist inside mitochondria, contributing, much like cytosolic LDH, to

promoting a lactate balance in cells by dehydrogenating lactate produced in the cytosol (Brooks

et al. 1999). While there was some debate about the existence of LDH in mitochondria

(Rasmussen et al. 2002), a few recent studies have unequivocally confirmed its presence

(Lemire et al. 2008; Passarella et al. 2014).

Signal Transducer and Activator of Transcription 1 (STAT1) is a transcription factor in humans,

traditionally thought to be a protein involved in interferon signalling. Pitroda et al. (2009)

identified STAT1 as a transcription factor of LDH-A, among many other proteins involved in

metabolic energy pathways. They suggested that STAT1 works closely to regulate glycolysis and

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 17: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

even demonstrated that STAT1 helps to rescue the expression of proteins essential to

metabolic energy pathways after x-ray induced suppression. Another paper studied the

exposure of STAT1 to UVA. After exposure to UVA, the modulation of STAT1 expression

assumed a biphasic characteristic, according to Mazière et al. (2000). They also found that this

effect was reduced with the introduction of antioxidants, indicating the presence of ROS and

oxidative stress probably play a role in the modulation of STAT1. Furthermore, the regulation

and activation of STAT1 is complicated by the need for tyrosine/serine phosphorylation and the

MAPK/ERK pathway, as inhibiting either phosphorylation or the MAPK/ERK pathway inhibits the

activation of STAT1. STAT1 is also particularly interesting because of its close involvement with

interferon signalling; pertinent to CFIDS, STAT1 is involved downstream of interferon gamma, a

signalling molecule modulated and typically decreased in lymphocytes of patients with CFIDS, in

one pathway (Klimas et al. 1990; Visser et al. 1998; Patarca 2001; Mulero et al. 2015; Baris et al.

2016). The potential role of STAT1 in regulating cellular metabolism and LDH-A could suggest its

involvement in a variety of diseases, in addition to its link to interferon signalling in the context

of CFIDS. Moreover, the ability of ionizing radiation to affect, let alone modulate, the

expression of STAT1 is cause enough for further research into the effects of ionizing radiation

on LDH and its regulators, the interplay of ROS, and ultimately the pathways’ possible

contribution to CFIDS. Further discussion of STAT1 in relation to the expression of RNase L is

discussed in the Ribonuclease L section.

Studies have also shown that radiation-induced senescence of human breast cancer cells also

caused the upregulation and activation of LDH (Liao et al. 2014). The study by Liao and his team

also showed that, by altering the monocarboxylate transporter 1 (MCT1), adenosine

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 18: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

monophosphate-activated protein kinase (AMPK), and the nuclear factor kappa-light-chain-

enhancer of activated B cells (NF-κB) signalling pathways, radiation can effectively induce the

senescence of cells by metabolic alterations. The AMPK pathway is particularly interesting, as it

mediates the cellular response when the AMP/ATP ratio becomes too high as the cell

experiences energetic stress. The study also showed that invasion of surrounding cells and

radiation-induced bystander signalling was dependent on the MCT1, AMPK, and NF-κB

pathways. Recent studies have linked the NF-κB pathway and loss of p53 directly to CFIDS

(Morris & Maes 2012). A very interesting review proposed a common etiology between post-

radiation syndrome and CFIDS. Pall (2008) described post-radiation syndrome as a CFIDS-like

disease and linked the entire illness to the nitric-oxide/peroxynitrite cycle (NO/ONOO-). The it

was proposed that the cycle involves NF-κB and interferon gamma signalling and oxidative

stress, culminating in a “vicious cycle” that chronically elevates levels of nitric oxide. It was also

concluded that radiation can, even at relatively low doses such as those experienced by

Chernobyl recovery workers, can lead to the activation of this cycle, primarily through NF-κB.

Moreover, another study (Badalà et al. 2008) found elevated ventricular lactate in CFIDS

patients and implicated oxidative stress as a cause of the disease. These findings suggest that,

while CFIDS may be a complicated disorder with many different symptoms, more research must

be done in order to rule out the direct involvement of LDH and its associated proteins in the

illness.

LDH is an important protein with respect to cellular metabolism and energy production. The

involvement of LDH and its associated pathways in the Warburg Effect, oxidative stress,

mitochondrial structural and functional integrity, STAT1 regulation pathways, and radiation-

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 19: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

induced activation and senescence all necessitate future endeavours to fully understand its

potential role in contributing to CFIDS symptoms (Figure 2).

Cytochrome c and Cytochrome c oxidase

Cytochrome c oxidase (CCO) is a large multi-protein complex located in the mitochondria of

eukaryotes. Also known as Complex IV, it is a main component of the electron transport chain,

where it donates four electrons to molecular oxygen and transfers four protons to it as well,

creating two molecules of water in the process. In addition to this, it pumps protons into the

intermembrane space of mitochondria to create the electrochemical gradient required to make

ATP (Denis 1986). Cytochrome c (CC) is a hemeprotein associated with the inner membrane of

mitochondria; essential for the electron transport chain as well, it has the capacity to carry a

single electron, to donate that electron during oxidative phosphorylation, and to shuttle

electrons between Complex III and Complex IV.

CC is crucial in many pathways in the cell. Its release from the mitochondria following

mitochondrial permeabilization is necessary for many pathways leading to apoptosis (Yang

1997). Its involvement in the electron transport chain makes its uncompromised role an

important factor in the viability of cells with respect to energy production. CC has the capability

to produce intramitochondrial ROS when interacting with several proteins, leading many to

believe that it is involved in signalling and mitochondrial damage through ROS-propagation

pathways (Giorgio et al. 2005; Marchi et al. 2012; Panieri & Santoro 2016).

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 20: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

A study by Tirelli et al. (1994) indicated that there were no significant differences in CCO

reduction between control groups and CFIDS patients. Indeed, a few other older scientific

articles note no significant disparities between the amount of CCO reduction in healthy

individuals versus CFIDS patients, particularly in muscle tissue where the energy demand is high

for cells (Byrne & Trounce 1987). However, more recent publications have noticed some

important connections between CCO and CFIDS.

The aforementioned link that RCS have to mitochondrial dysfunction and CFIDS has been

studied extensively in the literature. A study (Brown 2001) found that CCO can be reversibly

impaired by a radical nitrogen species, NO in this case, as CCO competes with it for access to

oxygen. It noted that prolonged or high dosages of NO or any of its derivatives can cause

irreversible deleterious effects, such as CCO’s uncoupling, mitochondrial permeability, and even

cell death. Another article noted that CC can be modified by nitration of its tyrosine residues

after low doses of peroxynitrite are administered (Marchi et al. 2012). The authors found that

the nitration of CC also resulted in the upregulation of peroxidase activity for hydrogen

peroxide in the cell and the impairment of membrane potential formation, likely a result of

damaged subunits in the electron transport chain. Oxidative damage to mitochondrial proteins

and complexes crucial to oxidative phosphorylation not only results in impaired ATP

production, but also unregulated ROS production by the affected complexes, which leads to

amplification of ROS in mitochondria and increased release into the cell (Hosseini et al. 2014;

Zorov et al. 2014). It has been shown that the production of peroxynitrite and its derivatives,

NO included, can be induced by x-ray exposure in mice (Hanaue et al. 2007), which could

potentially lead to CC nitration, multi-complex corruption, RCS proliferation, mitochondrial

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 21: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

membrane depolarization, and reduced ATP production leading to ROS contribution to fatigue

and even tissue insult as a result of mass cell death. The theme that the nitric oxide cycle and

elevation of NO has something to do with CFIDS in humans is pervasive in the literature; many

papers have been published on the topic, including a few linking the nitric oxide cycle to

radiation exposure and PRS (Pall and Satterlee 2001; Pall 2007, 2008, 2009).

The biggest problem with studying CC and CCO is their nature as incredibly complicated

macromolecules, particularly CCO. Be that as it may, the roles that they might have in CFIDS

should not be underestimated. Both molecules are already being studied heavily, and the fact

that have already been implicated in a number of diseases only further necessitates research.

The discovery of RCS signalling pathways and the involvement of CC in them, the potential for

exogenous RNS to affect the metabolism of mitochondria through direct covalent interactions

with CC and competition with CCO, and the demonstrated radiation-induced production of a

reactive species of nitrogen implicated in these cascades in vivo (Figure 3) beckons the need for

further research into CC and CCO.

αB-crystallin

αB-crystallin (αBC), encoded by the gene CRYAB, is a protein that is a member of the small heat

shock protein (sHsp) family and is involved in binding to unfolded proteins to inhibit their

aggregation and ultimately preventing apoptosis (Boelens 2014). αBC interacts with many

proteins, inhibiting the formation of mature α-syn fibrils (Hsu et al. 2000; Riedel et al. 2009),

reducing the ubiquitination and degradation of a pore-forming subunit of major cardiac voltage

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 22: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

gated sodium channel (Huang et al. 2016), and binding to p53 in addition to several pro-

apoptotic proteins and preventing their translocation to mitochondria (Liu S et al. 2007). In fact,

the interplay between p53 and αBC shows tight regulation on a transcriptional and post-

translational level and affects oxidative stress and the Warburg Effect in human cells. It was

discovered that p53 binds directly to the promoter region of CRYAB to enhance its expression;

transfection of a p53 overexpression vector into MCF-7 cells increased expression of αBC,

reduced intracellular ROS, caused the Warburg Effect, and increased ROS after p53 was

depleted (Liu S et al. 2014). It was also shown, as stated previously in the LDH section, that

reduced levels of p53 is indicative of mitochondrial dysfunction and directly linked to CFIDS.

Many sHSPs are mobilized during oxidative stress (Dimauro et al. 2016), and in case of αBC, a

few crucial mitochondrial pathways involved in ROS production and cell damage might be one

of the major reasons for this.

αBC was shown to be modified heavily by the reactive nitrogen species peroxynitrite, where

peroxynitrite enhanced its chaperone and antioxidant abilities (Morris & Maes 2012). This is

very interesting when noting that peroxynitrite is heavily involved in compromising CC and CCO,

as discussed in the Cytochrome c section, and is thought to be central to the etiology of CFIDS

and PRS (Pall and Satterlee 2001; Pall 2007; Meeus et al. 2008; Pall 2008, 2009). Specifically,

there appears to be the potential for rescue in the Cytochrome c and Cytochrome c oxidase-

mediated ROS pathway after radiation exposure when considering the scientifically-

corroborated observation of radiation-induced formation of peroxynitrite (Zorov et al. 2014).

αBC was also shown to inhibit the release of Cytochrome c from mitochondria (Xu et al. 2013),

potentially by binding to mitochondrial transmembrane channels. As shown by Chis et al.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 23: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

(2012), Cytochrome c co-localizes with αBC during periods of oxidative stress, as are other pro-

apoptotic proteins. αBC was also shown to have antioxidant properties by binding directly to

Cu2+ ions and rendering them inactive (Ahmad et al. 2008). Crystallin genes have been shown

to be highly induced following radiation exposure in human cells and are closely linked to

mitochondria-stress pathways (Andley et al. 2000; Parcellier et al. 2005; Chaudhry 2006).

Could αBC be implicated in CFIDS? While the connections made to CFIDS and radiation may not

be conclusive, this chaperone is clearly involved with many aspects of proper cellular function.

The fact that αBC is involved in regulating every protein discussed in this paper, the fact that

increased αBC is correlated with increased lactate levels and the Warburg Effect, and the fact

that is relatively sensitive to ionizing radiation only speaks to the need to research αBC with

respect to radiation and CFIDS (Figure 4).

Ribonuclease L

Ribonuclease L (RNase L) is a ribonuclease that, upon activation, destroys RNA that resembles

viral RNA. RNase L is mobilized after either a type I interferon signalling cascade involving the

stimulation of JAK kinases and after activation of STAT1 and/or STAT2 results in transcription of

2,5A Synthase (2-5OAS); this synthase is activated by ss and dsRNA to create 2’-5’-

oligodendylates (2-5A), which bind to and activate RNase L (Stark et al. 1998; Silverman 2007a).

RNAse L is also believed to be involved in apoptosis, specifically during the cell’s efforts to enter

apoptosis following viral infection (Chakrabarti et al. 2011). Indeed, after a certain point of

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 24: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

activation, RNAse L is capable of destroying essential RNA in the cell in an attempt to induce

apoptosis to prevent the infection from spreading (Silverman 2007b).

It has been believed for some time that RNase L cleavage is linked to CFIDS. The protein exists

in its functional 83 kDa form in both CFIDS and healthy groups. Various truncated forms of

RNase L also exist, albeit mostly in unremarkably similar concentrations in both groups.

Curiously, some studies suggest that the 37 kDa form of the protein exists preferentially in

CFIDS groups (Suhadolnik et al. 1997; De Meirleir et al. 2000). One study in particular

attempted to link the upregulated activity of elastase to the cleavage of RNase L (Meeus et al.

2008). The same study found significant amounts of truncated 37 kDa RNase L in patients with

CFIDS. Upregulation of 2-5OAS has been directly linked to CFIDS as well, and one study found a

significant correlation in the expression of 2-5OAS with fatigue and depression symptoms

stimulated by interferon alpha treatment in patients with Hepatitis C (Felger et al. 2012).

RNAse L has been shown to have capabilities to cleave 28s rRNA in mice as part of an attempt

to prevent viral proliferation (Iordanov et al. 2000); another study showed RNase L-

independent 28s cleavage can occur during viral infection (Banerjee et al. 2000). A study by

Iordanov et al. (2000) suggested that the degradation of 28s rRNA, which makes up the large

ribosomal subunit, could lead to overall reduced translation in the cell and induction of pro-

inflammatory cytokines to the cell as part of the innate immune response. Such an implication

would mean that the loss or dysregulation of RNase L in any way could lead to an inappropriate

response to viral infection and serious immune dysfunction, which could be a route to

developing the immune symptoms present in CFIDS.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 25: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

The possibilities for the involvement of radiation in this pathway are virtually limitless and

mostly unexplored in the literature. For instance, it has been shown that RNAse L plays an

important role in upregulating the cellular response to gamma radiation-induced DNA damage

by affecting the transcription of proteins such as p21 (Al-Haj et al. 2012). It has been shown

through several studies that various kinds of kinases, including MAP and ATM kinases, can be

activated by ionizing radiation to phosphorylate their targets (Chen et al. 1996; Canman et al.

1998; Iordanov et al. 2000). Results from the Maziere et al. paper (2000) imply that the biphasic

modulation of STAT1 following radiation exposure might very well have something to do with

MAPK, JAK, or other similar tyrosine kinases, as inhibition of tyrosine kinase activity inhibited

the UVA-effect on STAT1. STAT1 is required for the transcription of 2-5OAS, which is essential

for the activation of RNase L among other interferon response genes in the Type I pathway

expressed throughout the human body in a multitude of tissues during viral infection (Mullan et

al. 2005). STAT1 is also necessary and sufficient to induce transcription of interferon-stimulated

genes in the Type II pathway specific to Type II interferons released from activated T

lymphocytes (Durbin et al. 2000; Mullan et al. 2005).

As described in the STAT1 section, it is important to note that there are many potential

pathways that may link radiation to its pathway and eventually CFIDS. Dysregulation of the

RNase L pathway is an interesting possibility for the manifestation of CFIDS. Many questions

still remain unanswered in this pathway with regards to radiation and CFIDS, necessitating

further research.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 26: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Potential New Areas for Research

The literature discussed above confirms that ionizing radiation has been shown to be capable of

perturbing various pathways thought to be involved in the manifestation of CFIDS. Some areas

for future research are discussed below in the hope of contributing to our understanding of

CFIDS and its potential connection to radiation exposure.

Involvement of the Bystander Effect

Because of the potential for signal propagation through cell populations (Nagasawa and Little

1992; Mothersill and Seymour 1998, reviewed in Mothersill et al. 2017a; 2017b) as a

mechanism which can magnify the size of the target for effects after low doses, the following

experiments might help to determine if the effects can be extrapolated to living multicellular

organisms following localized radiation exposure. Such studies could lead to a better

understanding of the potential for cellular symptoms of radiation to proliferate in a cell

population to affect the system as a whole. Comparison of results with and without the

compounds discussed above would enable us to determine if the radiation response pathway

were actually impacted by the interference with the biochemical pathway.

The Peroxynitrite Cycle

The effect of the presence of peroxynitrite on αBC, CC, and CCO (Brown 2001; Andrekopoulos

et al. 2004) could be a primary focus as a potential pathway in the proliferation of ROS,

mitochondrial and cell damage, energy deficiency, and apoptosis. As noted earlier, the

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 27: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

formation of peroxynitrite can be induced by x-ray exposure, particularly in mice (Hanaue et al.

2007), and a few papers (Pall 2007, 2008, 2009) have noted that RNS pathways could be the

missing link between CFIDS and radiation exposure in humans.

The first step would be to identify a human cell line where the production of peroxynitrite

occurred. This would be of high interest with regard to CFIDS. A control with established

baseline concentrations of peroxynitrite and its derivatives would have to be determined,

before looking at the impact of ionizing radiation on other samples. The baseline production of

other ROS and RNS species could be assayed as well in this step. If initial results are promising

an experiment attempting to correlate the radiation dose, the peroxynitrite concentration, and

the nitration of Cytochrome c could be performed. Assaying the modification of Cytochrome c

could be done using sodium dodecyl sulfate polyacrylamide gel electrophoresis using a

molecular weight ladder and comparing the untreated lane to the varying radiation

dosages/concentrations of peroxynitrite. Using SDS-PAGE in this way could show a shift in mass

of cytochrome c between the untreated samples and the treated samples, answering the

question of whether increased radiation exposure leads to increased nitration of Cytochrome c.

If this does not work due to the change in mass being potentially too small to detect, other

methods could be discussed. Again, this experiment would only attempt to establish whether

increasing doses of gamma radiation exposure alter Cytochrome c through nitration

mechanisms.

Using immortalized human cell lines, preliminary clonogenic assays could potentially show the

reduction in viability of cells that undergo this kind of alteration to Cytochrome c, which could

potentially link the alteration to damaged electron transport chain (ETC) components.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 28: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Bystander signals are already known to impact the ETC (Le et al. 2017) This step would

putatively link reduced viability caused by radiation exposure to cellular respiration based on

Cytochrome c’s intimate involvement in oxidative phosphorylation. A direct assay of the

alterations in each Complex by various methods might reveal some damage as well, which

would support the theory, although not directly. An assay of the functionality of Cytochrome c

in irradiated cells would have to be done to implicate it in reduced cell viability.

αB-crystallin could be involved in several ways in this process. It is suspected, based on

literature review, that alterations to αBC by peroxynitrite enhance its antioxidant and

chaperone properties. There is the potential to measure the rescue of Cytochrome c by

showing that αBC binds directly to Cytochrome c after irradiation and consequent nitration, as

it has been shown to co-localize with unaltered Cytochrome c (Chis et al. 2012). This could be

confirmed using a co-immunoprecipitation assay. This would involve isolating modified

Cytochrome c using an antibody, then identifying potential binding partners using a Western

Blot in the hopes of identifying αBC. It would also be interesting, assuming Cytochrome c is

bound by αBC after irradiation, if the affinity increases or decreases with more nitration and/or

exposure to radiation.

Because Cytochrome c oxidase is involved in competition with nitrogen species, this could be a

novel mechanism induced by ionizing radiation that could be linked to oxidative stress and

CFIDS in the future. Considering that ROS production, Cytochrome c, and αB-crystallin are

involved in cell metabolism, energy production, and have been linked to CFIDS in some way,

this pathway that involves the production of ROS, the loss of function of Cytochrome c, and αB-

crystallin is interesting when considering its ties to ionizing radiation. This experiment would

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 29: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

clearly not determine if this pathway operates in vivo in those suffering from CFIDS, however

elucidation of these pathways and their associated proteins is most definitely a good start.

P53, αB-crystallin, and ROS

Some very interesting regulation can be found in the p53 and αBC pathway. It appears as

though αBC negatively regulates the apoptotic functionality of p53 (Liu S et al. 2007). P53

positively regulates the expression of αBC (Liu S et al. 2007; 2014) and a number of proteins

that probably interact with other proteins involved in cell metabolism in order to induce the

Warburg Effect (Morris & Maes 2012; Liu S et al. 2014; Panieri & Santoro 2016).

No published experiments have yet assayed αBC’s resilience to gamma radiation using small

doses. It would be interesting to irradiate a solution containing αBC in the soluble fraction at

low doses, run it on an SDS-PAGE gel, determine if there are any appreciable changes, and then

determine what percentage of αBC was lost due to the radiation compared to the control. The

same experiment could then be done, with cells. The cells could then be lysed, αBC isolated,

and then measured. This would be done alongside unirradiated controls to determine if the

fraction of functional αBC was appreciably altered. Clonogenic assays would be done at each

endpoint to determine if this effect can be correlated with survivability. If the results between

the experiments using the chemical alone or the chemical in cells are different, then we can

postulate that ROS had a role in the damage done to αBC in the cell. It could suggest that ROS

generated inside cells might have played a role in αBC’s reduction. This would suggest that

certain photosensitizers might help in the propagation of ROS and the damage that they do

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 30: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

inside cells. If the converse was seen, then some compound or protein in living cells might be

protecting αBC from alterations caused by radiation that is not present in the chemical solution.

Further studies could be done to determine what radioprotector this might be, potentially by

reviewing the literature to identify putative binding partners and using coimmunoprecipitation

to validate them.

Considering that there are p53 +/+ and p53 -/- cell lines available, similar experiments can be

done on both cell lines. Firstly, the disparity between cells with no p53 and cells with p53

concerning expression of αBC would have to be established. Then, experiments could be done

in a similar fashion as above to try to link the reduction of αBC to pro-apoptotic pathways

involving p53, a shift to more glucose in the cell, and ROS production. Even though these

experiments would be performed on human cell lines and the results would not be directly

applicable to CFIDS patients, determining how these pathways behave in certain situations

could give researchers more potential biomarkers to observe in CFIDS patients after follow-up

studies testing for relevance in such patients.

Lactate Dehydrogenase and the Warburg Effect

In one reviewed study, it was shown that ventricular lactate levels were significantly higher in

people with CFIDS (Badalà et al. 2008). This raises the question of whether or not rising

intracellular and intercellular lactate levels have a role to play in CFIDS. Perhaps damaged

mitochondria due to ROS induce a cellular response to use more aerobic glycolysis in the

cytosol while some mitochondria shut down. The problem with LDH is that it can be involved in

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 31: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

a number of ways in the onset or aggravation of CFIDS: LDH could protect the cell from

oxidative damage, facilitate the switch to aerobic glycolysis if the mitochondria are damaged, or

facilitate the switch indirectly through a number of different pathways.

The mechanisms of such a switch could be triggered by radiation exposure. It has been shown

that lactate could be a radioprotective molecule (Hirschhaeuser et al. 2011) and that LDH-A can

induce oxidative stress if it is downregulated in cancer cells (Panieri & Santoro 2016). If LDH is,

in fact, an indirect radioprotective element in normal cells, then its loss would render cells more

susceptible to radiation. To determine if ROS play a major role in the radioprotective nature of

LDH, an assay measuring the antioxidant properties of LDH might be appropriate where radical

chemical species’ levels are being measured. A simple experiment would be to perform an

assay in a solution where RCS can be generated by radiation. Different doses of radiation can be

applied to each flask. After irradiation and measuring of relative reactive species in the

solution, LDH can be added to each with varying concentrations. The assay would be performed

again to measure RCS. If the trials show reduced RCS, then it could suggested that LDH is

sufficient as an antioxidant and can protect against radiation-induced generation of RCS

irrespective of the presence of other proteins or chemicals. If no significant reduction is seen,

then LDH might not be an antioxidant at all; much more likely than this however, is that LDH

does not have intrinsic chemical antioxidant properties, but might reduce reactive species in

the cell in the proper chemical environment and when assisted with auxiliary proteins. The

same set of experiments can be done again, but this time with LDH present in the solution

before irradiation. If a significant difference in ROS is seen between the two groups, then LDH

might be adversely or positively affected by radiation directly, which contributes to its

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 32: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

functionality in a cellular environment. If there is no significant difference between the two

groups, then it can be concluded that LDH is not directly affected by ionizing radiation at the

doses and wavelengths used.

Another set of experiments could follow testing the effects of ionizing radiation of LDH in living

cells. These sets of experiments would involve irradiating cells and will attempt to link ionizing

radiation and generation of RCS to radioprotective effects in vivo. This would involve irradiating

cells at the same dosages as in the first set of experiments mentioned above. Assays measuring

the loss of LDH and generation of RCS would then be used to attempt to correlate the two. If

the results are positive, then the loss of LDH should correspond to the increase of RCS in the

cell. This would imply that the loss of LDH as a radioprotector might lead to the increase of RCS

in cells. In order to confirm this, a knockdown of LDH by means independent of radiation would

have to be performed to directly implicate the loss of LDH to increased RCS caused by radiation

in vivo. There are a few methods that could accomplish this, but the most direct one would

involve the use of RNAi. If this method is chosen, then LDH would be knocked down in cells by

RNAi and then subjected to radiation. The levels of LDH and RCS in the dosage groups would

have to be measured. When this experiment is complete, one could compare the numbers to

the previous experiment where RNAi-induced knockdown of LDH was not performed. If

significant disparities are seen between the groups, then it can be better concluded that LDH

plays a direct role in reducing RCS in human cells, either intrinsically or through some pathway

involving other proteins or chemicals.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 33: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

To take these experiments further, clonogenic assays should also be done to correlate this

effect to survivability. Lactate levels can also be measured in cells which would be anticipated

to go up following knockdown of LDH. The effects of this on cellular metabolism can also be

measured; membrane potentials of mitochondria can be measured using the JC-1 dye or

another stain for membrane polarization, the functionality of different complexes can be

assayed before and after, and so on. Every subsequent experiment would attempt to link the

reduction of LDH and radiation to metabolic dysfunction and contribute to identifying the

potential causes of cellular markers of CFIDS.

Alpha-synuclein and ROS

Researchers believe that the aggregation of α-syn causes ROS formation (Hsu et al. 2000;

Uversky et al. 2005; Boelens 2014). In a similar fashion, ROS production causes α-syn

aggregation (Hsu et al. 2000; Uversky et al. 2005; Riedel et al. 2011). One article suggests that

the proximity of α-syn to lipid membranes contributes to its oxidative modification upon lipid

peroxidation after irradiation, causing it to aggregate (Riedel et al. 2011). A few tests could

correlate the two. The purpose of these experiments would be to determine if α-syn can

undergo aggregation after irradiation, what mechanisms underlie this aggregation process, and

what effects are produced on the cell.

The first step would be to measure lipid peroxidation by some kind of assay after irradiation in

cells. There are several assays available to measure lipid peroxidation, with one of them being

an LDH assay. This would be done in live cells. Next, one could extract α-syn and run it on a gel

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 34: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

to determine how much was lost when compared to unirradiated controls. Next, one could

irradiate extracted α-syn in solution at different doses and run that on a PAGE gel to determine

if any significant alterations occurred. After this experiment is complete, an experiment should

be performed where membrane tissue is isolated from human cells. One could then incubate

extracted α-syn with membrane tissue, irradiate at different dosages, purify the α-syn solution,

run it on an SDS-PAGE gel, and determine if there are any differences between this gel and the

one with α-syn alone. This would tell researchers if presence of fatty tissue affects the

alteration of α-syn by radiation. This would directly link lipid peroxidation to α-syn alterations if

the results are positive. If the results are negative, the finding that radiation-induced lipid

peroxidation, radical propagation, and termination in α-syn is negligible, and alterations happen

either directly or through the presence of another protein or species that can oxidize α-syn.

After this experiment, another experiment would consider the radiation-induced alterations of

α-syn in vivo using similar methods as above. There are a few possibilities in the outcomes of

each experimental sequence. The following lists all of the possible results and conclusions

following the full sequence of experiments:

1. Incubating and irradiating α-syn together with lipids produces no significant change in

the alterations of α-syn

Conclusion: Lipid peroxidation mechanisms are negligible in radioactive alterations to

α-syn.

a. Irradiating live cells containing α-syn produced no significant change in the

alterations of α-syn compared to ex vivo trials

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 35: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Conclusion: If different than unirradiated controls, then α-syn probably

undergoes alterations caused by radiation directly. If the same as unirradiated

controls, then radiation probably has no significant effect on the alteration of

α-syn, both in vivo and ex vivo.

b. Irradiating live cells containing α-syn produced a significant change in the

alterations of α-syn compared to ex vivo trials

Conclusion: While lipid peroxidation mechanisms are negligible, some other

mechanism exists that affects the oxidation of α-syn. This probably occurs by

photosensitizer species and/or other proteins; further investigation is needed

to identify these.

2. Incubating and irradiating α-syn together with lipids produces a significant change in the

alterations of α-syn

Conclusion: Lipid peroxidation mechanisms are at play in radioactive alterations to α-

syn.

a. Irradiating live cells containing α-syn produced no significant change in the

alterations of α-syn compared to ex vivo trials

Conclusion: Lipid peroxidation is the primary pathway that is needed for

oxidative alterations of α-syn.

b. Irradiating live cells containing α-syn produced a significant change in the

alterations of α-syn compared to ex vivo trials

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 36: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Conclusion: Lipid peroxidation is one of the primary pathways that are needed

for oxidative alterations of α-syn. Other mechanisms affect the oxidation of α-

syn; further research is needed to identify these pathways.

It is important to note that the above results would not be mutually exclusive, as a dosage

threshold may have to be reached before any one of these deterministic effects are observed. It

is also important to note that, due to the findings by Saligan et al. (2013), radiation dosages can

cause elevated α-syn concentrations; this will need to be accounted for by using many controls

and measuring quantity of protein in the PAGE gels by comparing them to the concentration of

the molecular ladder. If this method is deemed too inaccurate, another assay measuring

protein expression must be done for the controls before the experiment and for the test groups

after the experiments.

Clonogenic assays would be done at each endpoint in the in vivo trials to determine if

survivability was affected. These assays would help correlate the survivability of cells with

reduced α-syn functionality in the trials. Further experimentation can be done to elucidate the

other mechanisms that might affect the radiation-induced dysfunction and aggregation of α-syn

where applicable, and implicate the effects of bystander signalling and radioprotectors in these

results in future studies.

Overall, the findings in these experiments would contribute to understanding one pathway in

which CFIDS could potentially manifest. The experiments described above would be relying on

the ideas described in Saligan et al. (2013), wherein the group described elevated fatigue

symptoms with radiation therapy and increased expression of α-syn. Considering that

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 37: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

subsequent studies will be needed to determine if this effect is relevant to CFIDS patients and

even if α-syn radiation-induced transcriptional and post-translational alterations cannot be

linked to fatigue symptoms for quite some time, these experiments will be a crucial first step in

determining the effects of radiation on α-syn from a molecular perspective.

A multitude of other experiments must be pursued in order to further understand the

involvement of these pathways in CFIDS, as these experiments are merely suggested directions

for future research. No one study will conclusively validate or disprove the connections that

these proteins have to CFIDS. Again, the purpose of all of these proposed experiments would

not be to indisputably and unassailably link any of the mentioned proteins to CFIDS, but rather

to help elucidate these pathways further and potentially give other researchers background

studies to begin making connections that could not be made before, potentially to CFIDS and

the influence of radiation.

Conclusion

Alpha-synuclein, cytochrome c, cytochrome c oxidase, lactate dehydrogenase, STAT1, and αB-

crystallin are but a few proteins potentially involved in the development of Chronic Fatigue

Syndrome. When taking into consideration the very real and exciting possibility of signal

propagation in cell systems via bystander signalling, the effects of radiation on the disease

might involve a completely novel mechanism of radiation-induced pathogenesis that could have

drastic implications in not only Chronic Fatigue Syndrome, but a number of other diseases as

well. Numerous very interesting biochemical pathways have been identified through literature

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 38: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

review that could shed further light on the mechanisms behind Chronic Fatigue Syndrome in

the context of ionizing radiation, radical chemical species formation, cellular damage, and

apoptosis.

The beauty of modern biology is that it allows scientists to investigate innumerable biochemical

pathways involved in diseases that will inevitably converge on a set of causes for the disorder.

While the proteins under investigation have a broad range of potential functions in the cell,

determining the connection between these proteins and the metabolic and cellular

characteristics of Chronic Fatigue Syndrome may provide invaluable insights into the disease.

Understanding the molecular biology and biochemistry behind a particular disorder paves the

road to patient studies, the development of targeted treatments specific to one or more

pathways, therapy options, clinical trials, and eventually a treatment. In this circumstance,

taking a broader approach by targeting processes and proteins shown to be involved in Chronic

Fatigue Syndrome as well as many cellular characteristics of disease will identify targets for

more research, and will, eventually, lead to a substantiated and comprehensive model for a

disease that has burdened too many for far too long.

Acknowledgments

The CFIDS Foundation Inc. who funded this review and in particular the director - Dr. Alan

Cocchetto who pointed the authors in the direction of many of the papers covered by the

review and provided encouragement and ideas.

Declaration of Interest

The authors declare no conflicts of interest.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 39: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Bibliography

Ahmad MF, Singh D, Taiyab A, Ramakrishna T, Raman B, Rao CM. 2008. Selective Cu2+ Binding,

Redox Silencing, and Cytoprotective Effects of the Small Heat Shock Proteins αA- and αB-

Crystallin. J Mol Biol. 382:812–824.

Ahmad SB, McNeill FE, Byun SH, Prestwich W V., Mothersill C, Seymour C, Armstrong A,

Fernandez C. 2013. Ultra-violet light emission from hpv-g cells irradiated with low let radiation

from 90Y; consequences for radiation induced bystander effects. Dose-Response. 11:498–516.

Al-Haj L, Blackshear PJ, Khabar KSA. 2012. Regulation of p21/CIP1/WAF-1 mediated cell-cycle

arrest by RNase L and tristetraprolin, and involvement of AU-rich elements. Nucleic Acids Res.

40:7739–7752.

Anderson VR, Jason L a, Hlavaty L, Porter N. 2013. A review and meta-synthesis of qualitative

studies on myalgic encephalomyelitis/chronic fatigue syndrome. Patient Educ Couns. 86:147–

155.

Andley UP, Song Z, Wawrousek EF, Fleming TP, Bassnett S. 2000. Differential protective activity

of αA-and αB-crystallin in lens epithelial cells. J. Biol. Chem. 275:36823–36831.

Andrekopoulos C, Zhang H, Joseph J, Kalivendi S, Kalyanaraman B. 2004. Bicarbonate enhances

alpha-synuclein oligomerization and nitration: intermediacy of carbonate radical anion and

nitrogen dioxide radical. Biochem J. 378:435–447.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 40: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Armeni T, Damiani E, Battino M, Greci L, Principato G. 2004. Lack of in vitro protection by a

common sunscreen ingredient on UVA-induced cytotoxicity in keratinocytes. Toxicology.

203:165–178.

Arnold DL, Radda GK, Bore PJ, Styles P, Taylor DJ. 1984. Excessive intracellular acidosis of

skeletal muscle on exercise in a patient with a post-viral exhaustion/fatigue syndrome: a 31P

nuclear magnetic resonance study. Lancet 323:1367–1369.

Azzam EI, Jay-Gerin J-P, Pain D. 2012. Ionizing radiation-induced metabolic oxidative stress and

prolonged cell injury. Cancer Lett. 327:48–60.

Badalà F, Nouri-mahdavi K, Raoof DA. 2008. Increased ventricular lactate in chronic fatigue

syndrome. III. Relationships to cortical glutathione and clinical symptoms implicate oxidative

stress in disorder pathophysiology. NMR Biomed. 144:724–732.

Banerjee S, An S, Zhou A, Silverman RH, Makino S. 2000. RNase L-independent specific 28S

rRNA cleavage in murine coronavirus-infected cells. J Virol.74:8793–802.

Barjaktarovic Z, Schmaltz D, Shyla A, Azimzadeh O, Schulz S, Haagen J, Dörr W, Sarioglu H,

Schäfer A, Atkinson MJ. 2011. Radiation–induced signaling results in mitochondrial impairment

in mouse heart at 4 weeks after exposure to X-rays. PLoS One 6:e27811.

Baris S, Alroqi F, Kiykim A, Karakoc-Aydiner E, Ogulur I, Ozen A, Charbonnier L-M, Bakır M,

Boztug K, Chatila TA. 2016. Severe early-onset combined immunodeficiency due to

heterozygous gain-of-function mutations in STAT1. J. Clin. Immunol. 36:641–648.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 41: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Bazyka D, Loganovsky K, Ilyenko I, Volovyk S, Perchuk I, Pleskach O, Nechayev S. 2010.

Psychophysiological, neuroimmune and gene expression changes in chronic fatigue syndrome

after low-dose radiation exposure. Int. J. Psychophysiol. 77:340.

Behan WMH, More IAR, Behan PO. 1991. Mitochondrial abnormalities in the postviral fatigue

syndrome. Acta Neuropathol. 83:61–65.

Bertell R. 2006. Depleted uranium: all the questions about DU and Gulf War syndrome are not

yet answered. Int. J. Heal. Serv. 36:503–520.

Boelens WC. 2014. Cell biological roles of alphaB-crystallin. Prog Biophys Mol Biol. 115:3–10.

Bogenhagen D, Clayton DA. 1977. Mouse L cell mitochondrial DNA molecules are selected

randomly for replication throughout the cell cycle. Cell 11:719–727.

Bonnekoh B, Farkas B, Geisel J, Mahrle G. 1990. Lactate dehydrogenase release as an indicator

of dithranol-induced membrane injury in cultured human keratinocytes - A time profile study.

Arch Dermatol Res. 282:325–329.

Booth NE, Myhill S, McLaren-Howard J. 2012. Mitochondrial dysfunction and the

pathophysiology of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Int. J. Clin.

Exp. Med. 5:208.

Boumezbeur F, Petersen KF, Cline GW, Mason GF, Behar KL, Shulman GI, Rothman DL,

Radiology D, Haven N. 2011. The Contribution of Blood Lactate to Brain Energy Metabolism in

Humans Measured by Dynamic 13 C Nuclear Magnetic Resonance Spectroscopy. J Neurosci.

30:13983–13991.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 42: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Braun N, Zacharias M, Peschek J, Kastenmueller A, Zou J, Hanzlik M, Weinkauf S. 2011.

Molecular architectures of the 24meric eye lens chaperone alphaB- crystallin elucidated by a

triple hybrid approach.

Broinowski A. 2013. Fukushima: life and the transnationality of radioactive contamination. Asia-

Pacific J. 11.

Brooks GA, Dubouchaud H, Brown M, Sicurello JP, Butz CE. 1999. Role of mitochondrial lactate

dehydrogenase and lactate oxidation in the intracellular lactate shuttle. Proc Natl Acad Sci U S

A. 96:1129–1134.

Brown CE, Badie B, Barish ME, Weng L, Julie R, Chang W, Naranjo A, Starr R, Wagner J, Wright C,

et al. 2016a. α-Synuclein binds TOM20 and inhibits mitochondrial protein import in Parkinson’s

disease. Sci Transl Med. 21:4062–4072.

Brown CE, Badie B, Barish ME, Weng L, Julie R, Chang W, Naranjo A, Starr R, Wagner J, Wright C,

et al. 2016b. Solid-State NMR Structure of a Pathogenic Fibril of Full-Length Human α-Synuclein.

Nat Struct Mol Biol. 21:4062–4072.

Brown GC. 2001. Regulation of mitochondrial respiration by nitric oxide inhibition of

cytochrome c oxidase. Biochim Biophys Acta - Bioenerg. 1504:46–57.

Bruinsma IB, Bruggink KA, Kinast K, Versleijen AAM, Segers-Nolten IMJ, Subramaniam V, Bea

Kuiperij H, Boelens W, de Waal RMW, Verbeek MM. 2011. Inhibition of alpha-synuclein

aggregation by small heat shock proteins. Proteins Struct Funct Bioinforma. 79:2956–2967.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 43: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Buist R. 1989. Elevated xenobiotics, lactate and pyruvate in CFS patients. J Orthomol. Med

4:170–172.

Byrne E, Trounce I. 1987. Chronic fatigue and myalgia syndrome: mitochondrial and glycolytic

studies in skeletal muscle. J Neurol Neurosurg Psychiatry. 50:743–746.

Byrne E, Trounce I, Dennett X. 1985. Chronic relapsing myalgia (? Post viral): clinical,

histological, and biochemical studies. Intern. Med. J. 15:305–308.

Canman CE, Lim DS, Cimprich KA, Taya Y, Tamai K, Sakaguchi K, Appella E, Kastan MB, Siliciano

JD, Lavin MF, et al. 1998. Activation of the ATM kinase by ionizing radiation and

phosphorylation of p53. Science. 281:1677–9.

Chakrabarti A, Jha BK, Silverman RH. 2011. New insights into the role of RNase L in innate

immunity. J Interf Cytokine Res. 31:49–57.

Chaudhry MA. 2006. Radiation‐induced gene expression profile of human cells deficient in 8‐

hydroxy‐2′‐deoxyguanine glycosylase. Int. J. cancer 118:633–642.

Chen Y, Wang X, Templeton D, Davis RJ, Tan T. 1996. The Role of c-Jun N-terminal Kinase ( JNK )

in Apoptosis Induced by Ultraviolet C and Y Radiation. J Biol Chem. 271:31929–31936.

Chis R, Sharma P, Bousette N, Miyake T, Wilson A, Backx PH, Gramolini AO. 2012. α-Crystallin B

prevents apoptosis after H 2 O 2 exposure in mouse neonatal cardiomyocytes. Am J Physiol Circ

Physiol. 303:H967–H978.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 44: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Coughlin SS, McNeil RB, Provenzale DT, Dursa EK, Thomas CM. 2013. Method issues in

epidemiological studies of medically unexplained symptom-based conditions in veterans. J. Mil.

Veterans. Health 21:4.

Dandona P, Thusu K, Cook S, Snyder B, Makowski J, Armstrong D, Nicotera T. 1996. Oxidative

damage to DNA in diabetes mellitus. Lancet. 347:444–445.

Denis M. 1986. Structure and function of cytochrome-c oxidase. Biochimie. 68:459–70.

Dimauro I, Mercatelli N, Caporossi D. 2016. Exercise-induced ROS in heat shock proteins

response. Free Radic Biol Med. 98:46–55.

Durbin JE, Fernandez-Sesma A, Lee C-K, Rao TD, Frey AB, Moran TM, Vukmanovic S, Garcia-

Sastre A, Levy DE. 2000. Type I IFN Modulates Innate and Specific Antiviral Immunity. J

Immunol. 164:4220–4228.

Durakoviæ A. 2003. Undiagnosed illnesses and radioactive warfare. Croat Med J 44:520–532.

Eustermann S, Wu WF, Langelier MF, Yang JC, Easton LE, Riccio AA, Pascal JM, Neuhaus D. 2015.

Structural Basis of Detection and Signaling of DNA Single-Strand Breaks by Human PARP-1. Mol

Cell. 60:742–754.

Evengård B, Schacterle RS, Komaroff a L. 1999. Chronic fatigue syndrome: new insights and old

ignorance. J Intern Med. 246:455–69.

Fantin VR, St-Pierre J, Leder P. 2006. Attenuation of LDH-A expression uncovers a link between

glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell. 9:425–434.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 45: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Felger JC, Cole SW, Pace TWW, Hu F, Woolwine BJ, Doho GH, Raison CL, Miller AH. 2012.

Molecular signatures of peripheral blood mononuclear cells during chronic interferon-α

treatment: relationship with depression and fatigue. Psychol. Med. 42:1591–1603.

Filler K, Lyon D, Bennett J, McCain N, Elswick R, Lukkahatai N, Saligan LN. 2014. Association of

mitochondrial dysfunction and fatigue: A review of the literature. BBA Clin. 1:12–23.

Flower TR, Chesnokova LS, Froelich CA, Dixon C, Witt SN. 2005. Heat shock prevents alpha-

synuclein-induced apoptosis in a yeast model of Parkinson’s disease. J Mol Biol. 351:1081–

1100.

Fujii N, Nakamura T, Sadakane Y, Saito T, Fujii N. 2007. Differential susceptibility of alpha A- and

alpha B-crystallin to gamma-ray irradiation. Biochim Biophys Acta - Proteins Proteomics.

1774:345–350.

Fulle S, Mecocci P, Fanó G, Vecchiet I, Vecchini A, Racciotti D, Cherubini A, Pizzigallo E, Vecchiet

L, Senin U, Beal MF. 2000. Specific oxidative alterations in vastus lateralis muscle of patients

with the diagnosis of chronic fatigue syndrome. Free Radic Biol Med. 29:1252–1259.

Gaziev AI, Shaĭkhaev GO. 2007. Ionizing radiation can activate the insertion of mitochondrial

DNA fragments in the nuclear genome. Radiatsionnaia Biol. Radioecol. 47:673–683.

Giorgio M, Migliaccio E, Orsini F, Paolucci D, Moroni M, Contursi C, Pelliccia G, Luzi L, Minucci S,

Marcaccio M, et al. 2005. Electron transfer between cytochrome c and p66Shc generates

reactive oxygen species that trigger mitochondrial apoptosis. Cell. 122:221–233.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 46: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

van Hall G, Str\omstad M, Rasmussen P, Jans O, Zaar M, Gam C, Quistorff B, Secher NH, Nielsen

HB. 2009. Blood lactate is an important energy source for the human brain. J Cereb Blood Flow

Metab. 29:1121–1129.

Hammer CT, Wills ED. 1979. The effect of ionizing radiation on the fatty acid composition of

natural fats and on lipid peroxide formation. Int J Radiat Biol Relat Stud Phys Chem Med.

35:323–332.

Hanaue N, Takeda I, Kizu Y, Tonogi M, Yamane G. 2007. Peroxynitrite formation in radiation-

induced salivary gland dysfunction in mice. Biomed Res. 28:147–151.

He Y, Yan C, Fang J, Inouye C, Tjian R, Ivanov I, Nogales E. 2016. Near-atomic resolution

visualization of human transcription promoter opening. Nature. 533:359–365.

Vander Heiden M, Cantley L, Thompson C. 2009. Understanding the Warburg effect: The

metabolic Requiremetns of cell proliferation. Science (80- ). 324:1029–1033.

Hirschhaeuser F, Sattler UGA, Mueller-Klieser W. 2011. Lactate: A metabolic key player in

cancer. Cancer Res. 71:6921–6925.

Holmes G, Kaplan J, Gantz N, Komaroff A, Schonberger B. 1988. Chronic fatigue syndrome: a

working case definition. Annu Intern Med. 108:387–389.

Hosseini M-J, Shaki F, Ghazi-Khansari M, Pourahmad J. 2014. Toxicity of Copper on Isolated

Liver Mitochondria: Impairment at Complexes I, II, and IV Leads to Increased ROS Production.

Cell Biochem Biophys. 70:367–381.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 47: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Hsu LJ, Sagara Y, Arroyo A, Rockenstein E, Sisk A, Mallory M, Wong J, Takenouchi T, Hashimoto

M, Masliah E. 2000. Alpha-Synuclein Promotes Mitochondrial Deficit and Oxidative Stress. Am J

Pathol. 157:401–10.

Hu B, Shen B, Su Y, Geard CR, Balajee AS. 2009. Protein kinase C epsilon is involved in ionizing

radiation induced bystander response in human cells. Int J Biochem Cell Biol. 41:2413–2421.

Huang Y, Wang Z, Liu Y, Xiong H, Zhao Y, Wu L, Yuan C, Wang L, Hou Y, Yu G, et al. 2016. alphaB-

crystallin interacts with Nav1.5 and regulates ubiquitination and internalization of cell surface

Nav1.5. J Biol Chem. 291:11030–11041.

Iordanov MS, Paranjape JM, Zhou a, Wong J, Williams BR, Meurs EF, Silverman RH, Magun BE.

2000. Activation of p38 mitogen-activated protein kinase and c-Jun NH(2)-terminal kinase by

double-stranded RNA and encephalomyocarditis virus: involvement of RNase L, protein kinase

R, and alternative pathways. Mol Cell Biol. 20:617–627.

Klimas NG, Salvato FR, Morgan R, Fletcher MA. 1990. Immunologic abnormalities in chronic

fatigue syndrome. J. Clin. Microbiol. 28:1403–1410.

Kim SH, Lee JM, Kim SC, Park CB, Lee PC. 2014. Proposed cytotoxic mechanisms of the saffron

carotenoids crocin and crocetin on cancer cell lines. Biochem Cell Biol. 92:105–111.

Kolappan S, Shen DL, Mosi R, Sun J, McEachern EJ, Vocadlo DJ, Craig L. 2015. Structures of

lactate dehydrogenase A (LDHA) in apo, ternary and inhibitor-bound forms. Acta Crystallogr

Sect D Biol Crystallogr. 71:185–195.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 48: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Kryston TB, Georgiev AB, Pissis P, Georgakilas AG. 2011. Role of oxidative stress and DNA

damage in human carcinogenesis. Mutat Res - Fundam Mol Mech Mutagen. 711:193–201.

Kumerova AO, Lece AG, Skesters AP, Orlikov GA, Seleznev J V, Rainsford KD. 2000. Antioxidant

defense and trace element imbalance in patients with postradiation syndrome. Biol. Trace

Elem. Res. 77:1–12.

Kuratsune H, Yamaguti K, Takahashi M, Misaki H, Tagawa S, Kitani T. 1994. Acylcarnitine

deficiency in chronic fatigue syndrome. Clin. Infect. Dis. 18:S62–S67.

Kurup RK, Kurup PA. 2003. Hypothalamic Digoxin, Cerebral Chemical Dominance and Myalgic

Encephalomyelitis. Int J Neurosci. 113:683–701.

Lane RJM, Barrett MC, Taylor DJ, Kemp GJ, Lodi R. 1998. Heterogeneity in chronic fatigue

syndrome: evidence from magnetic resonance spectroscopy of muscle. Neuromuscul. Disord.

8:204–209.

Le M, Fernandez-Palomo C, McNeill FE, Seymour CB, Rainbow AJ, Mothersill CE. 2017.

Exosomes are released by bystander cells exposed to radiation-induced biophoton signals:

Reconciling the mechanisms mediating the bystander effect. PLoS One. 12:e0173685.

Le M, McNeill FE, Seymour C, Rainbow AJ, Mothersill CE. 2015. An observed effect of ultraviolet

radiation emitted from beta-irradiated HaCaT cells upon non-beta-irradiated bystander cells.

Radiat Res. 183:279–90.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 49: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Le M, Mothersill CE, Seymour CB, Ahmad SB, Armstrong A, Rainbow AJ, McNeill FE. 2015.

Factors affecting ultraviolet-A photon emission from beta-irradiated human keratinocyte cells.

Phys Med Biol. 60:6371–6389.

Leach JK, Van Tuyle G, Lin P-S, Schmidt-Ullrich R, Mikkelsen RB. 2001. Ionizing radiation-

induced, mitochondria-dependent generation of reactive oxygen/nitrogen. Cancer Res.

61:3894–3901.

Lemire J, Mailloux RJ, Appanna VD. 2008. Mitochondrial lactate dehydrogenase is involved in

oxidative-energy metabolism in human astrocytoma cells (CCF-STTG1). PLoS One. 3:1–10.

Letts JA, Fiedorczuk K, Sazanov LA. 2016. The architecture of respiratory supercomplexes.

Naturet. 537:644–648.

Liao E-C, Hsu Y-T, Chuah Q-Y, Lee Y-J, Hu J-Y, Huang T-C, Yang P-M, Chiu S-J. 2014. Radiation

induces senescence and a bystander effect through metabolic alterations. Cell Death Dis.

5:e1255.

Liao JH, Lee JS, Chiou SH. 2002. Distinct roles of αA- and αB-crystallins under thermal and UV

stresses. Biochem Biophys Res Commun. 295:854–861.

Limoli CL, Ponnaiya B, Corcoran JJ, Giedzinski E, Kaplan MI, Hartmann A, Morgan WF. 2000.

Genomic instability induced by high and low LET ionizing radiation. Adv. Sp. Res. 25:2107–2117.

Lin MT, Beal MF. 2006. Mitochondrial dysfunction and oxidative stress in neurodegenerative

diseases. Nature. 443:787–795.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 50: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Liu Z, Prestwich, WV, Stewart RD, Byun SH, Mothersill CE, McNeill FE. 2007. Effective target size

for the induction of bystander effects in medium transfer experiments. Radiat Res. 168:627-

630.

Liu S, Li J, Tao Y, Xiao X. 2007. Small heat shock protein ??B-crystallin binds to p53 to sequester

its translocation to mitochondria during hydrogen peroxide-induced apoptosis. Biochem

Biophys Res Commun. 354:109–114.

Liu S, Yan B, Lai W, Chen L, Xiao D, Xi S, Jiang Y, Dong X, An J, Chen X, et al. 2014. As a novel p53

direct target, bidirectional gene HspB2/alphaB-crystallin regulates the ROS level and Warburg

effect. Biochim Biophys Acta - Gene Regul Mech. 1839:592–603.

Loganovsky K. 2009. Do low doses of ionizing radiation affect the human brain? Data Sci. J.

8:BR13-BR35.

Loganovsky K, Perchuk I, Marazziti D. 2016. Workers on transformation of the shelter object of

the Chernobyl nuclear power plant into an ecologically-safe system show qEEG abnormalities

and cognitive dysfunctions: A follow-up study. World J. Biol. Psychiatry 17:600–607.

Loganovsky KN. 2000. Vegetative-vascular dystonia and osteoalgetic syndrome or Chronic

Fatigue Syndrome as a characteristic after-effect of radioecological disaster: the Chernobyl

accident experience. J. Chronic Fatigue Syndr. 7:3–16.

Lorimore SA, Chrystal JA, Robinson JI, Coates PJ, Wright EG. 2008. Chromosomal instability in

unirradiated hemaopoietic cells induced by macrophages exposed in vivo to ionizing radiation.

Cancer Res. 68:8122–8126.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 51: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Mao X, Ren Z, Parker GN, Sondermann H, Pastorello MA, Wang W, McMurray JS, Demeler B,

Darnell JE, Chen X. 2005. Structural bases of unphosphorylated STAT1 association and receptor

binding. Mol Cell. 17:761–771.

Marchi S, Giorgi C, Suski JM, Agnoletto C, Bononi A, Bonora M, De Marchi E, Missiroli S,

Patergnani S, Poletti F, et al. 2012. Mitochondria-Ros Crosstalk in the Control of Cell Death and

Aging. J Signal Transduct. 2012:1–17.

Martin OA, Redon CE, Nakamura AJ, Dickey JS, Georgakilas AG, Bonner WM. 2011. Systemic

DNA damage related to cancer. Cancer Res. 71:3437–3441.

Mazière C, Dantin F, Dubois F, Santus R, Mazière JC. 2000. Biphasic effect of UVA radiation on

STAT1 activity and tyrosine phosphorylation in cultured human keratinocytes. Free Radic Biol

Med. 28:1430–1437.

McCauley LA, Joos SK, Barkhuizen A, Shuell T, Tyree WA, Bourdette DN. 2002. Chronic fatigue in

a population-based study of Gulf War veterans. Arch. Environ. Heal. An Int. J. 57:340–348.

McCully KK, Natelson BH, Iotti S, Sisto S, Leigh JS. 1996. Reduced oxidative muscle metabolism

in chronic fatigue syndrome. Muscle Nerve 19:621–625.

Meeus M, Nijs J, McGregor N, Meeusen R, De Schutter G, Truijen S, Frémont M, Van Hoof E, De

Meirleir K. 2008. Unravelling intracellular immune dysfunctions in chronic fatigue syndrome:

Interactions between protein kinase R activity, RNase L cleavage and elastase activity, and their

clinical relevance. In Vivo (Brooklyn). 22:115–122.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 52: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

De Meirleir K, Bisbal C, Campine I, De Becker P, Salehzada T, Demettre E, Lebleu B. 2000. A 37

kDa 2-5A binding protein as a potential biochemical marker for chronic fatigue syndrome. Am J

Med. 108:99–105.

Milano F. 2000. Gulf War Syndrome: the ’Agent Orange" of the nineties. Int. Soc. Sci. Rev.

75:16–25.

Mitra AK, Bhat N, Sarma A, Krishna M. 2005. Alteration in the expression of signaling

parameters following carbon ion irradiation. Mol. Cell. Biochem. 276:169–173.

Mohan N, Meltz ML. 1994. Induction of nuclear factor κB after low-dose ionizing radiation

involves a reactive oxygen intermediate signaling pathway. Radiat. Res. 140:97–104.

Moreno-Beltran B, Conte RD, Diaz-Quintana A, Rosa MD, P T, Diaz-Moreno I. 2016. NMR

structure of the Y48pCMF variant of human cytochrome c in its reduced state.

Morgan WF. 2003. Non-targeted and delayed effects of exposure to ionizing radiation: I.

Radiation-induced genomic instability and bystander effects in vitro. Radiat. Res. 159:567–580.

Morris G, Maes M. 2012. Increased nuclear factor-κB and loss of p53 are key mechanisms in

Myalgic Encephalomyelitis/chronic fatigue syndrome (ME/CFS). Med Hypotheses. 79:607–613.

Mothersill C, Seymour CB. 1998. Cell-cell contact during gamma irradiation is not required to

induce a bystander effect in normal human keratinocytes: Evidence for release during

irradiation of a signal controlling survival into the medium. Radiat. Res. 149:256–262.

Mothersill C, Seymour C. 1997. Medium from irradiated human epithelial cells but not human

fibroblasts reduces the clonogenic survival of unirradiated cells. Int. J. Radiat. Biol. 71:421–427.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 53: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Mothersill C. O’Reilly, D. Papworth, SJ Marsden, CB Seymour, EG Wright, C MAK. 2000. Dose–

and time–response relationships for lethal mutations and chromosomal instability induced by

ionizing radiation in an immortalized human keratinocyte cell line. Int. J. Radiat. Biol. 76:799–

806.

Mothersill C, Seymour C. 2016. Fundamental Mechanisms Underlying the Ill Health and Chronic

Fatigue Syndrome Suffered by Atomic and Gulf War Veterans: A Unifying Hypothesis. In:

Genetics, Evolution and Radiation. Springer. p. 347–356.

Mothersill C, Rusin A, Seymour C. 2017a. Low doses and non-targeted effects in environmental

radiation protection; where are we now and where should we go? Environ. Res. 159:484–490.

Mothersill CE, Rusin A, Fernandez-Palomo C, Seymour CB. 2017b. History of Bystander Effects

Research 1905-present; What’s in a name? Int. J. Radiat. Biol. 1–44. [accepted online].

Mulero P, Almansa R, Neri MJ, Bermejo-Martin JF, Archanco M, Arenillas JF, Téllez N. 2015.

Improvement of fatigue in multiple sclerosis by physical exercise is associated to modulation of

systemic interferon response. J. Neuroimmunol. 280:8–11.

Mullan PB, Hosey AM, Buckley NE, Quinn JE, Kennedy RD, Johnston PG, Harkin DP. 2005. The

2,5 oligoadenylate synthetase/RNaseL pathway is a novel effector of BRCA1- and interferon-γ-

mediated apoptosis. Oncogene. 24:5492–5501.

Myhill S, Booth NE, McLaren-Howard J. 2009. Chronic fatigue syndrome and mitochondrial

dysfunction. Int. J. Clin. Exp. Med. 2:1.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 54: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Naviaux RK, Naviaux JC, Li K, Bright AT, Alaynick WA, Wang L, Baxter A, Nathan N, Anderson W,

Gordon E. 2016. Metabolic features of chronic fatigue syndrome. Proc Natl Acad Sci U S A.

113:1607571113-.

Nagasawa H, Little JB. 1992. Induction of Sister Chromatid Exchanges by Extremely Low Doses

of α-Particles. Cancer Res. 52:6394 LP-6396.

Nicolson GL. 2003. Lipid replacement as an adjunct to therapy for chronic fatigue, anti-aging

and restoration of mitochondrial function. J Am Nutraceut Assoc 6:22–28.

Nicolson GL, Ellithorpe R. 2006. Lipid replacement and antioxidant nutritional therapy for

restoring mitochondrial function and reducing fatigue in chronic fatigue syndrome and other

fatiguing illnesses. J. Chronic Fatigue Syndr. 13:57–68.

Nugent S, Mothersill CE, Seymour C, McClean B, Lyng FM, Murphy JEJ. 2010. Altered

mitochondrial function and genome frequency post exposure to gamma-radiation and

bystander factors. Int. J. Radiat. Biol. 86:829–41. doi:10.3109/09553002.2010.486019.

Pall ML. 2007. Nitric oxide synthase partial uncoupling as a key switching mechanism for the

NO/ONOO–cycle. Med. Hypotheses 69:821–825.

Pall ML. 2008. Post-radiation syndrome as a NO/ONOO–cycle, chronic fatigue syndrome-like

disease. Med. Hypotheses 71:537–541.

Pall ML. 2009. The NO/ONOO-vicious cycle mechanism as the cause of chronic fatigue

syndrome/myalgic encephalomyelitis. New Res. chronic fatigue Syndr. New York Nov. Biomed.

Publ.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 55: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Pall ML, Satterlee JD. 2001. Elevated nitric oxide/peroxynitrite mechanism for the common

etiology of multiple chemical sensitivity, chronic fatigue syndrome, and posttraumatic stress

disorder. Ann. N. Y. Acad. Sci. 933:323–329.

Pandey BN, Gordon DM, De Toledo SM, Pain D, Azzam EI. 2006. Normal human fibroblasts

exposed to high-or low-dose ionizing radiation: differential effects on mitochondrial protein

import and membrane potential. Antioxid. Redox Signal. 8:1253–1261.

Panieri E, Santoro M. 2016. ROS homeostasis and metabolism: a dangerous liason in cancer

cells. Cell Death Dis. 7:1–12.

Parcellier A, Schmitt E, Brunet M, Hammann A, Solary E, Garrido C. 2005. Small heat shock

proteins HSP27 and αB-crystallin: cytoprotective and oncogenic functions. Antioxid. Redox

Signal. 7:404–413.

Passarella S, Paventi G, Pizzuto R. 2014. The mitochondrial L-lactate dehydrogenase affair. Front

Neurosci. 8:1–4.

Patarca R. 2001. Cytokines and chronic fatigue syndrome. Ann. N. Y. Acad. Sci. 933:185–200.

Pastel RH. 2002. Radiophobia: long-term psychological consequences of Chernobyl. Mil. Med.

167:134–136.

Perry AJ, Hulett JM, Likić VA, Lithgow T, Gooley PR. 2006. Convergent evolution of receptors for

protein import into mitochondria. Curr Biol. 16:221–229.

Pflugbeil S, Paulitz H, Claussen A, Schmitz-Fuerhake I. 2006. Health Effects of Chernobyl: 20

Years after the Reactor Catastrophe. Meta Anal. IPPNW.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 56: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Pitroda SP, Wakim BT, Sood RF, Beveridge MG, Beckett MA, MacDermed DM, Weichselbaum

RR, Khodarev NN. 2009. STAT1-dependent expression of energy metabolic pathways links

tumour growth and radioresistance to the Warburg effect. BMC Med. 7:1-10.

Plioplys A V, Plioplys S. 1995. Serum levels of carnitine in chronic fatigue syndrome: clinical

correlates. Neuropsychobiology 32:132–138.

Prasad A V, Mohan N, Chandrasekar B, Meltz ML. 1994. Activation of nuclear factor κB in

human lymphoblastoid cells by low-dose ionizing radiation. Radiat. Res. 138:367–372.

Rasmussen HN, van Hall G, Rasmussen UF. 2002. Lactate dehydrogenase is not a mitochondrial

enzyme in human and mouse vastus lateralis muscle. J Physiol. 541:575–80.

Rithidech KN, Tungjai M, Arbab E, Simon SR. 2005. Activation of NF-κB in bone marrow cells of

BALB/cJ mice following exposure in vivo to low doses of 137Cs γ-rays. Radiat. Environ. Biophys.

44:139–143.

Riedel M, Goldbaum O, Richter-Landsberg C. 2009. α-Synuclein promotes the recruitment of

tau to protein inclusions in oligodendroglial cells: Effects of oxidative and proteolytic stress. J

Mol Neurosci. 39:226–234.

Riedel M, Goldbaum O, Wille M, Richter-Landsberg C. 2011. Membrane lipid modification by

docosahexaenoic acid (DHA) promotes the formation of α-synuclein inclusion bodies

immunopositive for SUMO-1 in oligodendroglial cells after oxidative stress. J Mol Neurosci.

43:290–302.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 57: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Saligan LN, Hsiao CP, Wang D, Wang XM, John LS, Kaushal A, Citrin D, Barb JJ, Munson PJ,

Dionne RA. 2013. Upregulation of α-synuclein during localized radiation therapy signals the

association of cancer-related fatigue with the activation of inflammatory and neuroprotective

pathways. Brain Behav Immun. 27:63–70.

Schettino G, Folkard M, Michael BD, Prise KM. 2005. Low-dose binary behavior of bystander cell

killing after microbeam irradiation of a single cell with focused CK X rays. Radiat. Res. 163:332–

336.

Shi X, Seymour C, Mothersill C. 2016. The effects of chronic, low doses of Ra-226 on cultured

fish and human cells. Environ. Res. 148:303–309. doi:10.1016/j.envres.2016.04.009.

Silverman RH. 2007. Viral encounters with 2’,5’-oligoadenylate synthetase and RNase L during

the interferon antiviral response. J Virol. 81:12720–12729.

Silverman RH. 2007. A scientific journey through the 2-5A/RNase L system. Cytokine Growth

Factor Rev. 18:381–388.

Smith WW, Jiang H, Pei Z, Tanaka Y, Morita H, Sawa A, Dawson VL, Dawson TM, Ross CA. 2005.

Endoplasmic reticulum stress and mitochondrial cell death pathways mediate A53T mutant

alpha-synuclein-induced toxicity. Hum. Mol. Genet. 14:3801–3811.

Smith RW, Wang J, Bucking CP, Mothersill CE, Seymour CB. 2007. Evidence for a protective

response by the gill proteome of rainbow trout exposed to X-ray induced bystander signals.

Proteomics 7:4171–4180. doi:10.1002/pmic.200700573.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 58: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Smith RW, Seymour CB, Moccia RD, Mothersill CE. 2016. Irradiation of rainbow trout at early

life stages results in trans-generational effects including the induction of a bystander effect in

non-irradiated fish. Environ. Res. 145:26–38. doi:10.1016/j.envres.2015.11.019.

Spitz DR, Azzam EI, Li JJ, Gius D. 2004. Metabolic oxidation/reduction reactions and cellular

responses to ionizing radiation: a unifying concept in stress response biology. Cancer Metastasis

Rev. 23:311–322.

Stark GR, Kerr IM, Williams BRG, Silverman RH, Schreiber RD. 1998. How Cells Respond To

Interferons. Annu Rev Biochem. 67:227–264.

Suhadolnik RJ, Peterson DL, Brien KO, Cheney PR, Herst CVT, Reichenbach NL, Kon N, Horvath

SE, Iacono KT, Adelson ME, et al. 1997. Biochemical Evidence for a Novel Low Molecular Weight

2-5A-Dependent RNase L in Chronic Fatigue Syndrome CHRONi. J Interf Cytokine Res. 385:377–

385.

Szumiel I. 2015. Ionizing radiation-induced oxidative stress, epigenetic changes and genomic

instability: the pivotal role of mitochondria. Int. J. Radiat. Biol. 91:1–12.

Tada-Oikawa S, Oikawa S, Kawanishi S. 2000. Determination of DNA damage, peroxide

generation, mitochondrial membrane potential, and caspase-3 activity during ultraviolet A-

induced apoptosis summary. Methods Enzymol Singlet Oxyg UV-A, Ozone.:331–342.

Thomas MP, Chartrand K, Reynolds A, Vitvitsky V, Banerjee R, Gendelman HE. 2007. Ion channel

blockade attenuates aggregated alpha synuclein induction of microglial reactive oxygen species:

Relevance for the pathogenesis of Parkinson’s disease. J Neurochem. 100:503–519.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 59: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Tirelli U, Maotta G, Improta S, Pinto A. 1994. Immunological abnormalities in patients with

chronic fatigue syndrome. Scand J Immunol. 40:601–608.

Ulmer TS, Bax A, Cole NB, Nussbaum RL. 2005. Structure and Dynamics of Micelle-bound

Human -Synuclein. J Biol Chem. 280:9595–9603.

Uversky VN, Yamin G, Munishkina LA, Karymov MA, Millett IS, Doniach S, Lyubchenko YL, Fink

AL. 2005. Effects of nitration on the structure and aggregation of ??-synuclein. Mol Brain Res.

134:84–102.

Vainer R, Cohen S, Shahar A, Arivach R, Arbely E. 2016. Structural Basis for p53 Lys120-

Acetylation-Dependent DNA-Binding Mode. J Mol Biol. 428:3013–3025.

Vecchiet L, Montanari G, Pizzigallo E, Iezzi S, de Bigontina P, Dragani L, Vecchiet J,

Giamberardino MA. 1996. Sensory characterization of somatic parietal tissues in humans with

chronic fatigue syndrome. Neurosci. Lett. 208:117–120.

Visser J, Blauw B, Hinloopen B, Brommer E, Kloet ER de, Kluft C, Nagelkerken L. 1998. CD4 T

lymphocytes from patients with chronic fatigue syndrome have decreased interferon-γ

production and increased sensitivity to dexamethasone. J. Infect. Dis. 177:451–454.

Wawrzyniak NR, Joseph A, Levin DG, David M, Leeuwenburgh C, Sandesara B, Todd M. 2016.

Idiopathic chronic fatigue in older adults is linked to impaired mitochondrial content and

biogenesis signaling in skeletal muscle. 7.

Whiteside TL, Friberg D. 1998. Natural killer cells and natural killer cell activity in chronic fatigue

syndrome. Am J Med. 105:27S–34S.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 60: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Wilkes M, Madej MG, Kreuter L, Rhinow D, Heinz V, De Sanctis S, Ruppel S, Richter RM, Joos F,

Grieben M, et al. 2017. Molecular insights into lipid-assisted Ca2+ regulation of the TRP channel

Polycystin-2. Nat Struct Mol Biol. 24:123–130.

Wong R, Lopaschuk G, Zhu G, Walker D, Catellier D, Burton D, Teo K, Collins-Nakai R, Montague

T. 1992. Skeletal muscle metabolism in the chronic fatigue syndrome: in vivo assessment by 31P

nuclear magnetic resonance spectroscopy. Chest 102:1716–1722.

Wright EG. 1998. Radiation-induced genomic instability in haemopoietic cells. Int. J. Radiat. Biol.

74:681–687.

Wyss MT, Jolivet R, Buck A, Magistretti PJ, Weber B. 2011. In vivo evidence for lactate as a

neuronal energy source. J Neurosci. 31:7477–7485.

Xu F, Yu H, Liu J, Cheng L. 2013. αb-crystallin regulates oxidative stress-induced apoptosis in

cardiac H9c2 cells via the PI3K/AKT pathway. Mol Biol Rep. 40:2517–2526.

Yang J. 1997. Prevention of Apoptosis by Bcl-2: Release of Cytochrome c from Mitochondria

Blocked. Science. 275:1129-1132.

Yin E, Nelson DO, Coleman MA, Peterson LE, Wyrobek AJ. 2003. Gene expression changes in

mouse brain after exposure to low‐dose ionizing radiation. Int. J. Radiat. Biol. 79:759–775.

Yoshikawa S, Yano MN, Baba A S, Mochizuki M, T T. 2016. The Mg2 -containing water cluster of

mammalian cytochrome c oxidase collects four pumping proton equivalents in each catalytic

cycle. BBA Bioenerg. 1857.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 61: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Zhang C, Baumer A, Mackay IR, Linnane AW, Nagley P. 1995. Unusual pattern of mitochondrial

DNA deletions in skeletal muscle of an adult human with chronic fatigue syndrome. Hum. Mol.

Genet. 4:751–754.

Zorov DB, Juhaszova M, Sollott SJ. 2014. Mitochondrial Reactive Oxygen Species (ROS) and ROS-

Induced ROS Release. Physiol Rev. 94:909–950.

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 62: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 63: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018

Page 64: Chronic Fatigue and Immune Deficiency Syndrome (CFIDS

Accep

ted

Man

uscr

ipt

Dow

nloa

ded

by [

UN

IVE

RSI

TY

OF

AD

EL

AID

E L

IBR

AR

IES]

at 0

7:03

04

Janu

ary

2018