ArticlePDF Available

Abstract and Figures

Background: despite major advances in immunology and molecular biology, Multiple Sclerosis is poorly understood with regards to etiology and its immune trigger and causal pathways are substantially unknown. In recent years, vascular abnormalities associated with Multiple Sclerosis have been investigated, opening the doors to the hypothesis that in the pathogenesis of multiple sclerosis a vascular component could be important. Different forms of vascular abnormalities have been associated with MS: increased risk for ischemic disease, cerebral hypoperfusion, abnormalities of endothelial cells and impaired venous drainage. The aim of this review was to describe literature evidence concerning the correlation between Multiple Sclerosis and vascular dysfunction, cardiovascular risk factors and major cardiovascular events. Methods: A literature review was performed using the following databases and web search engines: PubMed –US National Library of Medicine; Google Scholar; and Ovid MEDLINE. The search included the following combination of terms: “multiple sclerosis” and endothelial dysfunction or vascular dysregulation or vascular hypothesis or risk factors or cardiovascular disease or venous thromboembolism or epidemiology. The selected articles were divided into six macro-groups according to the topic of the paper: cardiovascular risk; cardiovascular diseases; microcirculation factors; venous alterations; infectious pathogens; and vascular adverse effects of therapy. Results: patients with Multiple Sclerosis seem to have more cardiovascular risk factors and an increased risk for ischemic stroke. Several studies have demonstrated cerebral perfusion abnormalities. The relationship between the disturbances in cerebral venous outflow and neurological disorders remains an open issue that requires further studies. Conclusion: Recent evidence suggests that vascular components may be initiating triggers for neuronal pathology and subsequent neurological manifestations of the disease. The high degree of comorbidity between vascular disease and Multiple Sclerosis suggests that vascular pathology may be an important factor causing neuronal dysfunction or degeneration in multiple sclerosis.
Content may be subject to copyright.
Open Access
Review Article
Journal of Vascular
Medicine & Surgery
J
o
u
r
n
a
l
o
f
V
a
s
c
u
l
a
r
M
e
d
i
c
i
n
e
&
S
u
r
g
e
r
y
ISSN: 2329-6925
Caprio et al., Vasc Med Surg 2016, 4:2
http://dx.doi.org/10.4172/2329-6925.1000259
Volume 4 • Issue 2 • 1000259
J Vasc Med Surg
ISSN: 2329-6925 JVMS, an open access journal
*Corresponding author: Mancini M, Institute of Biostructures and Bioimaging,
National Research Council, via Tommaso De Amicis 95 -80145 Naples, Italy, Tel:
+39 081 2203411; Fax: +39 081 2203498; E-mail: direttore@ibb.cnr.it
Received February 23, 2016; Accepted March 04, 2016; Published March 11,
2016
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016)
Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med Surg
4: 259. doi:10.4172/2329-6925.1000259
Copyright: © 2016 Caprio MG, et al. This is an open-access article distributed
under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the
original author and source are credited.
Vascular Disease in Patients with Multiple Sclerosis: A Review
Caprio MG1, Russo C2, Giugliano A2, Ragucci M2 and Mancini M1*
1Institute of Biostructures and Bioimaging, National Research Council, Italy
2Department of Advanced Biomedical Sciences, University “Federico II”, Italy
Abstract
Background: despite major advances in immunology and molecular biology, Multiple Sclerosis is poorly
understood with regards to etiology and its immune trigger and causal pathways are substantially unknown. In recent
years, vascular abnormalities associated with Multiple Sclerosis have been investigated, opening the doors to the
hypothesis that in the pathogenesis of multiple sclerosis a vascular component could be important. Different forms of
vascular abnormalities have been associated with MS: increased risk for ischemic disease, cerebral hypoperfusion,
abnormalities of endothelial cells and impaired venous drainage. The aim of this review was to describe literature
evidence concerning the correlation between Multiple Sclerosis and vascular dysfunction, cardiovascular risk factors
and major cardiovascular events.
Methods: A literature review was performed using the following databases and web search engines: PubMed –
US National Library of Medicine; Google Scholar; and Ovid MEDLINE. The search included the following combination
of terms: “multiple sclerosis” and endothelial dysfunction or vascular dysregulation or vascular hypothesis or
risk factors or cardiovascular disease or venous thromboembolism or epidemiology. The selected articles were
divided into six macro-groups according to the topic of the paper: cardiovascular risk; cardiovascular diseases;
microcirculation factors; venous alterations; infectious pathogens; and vascular adverse effects of therapy.
Results: patients with Multiple Sclerosis seem to have more cardiovascular risk factors and an increased risk
for ischemic stroke. Several studies have demonstrated cerebral perfusion abnormalities. The relationship between
the disturbances in cerebral venous outow and neurological disorders remains an open issue that requires further
studies.
Conclusion: Recent evidence suggests that vascular components may be initiating triggers for neuronal
pathology and subsequent neurological manifestations of the disease. The high degree of comorbidity between
vascular disease and Multiple Sclerosis suggests that vascular pathology may be an important factor causing
neuronal dysfunction or degeneration in multiple sclerosis.
Keywords: Multiple sclerosis; Cardiovascular risk factors; Ischemic
stroke; Vascular abnormalities; Cerebral venous outow; Review
Introduction
Multiple sclerosis (MS) is a neurodegenerative disorder that
aects the central nervous system (CNS), sparing the peripheral
nervous system and revealing its rst signs in early adulthood, with a
variable clinical course ranging from a benign condition to a rapidly
evolving disabling disease [1]. MS is the most common disease of the
CNS and the most common cause of neurological disability in young
adults, aecting approximately 2.5 million worldwide, with variable
progression and prognosis [1,2]. e incidence and prevalence rates
of MS vary considerably between regions and populations, showing
a typical latitudinal gradient probably due to genetic and behavioral
variations [3]. Europe is considered a high frequency area for MS
(prevalence ≥ 30/100,000); other high prevalence regions include
the northern USA, Israel, Canada, Southern Australia, New Zealand
and Eastern Russia [4]. Chronic inammation, perivenular cung,
demyelization, gliosis and neuronal loss are hallmarks of the pathology,
producing plaque formation and tissue destruction not only in the
white matter but also in the cerebral cortex. Despite major advances
in immunology and molecular biology, MS is poorly understood with
regards to etiology and its immune trigger and causal pathways are
substantially unknown. Moreover, how etiopathological mechanisms
inuence the course of this disease remains unclear [5-8]. Studies
with magnetic resonance imaging (MRI) have demonstrated that
white matter lesions correlate weakly with neurological disability and
longitudinal studies involving MS patients have shown accelerated
grey matter atrophy as consequence of grey matter lesions. Grey
matter (GM) atrophy correlates with physical and cognitive disability
more strongly than white matter atrophy. Many theories have been
developed, and many potential causes have been identied: genetic
predisposition, environmental factors, infections, vascular risk factors,
and traumatic brain injury. Moreover, it is not known why some
patients develop clinically asymptomatic MS with a benign course,
while other patients experience relapses with permanent neurological
decits or progressive and massive disability. Vascular pathology in
MS patients was investigated by authors and noted by Charcot in his
identication of MS; however, vascular abnormalities associated with
MS have been investigated only in recent years, opening the doors to
the hypothesis that in the pathogenesis of multiple sclerosis a vascular
component could be important [1,9-11]. Assuming that endothelial
dysfunction and chronic inammation play an integral role in CNS
lesions pathogenesis, many authors have decided to elucidate the
primary and secondary eects of these mechanisms regarding the
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016) Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med
Surg 4: 259. doi:10.4172/2329-6925.1000259
Page 2 of 12
Volume 4 • Issue 2 • 1000259
J Vasc Med Surg
ISSN: 2329-6925 JVMS, an open access journal
classical and non-classical cardiovascular risk factors and their
relationship with MS will be considered separately.
Obesity: Observational studies analyzing the prevalence of
overweight and obesity in MS in comparison with the general
population are inconclusive. Among MS patients, the combined
prevalence of overweight (Body mass index (BMI) 25 kg/m2) and
obesity (BMI 30 kg/m2) is approximately between 50% and 65% in
large sample sizes. ese estimates are similar to those of the non-
diseased adult population. Some studies in childhood and adolescence
showed an association between BMI and the risk of developing multiple
sclerosis [16]. Instead this association was not found in adults. ese
data suggest that the prevention of obesity in adolescence may reduce
the risk of MS [17,18].
In other studies a lower prevalence of obesity in MS patients
compared to controls has been reported [19,20]. In contrast, the
prevalence of metabolic syndrome seems similar to that of the general
population and waist circumference seems to increase among disabled
MS patients. e paradox of the discrepancy between the prevalence
of high BMI and metabolic syndrome can be explained by the lower
ratio of muscle to fat in disabled patients. erefore, the use of waist
circumference, instead of BMI, as a cardiovascular risk factor in these
patients could be recommended.
e association between BMI and disability was examined in four
studies. In two of these studies, the BMI, was calculated using weight
and height self-reported. us represents a potential limitation because
overweight patients frequently to under-report their weight [17].
Only one study clearly demonstrated a correlation between BMI
and disability [21]. is study showed a correlation of BMI and
almost all the determinants of serum lipids and Apo lipoproteins with
disability as assessed by Expanded Disability Status Scale (EDSS). Aer
adjusting the analysis for BMI and physical activity the magnitude of
BMI and disability persisted.
Levels of vitamin D: e importance of vitamin D as a risk factor
of MS has been demonstrated in several studies [22-24]. One study
showed that women who took vitamin D supplements had a 40% lower
risk of developing MS [25]. Recent studies also describe a relationship
between low levels of vitamin D and relapses and disease progression
[26]. Moreover, low vitamin D levels are correlated with both MS
and cardiovascular disease [27,28]. e studies are not yet conclusive;
however, a growing body of evidence supports a protective role for
vitamin D in MS development.
vasculature in MS patients compared to that in the healthy population.
Fundamentally, dierent forms of vascular abnormalities have been
associated with MS: increased risk for ischemic disease, arterial cerebral
hypoperfusion, abnormalities of endothelial cells and impaired venous
drainage. e aim of this review is to describe literature evidence
concerning the relationship between MS and vascular dysfunction,
cardiovascular risk factors and major cardiovascular events.
Materials and Methods
A literature review was performed using the following databases
and web search engines: PubMed – US National Library of Medicine;
Google Scholar; and Ovid MEDLINE. A text search was also
performed: “e epidemiology of Neurological Disorders”, Compston
et al. BMJ Books, London, 1998; “Principles of Neurology”, Chapter
36, Multiple Sclerosis and Allied Demyelinating Diseases, Adams
et al., McGraw-Hill, 1997; “Neurological disorders: public health
challenges”, World Health Organization 2006, WHO Library; and
“McAlpine’s Multiple Sclerosis” - Fourth Edition, Alastair Compston,
Churchill Livingstone Elsevier, 2006. In addition, a manual search
of reference lists from original articles was performed. e search
included the following combination of terms: “multiple sclerosis” and
“endothelial dysfunction” or “vascular dysregulation” or “vascular
hypothesis” or “risk factors” or “cardiovascular disease” or “venous
thromboembolism” or “epidemiology”. We considered articles and
publications from 1830 to 2015; the research was limited to texts and
articles published in English or with available translation into English.
We referenced primary sources, using secondary papers exclusively
when the primary sources were not available. Our initial search yielded
3842 articles whose abstracts were reviewed; the selection of the articles
was performed based on the agreement between the authors MGC, CR,
AG and MM. Of the 3842, we excluded dierent studies from the same
research group concerning similar ndings or overlapping populations
with redundant collected data. We also excluded case-reports or
studies with a small sample population size (fewer than 20 patients).
At the end of the selection procedure, 3643 articles had been excluded.
e selected articles were divided into six macro-groups according
to the topic of the paper: cardiovascular risk; cardiovascular diseases;
microcirculation factors in MS; venous alterations; MS and infectious
pathogens; and vascular adverse eects of MS therapy (Figure 1) and
(Table 1 as supplementary material). All the articles mentioned in this
review are included in the nal reference list.
Results
Cardiovascular risk
Many authors have discussed the association between MS and
classical/non-classical cardiovascular (CV) risk factors, analyzing this
issue from dierent points of view and trying to elucidate the impact
of these factors on the underlying disease [10-13]. e most salient
evidence from the literature is the higher prevalence of traditional
risk factors in patients with MS and the change in clinical outcomes
produced by the presence of one or more CV risk factors. e prevalence
increases with age and is higher in man. It was also shown an increased
risk of ischemic heart disease, ischemic stroke and peripheral vascular
disease [14].
e simultaneous presence of individual or multiple CV risk
factors is related to the increased accumulation of white matter
lesions, decreased grey matter and cortical volume and consequential
accelerated brain atrophy [15]. In the subsections below, the various
Figure 1: This owchart describes the systematic literature review, which were
selected papers included in the study.
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016) Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med
Surg 4: 259. doi:10.4172/2329-6925.1000259
Page 3 of 12
Volume 4 • Issue 2 • 1000259
J Vasc Med Surg
ISSN: 2329-6925 JVMS, an open access journal
In 2012, La Vela and colleagues compared 1,142 male MS veterans
with healthy veterans and with the general population: hypertension
was more frequent in the rst group than in the second group and the
general population (respectively 47.6%, 41.2%, and 20.9%) [48].
Studies that evaluated the relationship between hypertension and
disability reported that the presence of arterial hypertension could
inuence the development of disability in MS patients. e presence
of hypertension at any time in the disease was associated with a 29%
increased risk of early gait disability [45] and a 32% increased risk of
visual disability [46].
Dyslipidemia: Case-control studies reported higher total and
LDL cholesterol in MS patients compared with control subjects
[42,49-51]. A signicant worsening in expanded disability status
scale was concomitant with higher baseline low-density lipoprotein
and total cholesterol levels [52]; a prospective study showed a direct
association between higher ratio of total cholesterol to high-density
lipoprotein and deterioration in EDSS [20]. Higher serum high-density
lipoprotein levels were associated with lower contrast-enhancing T1-
weigthed lesion volume, therefore with a protective eect in the acute
inammatory phase. Contrast enhancement in T1-weighted images
corresponds to focal areas of impairment of the blood-brain barrier
(BBB), associated with acute inammatory inltration [53]. erefore,
these enhancing lesions are a marker of active disease [54]. e
assumption is that apolipoprotein A-1 and paraoxonase anti-oxidant
enzyme associated with high-density lipoprotein may participate to its
anti-oxidant and anti-inammatory activities. [49,50,55-58].
A negative eect of hypercholesterolemia on the development of
disability was demonstrated by Weinstock-Guttman and colleagues in
2011 that reported the relationship between adverse lipid prole and an
increased number of new T2 lesions [59].
Moreover Swank and colleagues found that a diet low in saturated
fats and supplemented with vegetable oils provides benets on the
progression of MS and on disability [60,61].
Cigarette smoking: Compared to non-smokers, the risk of MS in
smokers is about 50% greater and is correlated with the duration and
with the amount of smoking [62-66].
In a study of a large number of nurses [61], the risk of MS is about
twice as high among women who smoked ≥ 25 pack-years (equivalent
to smoking a pack of cigarettes a day for more than 25 years) than non-
smokers. e negative eects of smoking on the risk of MS appear even
higher among men than women, with some studies reporting a nearly
three-fold increased risk in male never smokers compared to male
never smokers [66-68]. It was observed that only the tobacco smoke,
but not the snu, is associated with an increased risk of MS; from this
it follows that the combustion plays an important role for toxicity [66].
Mechanisms underlying the eects of smoking on the risk of MS are
still unknown. Mechanisms hypothesized are: eects on demyelination
[69,70], eects on the breaking of the blood-brain barrier [71],
immunomodulatory eects [72,73] and an increase in nitric oxide
metabolites and nitric oxide [74].
A study conducted on a population of 1465 MS patients showed a
negative inuence of smoking on the progression of the disease [75].
At baseline smokers have a worse disease than non-smokers and ex-
smokers in terms of EDSS and multiple sclerosis severity score (MSSS).
EDSS and MSSS were directly correlated with the number of pack-years
smoked at baseline. A longitudinal analysis reported no relationship
between smoking and progression EDSS at 2 and 5 years. However,
Vitamin D is a potent immunomodulatory [29] and some
studies have shown that administration of the active hormone
1,25-dihydroxyvitamin D prevents the onset and slows the progression
of the disease in mice [30,31].
It is unknown the exact mechanism of this protection; it could be
an indirect eect, possibly mediated by regulatory T cells [32] that are
suppressed in individuals with MS [33].
Serum levels of vitamin D are regulated by exposure to UV radiation
from sunlight. e exposure to UV light whole body suppresses EAE
in mice [34], enhances the function of regulatory T cells and increases
the production of immunosuppressive cytokines interleukin 4 and
interleukin 10 [35].
e risk of developing MS among migrants is higher when
migrations occurs in childhood and tend to decrease with increasing
age [36]. erefore the level of vitamin D in childhood may confer a
protection for the development of MS.
In literature, there are few data on vitamin D levels earlier in
life and risk of MS. At ages 16 and 19 years was demonstrated on
association between the risk of multiple sclerosis and the levels of
25-hydroxyvitamin D.
In conclusion, it is not clear if you can reduce the incidence
of MS in high-risk populations, increasing circulating levels of
25-hydroxyvitamin D. We need more evidence to support the
observational studies alone.
Diabetes: A cohort study [37] including 98% of the pediatric
diabetic population in Austria and Germany below the age of 21 years
analyzed the RR of MS co-occurrence and demonstrated that the RR
for MS in type 1 diabetes was three to almost ve times higher than that
in the healthy population.
is study shows that children and adolescents with diabetes have a
greater risk of developing MS. e risk for the development of MS was
higher in diabetic patients born during spring and summer; therefore
environmental factors can aect the risk for the onset of both diseases.
In studies on the Sardinian population, there was a prevalence of
type 1 diabetes by two to ve times greater in adult patients with MS
than the general population. [38,39].
Studies on the prevalence of type-2 diabetes (T2D) in patients with
MS compared to subjects without MS have contradictory results. e
studies on type -2 diabetes in MS patients are inconclusive. Hospitalized
MS patients showed a lower frequency of T2D compared to healthy
controls [19,40].
Outpatients with MS showed a higher prevalence of diabetes than
that in the general population [40,41] which was not observed in other
studies [42-44].
Marrie and colleagues using the NARCOMS registry, comparing
MS patients with and without T2D showed in the rst group a 29%
increased risk of early gait disability [43,45]. Furthermore, the
development of T2D during the course of the disease has led to a 35%
increased risk of mild visual impairment, 41% of moderate visual
disability and 54% of visual impairment [46].
Hypertension: e data on the prevalence of hypertension in
MS are contrasting. Some studies have reported no dierences or a
lower prevalence of hypertension in MS cases compared to controls
[13,19,40,44,45,47]. Kang and colleagues observed a higher frequency
of hypertension among MS cases (n = 898) compared to controls [42].
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016) Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med
Surg 4: 259. doi:10.4172/2329-6925.1000259
Page 4 of 12
Volume 4 • Issue 2 • 1000259
J Vasc Med Surg
ISSN: 2329-6925 JVMS, an open access journal
there was an earlier conversion from relapsing remitting multiple
sclerosis (RRMS) to secondary progressive multiple sclerosis (SPMS) in
smokers than in non-smokers and ex-smokers. MRI showed a greater
increase in the volume of T2 lesions and a greater reduction of brain
parenchymal fraction in smokers than non-smokers and ex-smokers
[62,76].
Hyperhomocysteinemia: Some authors have described
hyperhomocysteinemia (Hcy), a risk factor for thromboembolic
disease [77], in the serum of MS patients and linked this nding to
an increased risk of intercurrent vascular diseases [78-80]. Neurons
may be particularly susceptible to Hcy-induced excitotoxicity, with
the hypomethylation of myelin basic proteins and consequent lack
of myelin structure stability [81]. Signicantly higher serum levels of
homocysteine were found in MS patients compared to healthy controls,
even in the absence of the methylenetetrahydrofolate reductase
(MTHFR) mutation [82-85]. Hyperhomocysteinemia was also related
to cognitive impairment in patients with MS [86], and high levels of
Hcy with low levels of folate/vitamin B12 were associated with higher
EDSS scores [87]. However, the clinical implications of literature
ndings about the relationship between hyperhomocysteinemia and
MS are not completely clear, requiring further studies to elucidate the
prognostic implications of this evidence [82].
Cardiovascular diseases
Several studies have reported an increased cardiovascular
morbidity in MS patients and that the mortality related to stroke and
cardiovascular disease was higher in MS patients compared to the
general population [88,89].
MS patients who reported more than one CV risk factor at the time
of diagnosis had an increased chance of ambulatory disability and this
risk increased with the number of reported CV risk factors [9,45,90].
Although studies do not provide data on the pathogenesis of
cardiovascular disease in MS patients, inammation and autoimmunity
are central to the pathogenesis of MS and endothelial dysfunction,
which is a precursor of atherosclerosis and arterial vascular diseases.
e activated endothelium has pro-coagulant properties and is a
central component of the anti-phospholipid syndrome, associated with
MS and venous thromboembolism [91]. Moreover, platelet activation
in MS could increase thromboembolic events.
Stroke: Patients with chronic inammation of the central nervous
system, have a higher risk of developing cardiovascular disease,
including ischemic stroke [92-95]. e cause may be endothelial
dysfunction, induced by chronic inammation, resulting in progress of
atherosclerosis and cardiovascular disease [96,97] such as stroke.
Few studies have associated MS with the development of ischemic
stroke [11,98]. Allen et al. conducted a cross-sectional study on
hospitalized patients. MS was associated with a higher prevalence of
the concurrent primary diagnosis of ischemic stroke compared to
other inpatients but not with hemorrhagic stroke [19]. In addition,
other authors observed a higher risk of stroke and cerebrovascular
diseases in MS patients compared to the control population [42,98,99].
A large Danish cohort study showed an increased relative risk that
was very pronounced for young patients [11]. Similarly, in a recent
Swedish cohort study MS was associated with signicantly increased
relative risks for cerebrovascular disease. However, in this study, aer
the rst year of follow-up, the overall relative risk of occurrence of
cerebrovascular disease and in particular cerebral infarction was not
statistically signicant. In a recent study in Taiwan, a database of 22
million people, recruited by the National Health Insurance (NHI)
database, was used to evaluate the association between MS and ischemic
stroke in 1174 patients with multiple sclerosis and 4496 controls,
randomly selected and monitored in follow-up for 14 years. Patients
with multiple sclerosis showed an increased risk of stroke compared
to the control population in the rst 5 years of follow-up. Among
participants without comorbidities, multiple sclerosis group shows a
risk of ischemic stroke higher than healthy controls. Also in a subset
of the population aged <40 years, patients with multiple sclerosis had a
signicantly increased risk of stroke. erefore, patients with multiple
sclerosis, in particular young people, should be carefully monitored for
stroke prevention (99).
Ischemic heart disease: In a study by Allen et al. admission with
a principal diagnosis of myocardial infarction was less frequent in MS
patients than in other hospitalized patients [19]. In addition, Fleming
and Blake observed a lower frequency of acute myocardial infarction
and heart failure in MS cases than in matched hospitalized controls
[40]. A Swedish cohort study showed an overall non-statistically
signicant inverse association with ischemic heart disease [100]. In
contrast, many studies showed an increased relative risk for ischemic
heart disease in MS patients. A cross-sectional study by Lindegard
et al. comparing MS patients with the general population showed a
prevalence of myocardial infarction that was four to six times higher
in MS patients [101].
Two studies demonstrated that patients with multiple sclerosis
have an increased of risk of death from cardiovascular disease by 32-
34% compared to the general population [88-102]. Other studies have
also shown an increased risk of myocardial infarction and heart failure
in the MS group compared to the control group [11,98]. Finally other
studies reported a higher rate of coronary heart disease [42,98] and
ischemic heart disease [103] in MS patients compared to controls.
Venous thromboembolism: Many autoimmune diseases are
associated with an increased risk of venous thromboembolism
(VTE) [104,105]. An increased risk of VTE was observed in MS. An
increased incidence of VTE in patients with MS was reported [106-
107], especially during the rst year aer diagnosis [108]. Peeters et
al reported a risk of venous thromboembolism 2.5 times higher in MS
patients compared with healthy subjects. According to the authors, this
association can be explained by presence in MS patients of disability
and glucocorticoids therapy [109]. Only one study relieved a lower risk
of VTE in MS patients. e authors hypothesized that muscle spasm
could favor the venous return with a non-eective emptying of veins of
lower limb, reduction of venous stasis and consequently lower of risk
of deep venous thrombosis [110].
Microcirculation factors in MS
Endothelial dysfunction: Together with the loss of myelin and
the glial proliferation, perivascular inltration and endothelial damage
have been described as histological ndings in the brain tissue of
patients with MS [111]. is observation has led to the hypothesis
that a possible vascular factor should be involved in the pathogenesis
of multiple sclerosis [112]. In particular, vessels in the pathological
area showed changes that are typical of chronic inammation. Many
factors have been associated with chronic inammation and oxidative
damage in multiple sclerosis lesions [113-117]. Endothelin-1 (ET-
1) is a vasoconstrictor peptide that is regulated by a large number of
vasoactive agents [118] and, should play a role in endothelial damage
and vascular pathological reaction. It has been suggested that ET-1
plasma level dysregulation may lead to excessive vasoconstriction
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016) Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med
Surg 4: 259. doi:10.4172/2329-6925.1000259
Page 5 of 12
Volume 4 • Issue 2 • 1000259
J Vasc Med Surg
ISSN: 2329-6925 JVMS, an open access journal
or insucient vasodilatation, resulting in vasospasm and reduced
cerebral blood ow (CBF) [119]. In some studies, (ET-1) plasma levels
measured in a blood sample taken from one of the main routes of
venous eux from the brain were signicantly elevated in patients with
MS compared to healthy controls; this increase could lead to chronic
cerebral hypoperfusion, especially in the more susceptible white
matter, with resulting myelin damage and progressive neuronal loss
[118]. Moreover, during immunomodulatory treatment MS patients
showed a signicant reduction in ET-1 plasma levels [120]. Other
elements frequently associated with MS lesions include endothelial
micro-particles, endothelial cell adhesion molecules (selectins,
integrins, ICAMs, VCAMs, JAMs, lymphoid chemokines and their
receptors), pro-inammatory cytokines and vascular growth factors.
e overexpression of these molecules should participate in the
vascular remodeling that is typical of multiple sclerosis, in a deleterious
cascade. Over time these elements should lead to cerebral endothelial
junctional disorganization, pathological iron deposition, immune cell
extravasation and metabolic disorders leading to massive apoptosis,
which culminate in the loss rst of neural and then of glial cells [113-
117,121,122] associated with a variable degree of brain atrophy, with
permanent disability and cognitive impairment [123].
In addition, autoimmune mechanisms targeting the central
nervous system myelin have long been proposed, but not proved. 17
cells producing interleukin-17 and 1 cells producing interferon-
gamma are postulated to play major roles in initiating inammation
while regulatory T cell functions are dampened [124].
Alterations in peripheral blood mononuclear cell (PBMC) cytokine
production have been found in patients with MS compared to healthy
controls. Stimulated PBMC-produced TNF-α and IFN-γ modulated
MS relapse risk, such that increased TNF-α was protective, while
increased IFN-γ increased relapse risk. Zhou et al. reported the novel
modulation of TNF-α and IFN-γ associations with relapse by SNPs in
major cytokines [125].
Adult human astrocytes stimulated with interferon-γ, a common
inammatory cytokine that is evidently present in neuropathological
brains, exert potent neurotoxicity in vitro. is interferon- γ-induced
astrocytic neurotoxicity is mediated by the activation of the Janus kinase-
signal transducer and activator of transcription (STAT) 3 pathways in
astrocytes and involves the intracellular phosphorylation of STAT3 at
the tyrosine-705 residue. erefore, the control of STAT3 activation
in human astrocytes may be a promising new therapeutic strategy
for a broad spectrum of neurodegenerative and neuroinammatory
disorders where activated astrocytes may contribute to the pathology
[126].
Platelet abnormalities: In MS, as result of endothelial lesions, can
occur a platelet activation, which markers are Platelet microparticles
and CD62p expression. Sheremata et al showed, in MS patients,
elevated levels of plasmatic endothelial micro particles positive for
platelet endothelial cell adhesion molecule-1(CD31/PECAM). is
may be expression of the interaction of activated T-cells with the
endothelium. is interaction may play a role in the abnormalities
involved in the pathophysiology of MS.
e ndings of this study demonstrate that platelets are signicantly
activated in MS patients. However, their role in the pathogenesis of MS
remains unknown. [127].
Perfusion abnormalities: Perfusion-weighted MRI was used to
demonstrate abnormalities of regional cerebral hemodynamics in MS,
by using both exogenous tracers (eg, gadolinium-based contrast agents)
and endogenous arterial water (arterial spin labeling). It was found
that, while enhancing lesions showed increased perfusion, chronic MS
lesions showed reduced perfusion. In particular, disruption of the blood-
brain barrier was described before and during the development of focal
MS lesions and it is represented by a local gadolinium-enhancing area
on T1w MR imaging [128]. Diuse enhancing areas were characterized
by increased CBF and CBV [129]; while ring-enhancing areas CBF and
CBV were increased in the ring tissue, CBF was reduced inside the ring.
Because of the similarity with acute ischemia, this nding is evocative
for a focal central ischemic zone and it suggests that focal ischemia
should play a crucial role in the development of some type of focal MS
lesions [129,130]. Law et al, using dynamic susceptibility contrast MR
(DSC-MR) imaging, demonstrate a decreased cerebral blood ow and
a prolonged mean transit time in periventricular regions of normal
appearing white matter (NAWM) in MS patients compared with
controls [131]. Other studies showed diuse hypoperfusion in various
regions (NAWM, cortex and deep GM) independently of the clinical
MS variant of the patient [132,133]. ese changes in brain tissue
perfusion were correlated to clinical disability and neuropsychological
impairment [134]. One study [135] reported a correlation between
decreased perfusion and decreased mean diusivity in the corpus
callosum of patients with RRMS, a nding more consistent with what
is seen in primary ischemia than in secondary hypoperfusion. e data
of perfusion-weighted MR imaging were conrmed by a study in which
the spatial distribution of the MS lesions were compared with images
of a SPECT (Single-Proton Emission-Computed Tomography) atlas of
healthy individuals. Lesions were localized in relatively lower normal
perfusion areas than normal appearing white matter areas [136].
Venous alterations
Over a century ago, researchers were considering the role of
abnormal venous in MS. Subsequently, more recently a small but
important body of research, including Zamboni’s work, contributed to
extend early observations.
e rst observations on vascular anomalies were documented by
French anatomist Cruveilhier [137] in 1839, who studied the dierences
between areas of sclerosis and results of embolic events. In 1863,
Rindeisch [138] described an engorged blood vessel in the center of
the plaques in autopsy specimens of MS brains and in the same period,
Charcot [5] noted the constant presence of vascular obstruction in MS.
Further evidence of the vascular mechanical eect comes from the
observations of Putnam that studied the eects of obstructed venous
ow in the brain of dogs and recorded the development in these
animals of various abnormalities similar to encephalitis or multiple
sclerosis. Fog stated that MS lesions typically developed around small
veins [139,140]. Further evidence of the vascular mechanical eect
comes from Allen’s observations, who noticed the wide vascular beds
around veins and the central widening of the venous tree that testies
an intermittent increase in cerebral pressure [141]. During 1980’s,
studying MS plaque development, Adams conrmed the crucial
contribution of vascular anomalies [121].
In 2006 [142], Dr. Zamboni presented a paper recorded some
similarities between Chronic Venous Disease and MS, including
presence of iron/brinogen deposition. From the rst observation of
MS up to modern MRI studies, a possible connection between MS and
vascular system is evident.
In MS the visibility of small medullary vein is reduced [143], and the
cerebral circulation time is increased [144]. All these results demonstrate
that hemodynamic abnormalities and a vascular compromise are
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016) Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med
Surg 4: 259. doi:10.4172/2329-6925.1000259
Page 6 of 12
Volume 4 • Issue 2 • 1000259
J Vasc Med Surg
ISSN: 2329-6925 JVMS, an open access journal
associated with MS. Zamboni and colleagues proposed that chronic
cerebrospinal venous insuciency (CCSVI) and secondarily venous
hypertension can lead to increase iron deposition along venous
pathway. e iron deposition within the brain parenchyma leads to the
inammation and tissue injury seen in MS [145]. Whether observed
increase in iron in the central nervous system is etiologic to MS or
the result of MS is a fundamental and yet unanswered question in the
debate of CCSVI and MS. e morphological basis of the abnormalities
in venous return are intraluminal defects as septa, membranes and
anomalies of valves in the internal jugular vein (IJV) and azygos vein,
compression muscle, hypoplasia or agenesis.
In 2009 [146], Dr. Zamboni using duplex ultrasonography
identied CCSVI as a vascular condition characterized by anatomical
and functional abnormalities of the intra- and extra-cranial veins
with ow anomalies and presence of collateral venous channels and
demonstrated a correlation between CCSVI and MS.
e outow anomaly focused in particular on 5 anomalous
parameters of cerebral venous drainage:
1. Reux constantly present in IJV and/or Vertebral Veins (VVs)
with the head at 0° and 90° assessed as ow reversal from
physiologic direction for a duration of >0.88 s.
2. Bidirectional ow (or reux) in the intracranial veins and
sinuses.
3. High-resolution B-mode ultrasound evidence of proximal
stenosis of the IJV (Cross sectional area (CSA) < 0.3 cm2).
4. Absence of detectable ow in the IJV and/or VVs despite
numerous deep inspirations with the head positioned at 0° and
90°.
5. Abnormal change of the IJV CSA with change in position
(negative ∆ CSA).
In the study of Zamboni the presence of at least two of these
parameters in a single subject was dened as abnormality. Heterogeneity
among studies investigating chronic cerebrospinal venous insuciency
detected by duplex sonography for the diagnosis of MS has been wide,
with reported sensitivity ranging from 0% to 100%. [146-154].
Unfortunately, B-mode anomalies of veins (intraluminal defects
such as aps, septa, membranes and valve defects) are dicult to
detect with ultrasound. Several potential factors, both physiologic
and technical, may aect the acquisition and interpretation of duplex
sonograms (respiratory variability, heart rate cardiac rhythm, the
hydration status of the patient, eects of head positioning, degree
of external compression, pulse repetition frequency setting, cross-
sectional area measurement by B-mode versus color Doppler imaging,
and sample volume placement during Doppler imaging).
Moreover, the innovation work of Zamboni is limited by the
absence of a reference standard for intracranial evaluation, and they
used a technology called Quality Doppler Proles (QDP) that is not
available on all ultrasonic scanning systems.
e dierence in CCSVI prevalence between dierent published
studies that use noninvasive or invasive imaging techniques
underscored the central importance of a multimodal imaging approach
for an optimal understanding of venous abnormalities indicative of
CCSVI [155]. At the current state, researchers have been unable to
dene if CCSVI has a crucial role in multiple sclerosis pathogenesis
and this point has raised serious objections to accept the hypothesis of
CCSVI as mechanism originating MS. Actually, additional researches
on the subject are in progress and the results are still not determining.
A 2013 study found out that CCSVI is uncommon both in MS patients
and healthy controls, while narrowing of the cervical veins is equally
common [156].
Lanzillo et al. observed a positive correlation between presence
of CCSVI and the patients’ age. However, no correlation was seen
between CCSVI and clinical forms of MS, except for IJVs blocked ow
that was more prevalent in progressive forms than in non-progressive
forms [157]. Other authors also observed a positive correlation between
age, disease duration and outow block in the cervical veins [158]. e
observation seems to conrm the hypothesis that venous abnormalities
detectable in patients with MS, and in particular the ow block, may be
a secondary and progressive impairment of the veins and not a primary
causal factor.
Valvular IJV abnormalities are strongly associated with venous
blood reux in the intracranial veins, are more frequent in SP form
and related to the EDSS class [159]. Pathologic studies identied
vascular pathology in any layer of the jugular wall. Intraluminal defects
are considered one the mechanisms causing a delay of jugular ow in
patients with CCSVI.
In recent years, some authors have tried to study the ultra structure
of extra cranial veins in patients with MS. e authors evaluated
the expression of collagen type I and III, cytoskeletal protein and
inammatory markers (CD3 and CD8), in patients with CCSVI
who underwent IJV reconstruction for restenosis aer percutaneous
transluminal angioplasty and in individuals without MS or other
neurological diseases who underwent carotid endarterectomy with
autologous external JV patch because of high-grade carotid stenosis.
Collagen deposition was assessed by polarized light examination. e
extra cranial veins of MS patients showed focal thickenings of the wall,
with an altereted proportion of collagen I and III within the adventitia,
due to a relative increase in type III collagen deposition. No dierences
in cytoscheletal protein and inammatory marker expression were
observed [160]. Pascolo et al. evaluated jugular tissue samples by both
advanced synchrotron X-Ray Fluorescence (XRF) microscopy, and
histological examination in two MS patients with CCSVI and in two
control subjects. XRF analyses showed an increased Ca presence in
the pathological samples, corresponding to rare and small basophilic
bodies suggestive of micro calcications. e samples of healthy
controls showed no structural alterations at histological examination
at XRF [161]. Recently Zamboni et al. studied the ultrastructure of IJV
using a scanning electron microscopy (SEM) in 7 patients with CCSVI,
underwent to vein open surgery and in 3 healthy controls who underwent
to vein repair for traumatic causes. e veins of CCSVI patients
showed partially detached endothelial cells, while the veins of healthy
controls showed an intact endothelium, with regular arrangement
of cells [162]. In conclusions, the IJVs of MS patients demonstrated
changes in collagen and increased calcium content in adventitia, and
disruption of endothelial layer, without coexistent inammation. ese
alterations could be due to endothelial chronic stress secondary to
altered hemodynamic. Moreover, the venous alterations as obstruction
and/or reux determine stasis and increased permeability to red blood
cells and brinogen. e brinogen extravagated out of micro vessels
polymerize to brin that, deposited outside the vessel, reduces tissue
oxygenation. e brinogen can also induce local activation of myelin
antigen-specic T-cells, and macrophage recruitment [163].
erefore the link between venous circulation and immune response
could be the extravasation of macromolecules into the interstitium
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016) Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med
Surg 4: 259. doi:10.4172/2329-6925.1000259
Page 7 of 12
Volume 4 • Issue 2 • 1000259
J Vasc Med Surg
ISSN: 2329-6925 JVMS, an open access journal
of brain that can induce neuroinammation, autoimmunity and
demyelization and macrophage recruitment (Figure 2).
MS and infectious pathogens
Over the years, many studies have been published about
MS geographical distribution and about the consistency of the
hypothesis that environmental determinants should play a role in
MS pathogenesis [3,4,164-168]. Among environmental factors,
epidemiologic evidence and biochemical and immunological data from
dierent populations suggest the involvement of an infective agent. e
hypothesized mechanism is that an infectious pathogen, possibly with
the concurrence of other inducing agents, causes a deregulation of
lymphocyte function that leads to an uncontrolled immune reaction
in the CNS, resulting; in early histological changes with an alteration of
the blood-brain barrier that gradually evolve into the more typical CNS
lesions [111,168-170]. Molecular mimicry between protein sequences
from a particular microorganism and structural proteins present in the
normal CNS is a possible explanation for autoimmune reactions in MS
[171]. Many potential infective agents have been indicated as possible
etiologic candidates, but no common interpretation of literature
evidence has emerged [172,173]; the strongest associations are those
with the gut microbiome [174-176], Rickettsia [177] and Chlamydia
[178-180] among bacteria and the Epstein-Barr [181,182] and herpes
viruses among viruses [183-185]. Recently a chronic persisting
venulitis aecting the cerebrospinal venous system, determined by
Chlamydophila pneumonia respiratory infection observed in certain
patients with MS, was hypothesized to lead to typical neural damage
[186,187].
Vascular adverse eects of MS therapy
e impact of the more-or-less recently introduced disease-
modifying drugs (DMDs) has changed patients ‘outcome [188] but
in some cases has shown negative eects at the cardiovascular level
in various ways, whether directly or indirectly [44,189]. Systemic
glucocorticoids have been associated with an increased risk of
cardiovascular diseases including stroke, myocardial infarction, and
atrial brillation [190,191]. Congestive heart failure was reported for
both interferons and mitoxantrone, but data are scarce and causality
is doubtful [192,193]. Mitoxantrone induces congestive heart failure,
although it is usually asymptomatic [193]. Fingolimod has been
associated with hypertension, bradycardia and atrioventricular blocks
[194,195] and, in a case report with ischemia of ngers, arterial
vasospasm [196]. In contrast, the use of natalizumab was associated
with lower cardiovascular risk factors [44].
Figure 2: The immunological factors and vascular factors are connected. The scheme depicts the possible interplay between changes in macro and microcirculation
and immunological factors. An altered macro-and microcirculation induces changes in endothelial cells that may determine cell-mediated immunity reactions:
presenting antigens to lymphocytes, promoting inammation in response to cytokines, and responding to oxidant injury. The autoimmune inammation can
determine iron deposition and increased leukocyte inltration. In turn the vascular mediators released by immune cells, and the activated endothelium can cause
neovascularization, lead to changes in microcirculation and recruitment of further effector cells. Venous alterations as obstruction and/or reux determine stasis
and increased permeability to red blood cells and brinogen. The brinogen extravasation reduce tissue oxygenation and induce macrophage recruitment and local
activation of myelin antigen-specic T-cells. All factors affect the neurovascular unit.
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016) Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med
Surg 4: 259. doi:10.4172/2329-6925.1000259
Page 8 of 12
Volume 4 • Issue 2 • 1000259
J Vasc Med Surg
ISSN: 2329-6925 JVMS, an open access journal
Interferon beta-1b is an immunomodulatory drug used for a
treatment of several diseases including MS. In 1993 recombinant
interferon beta-1b (Betaseron) was approved for the treatment of
exacerbations of multiple sclerosis. e drug’s eectiveness was
modest, with 30% of relapses and the appearance of side eects such as
local irritation, and a u-like syndrome [197,198].
Conclusions
e etiology of MS and neurologic involvement are still unknown
and the clinical course extremely variable. It can take various forms,
including those relapsing and progressive ones. Each of these
forms involves numerous disease mechanisms, and as such, MS is a
multifactorial disease.
e typical characteristics of MS pathophysiology include
inammatory and vascular factors that lead to the molecular
interactions between cellular components (brain endothelial cells,
astrocytes, pericytes, inammatory cells, and neurons) and myelin
components, resulting in increased blood-brain barrier permeability,
neurovascular uncoupling and dysfunction and neuronal damage.
is review focuses on vascular abnormalities that have been
described in MS. Recent hypotheses and evidence suggest that vascular
components may be initiating triggers for neuronal pathology and
subsequent neurological manifestations of the disease.
Patients with MS have more cardiovascular risk factors, an
increased risk for ischemic stroke and an increased risk of venous
thrombosis and pulmonary embolism. Vascular comorbidities are
common in MS and adversely inuence disability. ere is evidence
suggesting that MS patients are more susceptible to cardiovascular risk
factors than healthy controls.
Several studies have demonstrated regional cerebral perfusion
abnormalities in MS compared to controls. e relationship between
the disturbances in cerebral venous outow and neurological disorders
remains an open issue that requires further studies.
e debate on whether vascular events are the primary cause of
neurological diseases or rather a mere participant recruited from a
primary neuronal origin of pathology is still open.
e high degree of comorbidity between vascular disease and MS
suggests that vascular pathology may be an important factor causing
neuronal dysfunction or degeneration in multiple sclerosis.
In our opinion, this review is useful to shed light on a hot topic in
which literature is still lacking.
e results in literature are not always consistent, both for the
frequency of association between vascular disease and neurological
disease, both for the relationship between vascular disease, cerebral
damage and clinical disability. Several studies are mostly “case-control”
and studies on population are few. Furthermore, the authors of studies
do not always come from centers with expertise in epidemiological
assessment. It is possible that many of the changes reported in the
literature are a result of bias in the interpretation or eects of the
disease itself, or activation of the immune system or of drug treatment.
erefore, more rigorous epidemiological and pathophysiological
studies are needed to verify the biological plausibility between vascular
damage and demyelinating-neurodegenerative diseases.
Acknowledgement
This work was supported by a research grant from the Italian Ministry for
Education University and Research in the framework of PRIN (2010XE5L2R_004).
The funders had no role in study design, data collection and analysis, decision to
publish, or preparation of the manuscript. The authors thank Dr. Marianna Malgieri
for the editing of the manuscript.
References
1. Noseworthy JH, Lucchinetti C, Rodriguez M, Weinshenker BG (2000) Multiple
sclerosis. N Engl J Med 343: 938-952.
2. Ramagopalan SV, Dobson R, Meier UC, Giovannoni G (2010) Multiple
sclerosis: risk factors, prodromes, and potential causal pathways. Lancet
Neurol 9: 727-739.
3. Ebers GC, Sadovnick AD (1993) The geographic distribution of multiple
sclerosis: a review. Neuroepidemiology 12: 1-5.
4. Kurtzke JF (2000) Multiple sclerosis in time and space-geographic clues to
cause. J Neurovirol 6 Suppl 2: S134-140.
5. Charcot J (1868) Histologie de la sclerose en plaques. Gaz Hopit Civilis Milit
4: 554-566.
6. Adams CW (1988) Perivascular iron deposition and other vascular damage in
multiple sclerosis. J Neurol Neurosurg Psychiatry 51: 260-265.
7. Fog T (1965) The topography of plaques in multiple sclerosis with special
reference to cerebral plaques. Acta Neurol Scand Suppl 15: 1-161.
8. Tracy J, Putnam MD (1933) The pathogenesis of multiple sclerosis, a possible
vascular factor. N Engl J Med 209: 786-790.
9. Tettey P, Simpson S Jr, Taylor BV, van der Mei IA (2014) Vascular
comorbidities in the onset and progression of multiple sclerosis. J Neurol Sci
347: 23-33.
10. Karmon Y, Ramanathan M, Minagar A, Zivadinov R, Weinstock-Guttman B
(2012) Arterial, venous and other vascular risk factors in multiple sclerosis.
Neurol Res 34: 754-760.
11. Christiansen CF, Christensen S, Farkas DK, Miret M, Sørensen HT et al.
(2010) Risk of arterial cardiovascular diseases in patients with multiple
sclerosis: a population-based cohort study. Neuroepidemiology 35: 267-274.
12. Moccia M, Lanzillo R, Palladino R2,3, Maniscalco GT, et al. (2015) The
Framingham cardiovascular risk score in multiple sclerosis. Eur J Neurol 22:
1176-1183.
13. Sternberg Z, Leung C, Sternberg D, Li F, Karmon Y, et al. (2013) The
prevalence of the classical and non-classical cardiovascular risk factors in
multiple sclerosis patients. CNS Neurol Disord Drug Targets 12: 104-111.
14. Marrie RA, Reider N, Cohen J, Stuve O, Trojano M, et al. (2015) A systematic
review of the incidence and prevalence of cardiac, cerebrovascular, and
peripheral vascular disease in multiple sclerosis. Mult Scler 21: 318-331.
15. Kappus N, Weinstock-Guttman B, Hagemeier J, Kennedy C, Melia R, et al.
(2015) Cardiovascular risk factors are associated with increased lesion burden
and brain atrophy in multiple sclerosis. J Neurol Neurosurg Psychiatry.
16. Munger KL, Bentzen J, Laursen B, Stenager E, Koch-Henriksen N, et al.
(2013) Childhood body mass index and multiple sclerosis risk: a long-term
cohort study. Mult Scler 19: 1323-1329.
17. Pilutti LA, Dlugonski D, Pula JH, Mot RW (2012) Weight status in persons with
multiple sclerosis: implications for mobility outcomes. J Obes 2012: 868256.
18. Pinhas-Hamiel O, Livne M, Harari G, Achiron A (2015) Prevalence of
overweight, obesity and metabolic syndrome components in multiple sclerosis
patients with signicant disability. Eur J Neurol.
19. Allen NB, Lichtman JH, Cohen HW, Fang J, Brass LM, et al. (2008) Vascular
disease among hospitalized multiple sclerosis patients. Neuroepidemiology
30: 234-238.
20. Khurana SR, Bamer AM, Turner AP, Wadhwani RV, Bowen JD, et al. (2009)
The prevalence of overweight and obesity in veterans with multiple sclerosis.
Am J Phys Med Rehabil 88: 83-91.
21. Tettey P, Simpson S Jr, Taylor B, Blizzard L, Ponsonby AL, et al. (2014) An
adverse lipid prole is associated with disability and progression in disability,
in people with MS. Mult Scler 20: 1737-1744.
22. Lucas RM, Ponsonby AL, Dear K, Valery PC, Pender MP, et al. (2011) Sun
exposure and vitamin D are independent risk factors for CNS demyelination.
Neurology 76: 540-548.
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016) Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med
Surg 4: 259. doi:10.4172/2329-6925.1000259
Page 9 of 12
Volume 4 • Issue 2 • 1000259
J Vasc Med Surg
ISSN: 2329-6925 JVMS, an open access journal
23. Munger KL, Levin LI, Hollis BW, Howard NS, Ascherio A (2006) Serum
25-hydroxyvitamin D levels and risk of multiple sclerosis. JAMA 296: 2832-
2838.
24. van der Mei IA, Ponsonby AL, Dwyer T, Blizzard L, Simmons R, et al. (2003)
Past exposure to sun, skin phenotype, and risk of multiple sclerosis: case-
control study. BMJ 327: 316.
25. Weinstock-Guttman B, Mehta BK, Ramanathan M, Karmon Y, Henson LJ, et
al. (2012) Vitamin D and multiple sclerosis. Neurologist 18: 179-183.
26. Kappus N, Weinstock-Guttman B, Hagemeier J, Kennedy C, Melia R, et al. (2016)
Cardiovascular risk factors are associated with increased lesion burden and
brain atrophy in multiple sclerosis. J Neurol Neurosurg Psychiatry 87:181-187.
27. McGreevy C, Williams D (2011) New insights about vitamin D and
cardiovascular disease: a narrative review. Ann Intern Med 155: 820-826.
28. Ascherio A, Munger KL, Simon KC (2010) Vitamin D and multiple sclerosis.
Lancet Neurol 9: 599-612.
29. Hayes CE, Nashold FE, Spach KM, Pedersen LB (2003) The immunological
functions of the vitamin D endocrine system. Cell Mol Biol (Noisy-le-grand)
49: 277-300.
30. Lemire JM, Archer DC (1991) , 25-dihydroxyvitamin D3 prevents the in vivo
induction of murine experimental autoimmune encephalomyelitis. J Clin Invest
87: 1103-1107.
31. Cantorna MT, Hayes CE, DeLuca HF (1996) ,25-Dihydroxyvitamin D3
reversibly blocks the progression of relapsing encephalomyelitis, a model of
multiple sclerosis. Proc Natl Acad Sci USA 93: 7861-7864.
32. Hayes CE, Nashold FE, Hoag KA, Goverman J, Hayes CE (2001) Rag-1-
dependent cells are necessary for ,25-dihydroxyvitamin D(3) prevention of
experimental autoimmune encephalomyelitis. J Neuroimmunol 119:16-29.
33. Viglietta V, Baecher-Allan C, Weiner HL, Haer DA (2004) Loss of functional
suppression by CD4+CD25+ regulatory T cells in patients with multiple
sclerosis. J Exp Med 199: 971-979.
34. Hauser SL, Weiner HL, Che M, Shapiro ME, Gilles F, et al. (1984) Prevention
of experimental allergic encephalomyelitis (EAE) in the SJL/J mouse by whole
body ultraviolet irradiation. J Immunol 132: 1276-1281.
35. Aubin F (2003) Mechanisms involved in ultraviolet light-induced
immunosuppression. Eur J Dermatol 13: 515-523.
36. Gale CR, Martyn CN (1995) Migrant studies in multiple sclerosis. Prog
Neurobiol 47: 425-448.
37. Bechtold S, Blaschek A, Raile K, Dost A, Freiberg C, et al. (2014) Higher
relative risk for multiple sclerosis in a pediatric and adolescent diabetic
population: analysis from DPV database. Diabetes Care 37: 96-101.
38. Marrosu MG, Motzo C, Murru R, Lampis R, Costa G, et al. (2004) The co-
inheritance of type 1 diabetes and multiple sclerosis in Sardinia cannot be
explained by genotype variation in the HLA region alone. Hum Mol Genet 13:
2919-2924.
39. Pitzalis M, Zavattari P, Murru R, Deidda E, Zoledziewska M, et al. (2008)
Genetic loci linked to type 1 diabetes and multiple sclerosis families in
Sardinia. BMC Med Genet 9: 3.
40. Fleming ST, Blake RL Jr (1994) Patterns of comorbidity in elderly patients with
multiple sclerosis. J Clin Epidemiol 47: 1127-1132.
41. Hussein WI, Reddy SS (2006) Prevalence of diabetes in patients with multiple
sclerosis. Diabetes Care 29: 1984-1985.
42. Kang JH, Chen YH, Lin HC (2010) Comorbidities amongst patients with multiple
sclerosis: a population-based controlled study. Eur J Neurol 17: 1215-1219.
43. Marrie RA, Yu BN, Leung S, Elliott L, Caetano P, et al. (2012) Rising
prevalence of vascular comorbidities in multiple sclerosis: validation of
administrative denitions for diabetes, hypertension, and hyperlipidemia. Mult
Scler 18: 1310-1319.
44. Sternberg Z, Leung C, Sternberg D, Yu J, Hojnacki D (2014) Disease
modifying therapies modulate cardiovascular risk factors in patients with
multiple sclerosis. Cardiovasc Ther 32: 33-39.
45. Marrie RA, Rudick R, Horwitz R, Cutter G, Tyry T, et al. (2010) Vascular
comorbidity is associated with more rapid disability progression in multiple
sclerosis. Neurology 74:1041-1047.
46. Marrie RA, Cutter G, Tyry T (2011) Substantial adverse association of visual
and vascular comorbidities on visual disability in multiple sclerosis. Mult Scler
17:1464-1471.
47. Pinhas-Hamiel O, Livne M, Harari G, Achiron A (2015) Prevalence of
overweight, obesity and metabolic syndrome components in multiple sclerosis
patients with signicant disability. Eur J Neurol 22:1275-1279.
48. Lavela SL, Prohaska TR, Furner S, Weaver FM (2012) Chronic diseases in
male veterans with multiple sclerosis. Prev Chronic Dis 9: E55.
49. Giubilei F, Antonini G, Di Legge S, Sormani MP, Pantano P, et al. (2002) Blood
cholesterol and MRI activity in rst clinical episode suggestive of multiple
sclerosis. Acta Neurol Scand 106: 109-112.
50. Jamroz-Wisniewska A, Beltowski J, Stelmasiak Z, Bartosik-Psujek H (2009)
Paraoxonase 1 activity in different types of multiple sclerosis. Mult Scler 15:
399-402.
51. Salemi G, Gueli MC, Vitale F, Battaglieri F, Guglielmini E, et al. (2010) Blood
lipids, homocysteine, stress factors, and vitamins in clinically stable multiple
sclerosis patients. Lipids Health Dis 18: 9-19.
52. Palavra F, Marado D, Mascarenhas-Melo F, Sereno J, Teixeira-Lemos E, et al.
(2013) New markers of early cardiovascular risk in multiple sclerosis patients:
oxidized-LDL correlates with clinical staging. Dis Markers 34: 341-348.
53. Kappos L, Moeri D, Radue EW, Schoetzau A, Schweikert K, et al. (1999)
Predictive value of gadolinium-enhanced magnetic resonance imaging for
relapse rate and changes in disability or impairment in multiple sclerosis: a
meta-analysis. Gadolinium MRI Meta-analysis Group. Lancet 353: 964-969.
54. He J, Grossman RI, Ge Y, Mannon LJ (2001) Enhancing patterns in multiple
sclerosis: evolution and persistence. AJNR Am J Neuroradiol 22: 664-669.
55. Weinstock-Guttman B, Zivadinov R, Mahfooz N, Carl E, Drake A, et al. (2011)
Serum lipid proles are associated with disability and MRI outcomes in multiple
sclerosis. J Neuroinammation 8: 127.
56. Dietschy JM, Turley SD (2004) Thematic review series: brain Lipids. Cholesterol
metabolism in the central nervous system during early development and in the
mature animal. J Lipid Res 45: 1375-1397.
57. Leoni V, Caccia C (2011) Oxysterols as biomarkers in neurodegenerative
diseases. Chem Phys Lipids 164: 515-524.
58. Fessler MB, Parks JS (2011) Intracellular lipid ux and membrane microdomains
as organizing principles in inammatory cell signaling. J Immunol 187: 1529-
1535.
59. Weinstock-Guttman B, Zivadinov R, Horakova D, Havrdova E, Qu J, et al.
(2013) Lipid proles are associated with lesion formation over 24 months in
interferon-ß treated patients following the rst demyelinating event. J Neurol
Neurosurg Psychiatry 84:1186-1191.
60. Swank RL, Goodwin J (2003) Review of MS patient survival on a Swank low
saturated fat diet. Nutrition 19: 161-162.
61. Swank RL, Goodwin JW (2003) How saturated fats may be a causative factor
in multiple sclerosis and other diseases. Nutrition 19: 478.
62. Hernán MA, Olek MJ, Ascherio A (2001) Cigarette smoking and incidence of
multiple sclerosis. Am J Epidemiol 154: 69-74.
63. Hernán MA, Jick SS, Logroscino G, Olek MJ, Ascherio A, et al. (2005) Cigarette
smoking and the progression of multiple sclerosis. Brain 128: 1461-1465.
64. Villard-Mackintosh L, Vessey MP (1993) Oral contraceptives and reproductive
factors in multiple sclerosis incidence. Contraception 47: 161-168.
65. Thorogood M, Hannaford PC (1998) The inuence of oral contraceptives on
the risk of multiple sclerosis. Br J Obstet Gynaecol 105: 1296-1299.
66. Hedström AK, Bäärnhielm M, Olsson T, Alfredsson L (2009) Tobacco smoking,
but not Swedish snuff use, increases the risk of multiple sclerosis. Neurology
73: 696-701.
67. Riise T, Nortvedt MW, Ascherio A (2003) Smoking is a risk factor for multiple
sclerosis. Neurology 61: 1122-1124.
68. Sundström P, Nyström L, Hallmans G (2008) Smoke exposure increases the
risk for multiple sclerosis. Eur J Neurol 15: 579-583.
69. Bass NH (1968) Pathogenesis of myelin lesions in experimental cyanide
encephalopathy. A microchemical study. Neurology 18: 167-177.
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016) Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med
Surg 4: 259. doi:10.4172/2329-6925.1000259
Page 10 of 12
Volume 4 • Issue 2 • 1000259
J Vasc Med Surg
ISSN: 2329-6925 JVMS, an open access journal
70. Lessell S (1971) Experimental cyanide optic neuropathy. Arch Ophthalmol 86:
194-204.
71. Chen JL, Wei L, Bereczki D, Hans FJ, Otsuka T, et al. (1995) Nicotine raises
the inux of permeable solutes across the rat blood-brain barrier with little or
no capillary recruitment. J Cereb Blood Flow Metab 15: 687-698.
72. Sopori ML, Kozak W (1998) Immunomodulatory effects of cigarette smoke. J
Neuroimmunol 83: 148-156.
73. Francus T, Klein RF, Staiano-Coico L, Becker CG, Siskind GW (1988) Effects
of tobacco glycoprotein (TGP) on the immune system. II. TGP stimulates the
proliferation of human T cells and the differentiation of human B cells into Ig
secreting cells. J Immunol 140: 1823-1829.
74. Rejdak K, Eikelenboom MJ, Petzold A, Thompson EJ, Stelmasiak Z, et
al. (2004) CSF nitric oxide metabolites are associated with activity and
progression of multiple sclerosis. Neurology 63: 1439-1445.
75. Healy BC, Ali EN, Guttmann CR, Chitnis T, Glanz BI, et al. (2009) Smoking
and disease progression in multiple sclerosis. Arch Neurol 66: 858-864.
76. Handel AE, Williamson AJ, Disanto G, Dobson R, Giovannoni G, et al. (2011)
Smoking and multiple sclerosis: an updated meta-analysis. PLoS One 6:
e16149.
77. Hankey GJ, Eikelboom JW (1999) Homocysteine and vascular disease.
Lancet 354: 407-413.
78. Zhu Y, He ZY, Liu HN (2011) Meta-analysis of the relationship between
homocysteine, vitamin B12, folate and multiple sclerosis. J Clin Neurosci 18:
933-938.
79. Ansari R, Mahta A, Mallack E2, Luo JJ (2014) Hyperhomocysteinemia and
neurologic disorders: a review. J Clin Neurol 10: 281-288.
80. Aksungar FB, Topkaya AE, Yildiz Z, Sahin S, Turk U (2008) Coagulation
status and biochemical and inammatory markers in multiple sclerosis. J Clin
Neurosci 15: 393-397.
81. Ho PI, Ortiz D, Rogers E, Shea TB (2002) Multiple aspects of homocysteine
neurotoxicity: glutamate excitotoxicity, kinase hyperactivation and DNA
damage. J Neurosci Res 70: 694-702.
82. Triantafyllou N, Evangelopoulos ME, Kimiskidis VK, Kararizou E, Boudou F,
et al. (2008) Increased plasma homocysteine levels in patients with multiple
sclerosis and depression. Ann Gen Psychiatry 7: 17.
83. Vrethem M, Mattsson E, Hebelka H, Leerbeck K, Osterberg A, et al. (2003)
Increased plasma homocysteine levels without signs of vitamin B12 deciency
in patients with multiple sclerosis assessed by blood and cerebrospinal uid
homocysteine and methylmalonic acid. Mult Scler 9: 239-245.
84. Ramsaransing GS, Fokkema MR, Teelken A, Arutjunyan AV, Koch M, et al.
(2006) Plasma homocysteine levels in multiple sclerosis. J Neurol Neurosurg
Psychiatry 77: 189-192.
85. Río J, Montalban J, Tintoré M, Codina A, Malinow MR (1994) Serum
homocysteine levels in multiple sclerosis. Arch Neurol 51: 1181.
86. Russo C, Morabito F, Luise F, Piromalli A, Battaglia L, et al. (2008)
Hyperhomocysteinemia is associated with cognitive impairment in multiple
sclerosis. J Neurol 255: 64-69.
87. Moghaddasi M, Mamarabadi M, Mohebi N, Razjouyan H, Aghaei M (2013)
Homocysteine, vitamin B12 and folate levels in Iranian patients with Multiple
Sclerosis: a case control study. Clin Neurol Neurosurg 115: 1802-1805.
88. Koch-Henriksen N, Brønnum-Hansen H, Stenager E (1998) Underlying cause
of death in Danish patients with multiple sclerosis: results from the Danish
Multiple Sclerosis Registry. J Neurol Neurosurg Psychiatry 65: 56-59.
89. Hirst C, Swingler R, Compston DA, Ben-Shlomo Y, Robertson NP (2008)
Survival and cause of death in multiple sclerosis: a prospective population-
based study. J Neurol Neurosurg Psychiatry 79: 1016-1021.
90. Marrie RA, Horwitz R, Cutter G, Tyry T, Campagnolo D, et al. (2009)
Comorbidity delays diagnosis and increases disability at diagnosis in MS.
Neurology 72: 117-124.
91. Ruiz-Irastorza G, Crowther M, Branch W, Khamashta MA (2010)
Antiphospholipid syndrome. Lancet 376: 1498-1509.
92. Peterson LK, Fujinami RS (2007) Inammation, demyelination,
neurodegeneration and neuroprotection in the pathogenesis of multiple
sclerosis. J Neuroimmunol 184: 37-44.
93. Brück W (2005) The pathology of multiple sclerosis is the result of focal
inammatory demyelination with axonal damage. J Neurol 252 Suppl 5: v3-9.
94. Slawta JN, Wilcox AR, McCubbin JA, Nalle DJ, Fox SD, et al. (2003) Health
behaviors, body composition, and coronary heart disease risk in women with
multiple sclerosis. Arch Phys Med Rehabil 84: 1823-1830.
95. Marrie R, Horwitz R, Cutter G, Tyry T, Campagnolo D, et al. (2009) High
frequency of adverse health behaviors in multiple sclerosis. Mult Scler 15:
105-113.
96. Ross R (1999) Atherosclerosis-an inammatory disease. N Engl J Med 340:
115-126.
97. O’Keefe JH, Carter MD, Lavie CJ (2009) Primary and secondary prevention
of cardiovascular diseases: a practical evidence-based approach. Mayo Clin
Proc 84: 741-757.
98. Jadidi E, Mohammadi M, Moradi T (2013) High risk of cardiovascular diseases
after diagnosis of multiple sclerosis. Mult Scler 19: 1336-1340.
99. Tseng CH, Huang WS, Lin CL, Chang YJ. (2015) Increased risk of ischaemic
stroke among patients with multiple sclerosis. Eur J Neurol 22: 500-506.
100. Roshanisefat H, Bahmanyar S, Hillert J, Olsson T, Montgomery S (2014)
Multiple sclerosis clinical course and cardiovascular disease risk - Swedish
cohort study. Eur J Neurol 21: 1353-1353e88.
101. Lindegard B (1985) Diseases associated with multiple sclerosis and epilepsy.
A population cohort study of 1,59,200 middle-aged, urban, native Swedes
observed over 10 years (1970-79). Acta Neurol Scand 71: 267-277.
102. Brønnum-Hansen H, Koch-Henriksen N, Stenager E (2004) Trends in survival
and cause of death in Danish patients with multiple sclerosis. Brain 127: 844-
850.
103. Marrie RA, Yu BN, Leung S, Elliott L, Caetano P, et al. (2013) Prevalence and
incidence of ischemic heart disease in multiple sclerosis: A population-based
validation study. Mult Scler Relat Disord 2: 355-361.
104. Zöller B, Li X, Sundquist J, Sundquist K (2012) Risk of pulmonary embolism
in patients with autoimmune disorders: a nationwide follow-up study from
Sweden. Lancet 379: 244-249.
105. Ramagopalan SV, Wotton CJ, Handel AE, Yeates D, Goldacre MJ (2011)
Risk of venous thromboembolism in people admitted to hospital with selected
immune-mediated diseases: record-linkage study. BMC Med 9: 1.
106. Christensen S, Farkas DK, Pedersen L, Miret M, Christiansen CF, et al. (2012)
Multiple sclerosis and risk of venous thromboembolism: a population-based
cohort study. Neuroepidemiology 38: 76-83.
107. Arpaia G, Bavera PM, Caputo D, Mendozzi L, Cavarretta R, et al. (2010) Risk
of deep venous thrombosis (DVT) in bedridden or wheelchair-bound multiple
sclerosis patients: a prospective study. Thromb Res 125: 315-317.
108. Umpleby H (2012) Autoimmune disorders increase the risk of developing
pulmonary embolism. Thorax 67: 907-912.
109. Peeters PJ, Bazelier MT, Uitdehaag BM, Leufkens HG, De Bruin ML, et al.
(2014) The risk of venous thromboembolism in patients with multiple sclerosis:
the Clinical Practice Research Datalink. J Thromb Haemost 12: 444-451.
110. Kaufman J, Khatri BO, Riendl P (1988) Are patients with multiple sclerosis
protected from thrombophlebitis and pulmonary embolism? Chest 94: 998-
1001.
111. Poser CM (1986) Pathogenesis of multiple sclerosis. A critical reappraisal.
Acta Neuropathol 71: 1-10.
112. Rae-Grant AD, Wong C, Bernatowicz R, Fox RJ (2014) Observations on the
brain vasculature in multiple sclerosis: A historical perspective. Mult Scler
Relat Disord 3: 156-162.
113. Werner P, Pitt D, Raine CS (2001) Multiple sclerosis: altered glutamate
homeostasis in lesions correlates with oligodendrocyte and axonal damage.
Ann Neurol 50: 169-180.
114. Minagar A, Jy W, Jimenez JJ, Alexander JS (2006) Multiple sclerosis as a
vascular disease. Neurol Res 28: 230-235.
115. Frohman EM, Racke MK, Raine CS (2006) Multiple sclerosis-the plaque and
its pathogenesis. N Engl J Med 354: 942-955.
116. Haider L, Fischer MT, Frischer JM, Bauer J, Höftberger R, et al. (2011)
Oxidative damage in multiple sclerosis lesions. Brain 134: 1914-1924.
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016) Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med
Surg 4: 259. doi:10.4172/2329-6925.1000259
Page 11 of 12
Volume 4 • Issue 2 • 1000259
J Vasc Med Surg
ISSN: 2329-6925 JVMS, an open access journal
117. van Horssen J, Witte ME, Schreibelt G, de Vries HE (2011) Radical changes in
multiple sclerosis pathogenesis. Biochim Biophys Acta 1812: 141-150.
118. Yanagisawa M, Kurihara H, Kimura S, Tomobe Y, Kobayashi M, et al. (1988)
A novel potent vasoconstrictor peptide produced by vascular endothelial cells.
Nature 332: 411-415.
119. D’haeseleer M, Beelen R, Fierens Y, Cambron M, Vanbinst AM, et al. (2013)
Cerebral hypoperfusion in multiple sclerosis is reversible and mediated by
endothelin-1. Proc Natl Acad Sci U S A 110: 5654-5658.
120. Jankowska-Lech I, Terelak-Borys B, Grabska-Liberek I, Palasik W, Bik W, et
al. (2015) Decreased endothelin-1 plasma levels in multiple sclerosis patients:
a possible factor of vascular dysregulation?. Med Sci Monit 2: 1066-1071.
121. Zivadinov R, Weinstock-Guttman B, Pirko I (2011) Iron deposition and
inammation in multiple sclerosis. Which one comes rst? BMC Neuroscience
20: 60-65.
122. Hammond KE, Metcalf M, Carvajal L, Okuda DT, Srinivasan R, et al. (2008)
Quantitative in vivo magnetic resonance imaging of multiple sclerosis at 7
Tesla with sensitivity to iron. Ann Neurol 64: 707-713.
123. Hagemeier J, Weinstock-Guttman B, Heininen-Brown M, Poloni GU,
Bergsland N, et al. (2013) Gray matter SWI-ltered phase and atrophy are
linked to disability in MS. Front Biosci (Elite Ed) 5: 525-532.
124. Kira J (2014) (Disease concept, etiology and mechanisms of multiple sclerosis).
Nihon Rinsho 72: 1884-1894.
125. Zhou Y, Taylor B, van der Mei I, Stewart N, Charlesworth J, et al. (2015)
Genetic variation in PBMC-produced IFN-γ and TNF-α ± associations with
relapse in multiple sclerosis. J Neurol Sci 349: 40-44.
126. Hashioka S, McGeer EG, Miyaoka T, Wake R, Horiguchi J, et al. (2015)
Interferon-γ-induced neurotoxicity of human astrocytes. CNS Neurol Disord
Drug Targets 14: 251-256.
127. Sheremata WA, Jy W, Horstman LL, Ahn YS, Alexander JS, et al. (2008)
Evidence of platelet activation in multiple sclerosis. J Neuroinammation 5: 27.
128. Wuerfel J, Paul F, Zipp F (2007) Cerebral blood perfusion changes in multiple
sclerosis. J Neurol Sci 259: 16-20.
129. Wuerfel J, Bellmann-Strobl J, Brunecker P, Aktas O, McFarland H, et al.
(2004) Changes in cerebral perfusion precede plaque formation in multiple
sclerosis: a longitudinal perfusion MRI study. Brain 127: 111-119.
130. Rosso C, Remy P, Creange A, Brugieres P, Cesaro P, et al. (2006) Diffusion-
weighted MR imaging characteristics of an acute strokelike form of multiple
sclerosis. AJNR Am J Neuroradiol 27: 1006-1008.
131. Law M, Saindane AM, Ge Y, Babb JS, Johnson G, et al. (2004) Microvascular
abnormality in relapsing-remitting multiple sclerosis: perfusion MR imaging
ndings in normal-appearing white matter. Radiology 231: 645-652.
132. Inglese M, Park SJ, Johnson G, Babb JS, Miles L, et al. (2007) Deep gray
matter perfusion in multiple sclerosis: dynamic susceptibility contrast perfusion
magnetic resonance imaging at 3 T. Arch Neurol 64: 196-202.
133. Varga AW, Johnson G, Babb JS, Herbert J, Grossman RI, et al. (2009) White
matter hemodynamic abnormalities precede sub-cortical gray matter changes
in multiple sclerosis. J Neurol Sci 282: 28-33.
134. Inglese M, Adhya S, Johnson G, Babb JS, Miles L, et al. (2008) Perfusion
magnetic resonance imaging correlates of neuropsychological impairment in
multiple sclerosis. J Cereb Blood Flow Metab 28: 164-171.
135. Saindane AM, Law M, Ge Y, Johnson G, Babb JS, et al. (2007) Correlation
of diffusion tensor and dynamic perfusion MR imaging metrics in normal-
appearing corpus callosum: support for primary hypoperfusion in multiple
sclerosis. AJNR Am J Neuroradiol 28: 767-772.
136. Holland CM, Charil A, Csapo I, Liptak Z, Ichise M, et al. (2012) The relationship
between normal cerebral perfusion patterns and white matter lesion distribution
in ,249 patients with multiple sclerosis. J Neuroimaging 22: 129-136.
137. Cruveilhier J (1829-1842) Anatomie pathologique du corps humain. Bailliere,
Paris, France.
138. Reindeisch E (1863) Histologisches detail zu der grauen degeneration von
gehirn und rueckenmark. Arch Path Anat Physiol Klin Med 26: 474-483.
139. Tracy J, Putnam MD (1937) Evidences of vascular occlusion in multiple
sclerosis and “encephalomyelitis”. Arch Neur Psych 37: 1298-1321.
140. Fog T (1963) On the vessel-plaque relations in the brain in multiple sclerosis.
Acta Psychiat Neurol Scand 39: 257-262.
141. Allen IV (1981) The pathology of multiple sclerosis-fact, ction and hypothesis.
Neuropathol Appl Neurobiol 7: 169-182.
142. Zamboni P (2006) The big idea: iron-dependent inammation in venous
disease and proposed parallels in multiple sclerosis. J R Soc Med 99: 589-
593.
143. Ge Y, Zohrabian VM, Osa EO, Xu J, Jaggi H, et al. (2009) Diminished visibility
of cerebral venous vasculature in multiple sclerosis by susceptibility-weighted
imaging at 3.0 Tesla. J Magn Reson Imaging 29: 1190-1194.
144. Ge Y, Law M, Johnson G, Herbert J, Babb JS, et al. (2005) Dynamic
susceptibility contrast perfusion MR imaging of multiple sclerosis lesions:
characterizing hemodynamic impairment and inammatory activity. AJNR Am
J Neuroradiol 26: 1539-1547.
145. Singh AV, Zamboni P (2009) Anomalous venous blood ow and iron deposition
in multiple sclerosis. J Cereb Blood Flow Metab 29: 1867-1878.
146. Zamboni P, Galeotti R, Menegatti E, Malagoni AM, Tacconi G, et al. (2009)
Chronic cerebrospinal venous insufciency in patients with multiple sclerosis.
J Neurol Neurosurg Psychiatry 80: 392-399.
147. Fox RJ, Rae-Grant A (2011) Chronic cerebrospinal venous insufciency: have
we found the cause and cure of MS? Neurology 77: 98-100.
148. Doepp F, Paul F, Valdueza JM, Schmierer K, Schreiber SJ (2010) No
cerebrocervical venous congestion in patients with multiple sclerosis. Ann
Neurol 68: 173-183.
149. Zamboni P, Morovic S, Menegatti E, Viselner G, Nicolaides AN (2011)
Screening for chronic cerebrospinal venous insufciency (CCSVI) using
ultrasound-recommendations for a protocol. Int Angiol 30: 571-597.
150. Laupacis A, Lillie E, Dueck A, Straus S, Perrier L, et al. (2011) Association
between chronic cerebrospinal venous insufciency and multiple sclerosis: a
meta-analysis. CMAJ 183: E1203-1212.
151. Zamboni P, Menegatti E, Galeotti R, Malagoni AM, Tacconi G, et al. (2009)
The value of cerebral Doppler venous haemodynamics in the assessment of
multiple sclerosis. J Neurol Sci 282: 21-27.
152. Sundström P, Wåhlin A, Ambarki K, Birgander R, Eklund A, et al. (2010)
Venous and cerebrospinal uid ow in multiple sclerosis: a case-control study.
Ann Neurol 68: 255-259.
153. Baracchini C, Perini P, Calabrese M, Causin F, Rinaldi F, et al. (2011) No
evidence of chronic cerebrospinal venous insufciency at multiple sclerosis
onset. Ann Neurol 69: 90-99.
154. Mayer CA, Pfeilschifter W, Lorenz MW, Nedelmann M, Bechmann I, et
al. (2011) The perfect crime? CCSVI not leaving a trace in MS. J Neurol
Neurosurg Psychiatry 82: 436-440.
155. Dolic K, Siddiqui AH, Karmon Y, Marr K, Zivadinov R. (2013) The role of
noninvasive and invasive diagnostic imaging techniques for detection of extra-
cranial venous system anomalies and developmental variants. BMC Med.
156. Comi G, Battaglia MA, Bertolotto A, Del Sette M, Ghezzi A, et al. (2013)
CoSMo Collaborative Study Group. Observational case-control study of the
prevalence of chronic cerebrospinal venous insufciency in multiple sclerosis:
results from the CoSMo study. Mult Scler 19:1508-1517.
157. Lanzillo R, Mancini M, Liuzzi R, Di Donato O, Salvatore E, et al. (2013) Chronic
cerebrospinal venous insufciency in multiple sclerosis: a highly prevalent
age-dependent phenomenon. BMC Neurol.
158. Ciciarello F, Mandolesi S, Galeandro AI, Marceca A, Rossi M, et al. (2014)
Age-related vascular differences among patients suffering from multiple
sclerosis. Curr Neurovasc Res 11: 23-30.
159. Ciccone MM, Galeandro AI, Scicchitano P, Zito A, Gesualdo M et al. (2012)
Multigate quality Doppler proles and morphological/hemodynamic alterations
in multiple sclerosis patients. Curr Neurovasc Res 9: 120-127.
160. Pascolo L, Gianoncelli A, Rizzardi C, Tisato V, Salomé M, et al. (2014)
Calcium micro-depositions in jugular truncular venous malformations revealed
by Synchrotron-based XRF imaging. Sci Rep 4: 6540.
161. Zamboni P, Tisato V, Menegatti E, Mascoli F, Gianesini S, et al. (2014)
Ultrastructure of internal jugular vein defective valves. Phlebology 30: 644-
647.
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016) Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med
Surg 4: 259. doi:10.4172/2329-6925.1000259
Page 12 of 12
Volume 4 • Issue 2 • 1000259
J Vasc Med Surg
ISSN: 2329-6925 JVMS, an open access journal
162. Ryu JK, Petersen MA, Murray SG, Baeten KM, Meyer-Franke A et al.
(2015) Blood coagulation protein brinogen promotes autoimmunity and
demyelination via chemokine release and antigen presentation. Nat Commun.
163. Acheson ED, Bachrach CA, Wright FM (1960) Some comments on the
relationship of the distribution of multiple sclerosis to latitude, solar radiation,
and other variables. Acta Psychiatr Scand Suppl 35: 132-147.
164. Kurtzke JF, Hyllested K (1987) Multiple sclerosis in the Faroe Islands. III. An
alternative assessment of the three epidemics. Acta Neurol Scand 76: 317-339.
165. Miller DH, Hammond SR, McLeod JG, Purdie G, Skegg DC (1990) Multiple
sclerosis in Australia and New Zealand: are the determinants genetic or
environmental? J Neurol Neurosurg Psychiatry 53: 903-905.
166. Simpson S Jr, Blizzard L, Otahal P, Van der Mei I, Taylor B (2011) Latitude
is signicantly associated with the prevalence of multiple sclerosis: a meta-
analysis. J Neurol Neurosurg Psychiatry 82: 1132-1141.
167. Kurtzke JF (1993) Epidemiologic evidence for multiple sclerosis as an
infection. Clin Microbiol Rev 6: 382-427.
168. Cuzner ML (1980) Annotation. Recent biochemical and immunological
observations in multiple sclerosis. Neuropathol Appl Neurobiol 6: 405-414.
169. Adams CW, Poston RN, Buk SJ, Sidhu YS, Vipond H (1985) Inammatory
vasculitis in multiple sclerosis. J Neurol Sci 69: 269-283.
170. Westall FC (2006) Molecular mimicry revisited: gut bacteria and multiple
sclerosis. J Clin Microbiol 44: 2099-2104.
171. Steiner I, Nisipianu P, Wirguin I (2001) Infection and the etiology and
pathogenesis of multiple sclerosis. Curr Neurol Neurosci Rep 1: 271-276.
172. Giovannoni G, Cutter GR, Lunemann J, Martin R, Münz C, et al. (2006)
Infectious causes of multiple sclerosis. Lancet Neurol 5: 887-894.
173. Mielcarz DW, Kasper LH (2015) The gut microbiome in multiple sclerosis. Curr
Treat Options Neurol 17: 344.
174. Petra AI, Panagiotidou S, Hatziagelaki E, Stewart JM, Conti P, et al. (2015)
Gut-Microbiota-Brain axis and its effect on neuropsychiatric disorders with
suspected immune dysregulation. Clin Ther 37: 984-995.
175. Bhargava P, Mowry EM (2014) Gut microbiome and multiple sclerosis. Curr
Neurol Neurosci Rep 14: 492.
176. Le Gac P (1960) The treatment of multiple sclerosis of rickettsial or neo-
rickettsial origin. J Med Bord 137: 577-589.
177. Sriram S, Mitchell W, Stratton C (1998) Multiple sclerosis associated with
Chlamydia pneumoniae infection of the CNS. Neurology 50: 571-572.
178. Parratt J, Tavendale R, O’Riordan J, Parratt D, Swingler R (2008) Chlamydia
pneumoniae-specic serum immune complexes in patients with multiple
sclerosis. Mult Scler 14: 292-299.
179. Grimaldi LME, Pincherle A, Martinelli-Boneschi F, Filippi M, Patti F, et al.
(2003) An MRI study of Chlamydia pneumoniae infection in Italian multiple
sclerosis patients. Mult Scler 9: 467-471.
180. Gutiérrez J, Koppel B, Kleiman A, Akrat G (2008) Multiple sclerosis and
Epstein-Barr virus: a growing association. Rev Med Inst Mex Seguro Soc
46: 639-642.
181. Abdelrahman HS, Selim HS, Hashish MH, Sultan LI (2014) Epstein-Barr virus
in multiple sclerosis. J Egypt Public Health Assoc 89: 90-95.
182. Leibovitch EC, Jacobson S2 (2014) Evidence linking HHV-6 with multiple
sclerosis: an update. Curr Opin Virol 9: 127-133.
183. Broccolo F, Fusetti L, Ceccherini-Nelli L (2013) Possible role of human
herpesvirus 6 as a trigger of autoimmune disease. ScienticWorldJournal
2013: 867389.
184. Ferrò MT, Franciotta D, Prelle A, Bestetti A, Cinque P (2012) Active intrathecal
herpes simplex virus type 1 (HSV-1) and human herpesvirus-6 (HHV-6)
infection at onset of multiple sclerosis. J Neurovirol 18: 437-440.
185. Stratton CW, Wheldon DB (2006) Multiple sclerosis: an infectious syndrome
involving Chlamydophila pneumoniae. Trends Microbiol 14: 474-479.
186. Thibault PK (2012) Multiple sclerosis: a chronic infective cerebrospinal
venulitis? Phlebology 27: 207-218.
187. Ransohoff RM, Haer DA, Lucchinetti CF (2015) Multiple sclerosis-a quiet
revolution. Nat Rev Neurol 11: 134-142.
188. Christiansen CF1 (2012) Risk of vascular disease in patients with multiple
sclerosis: a review. Neurol Res 34: 746-753.
189. Wei L, MacDonald TM, Walker BR (2004) Taking glucocorticoids by
prescription is associated with subsequent cardiovascular disease. Ann Intern
Med 141: 764-770.
190. Christiansen CF, Christensen S, Mehnert F, Cummings SR, Chapurlat RD, et
al. (2009) Glucocorticoid use and risk of atrial brillation or utter: a population-
based, case-control study. Arch Intern Med 169: 1677-1683.
191. Sonnenblick M, Rosin A (1991) Cardiotoxicity of interferon. A review of 44
cases. Chest 99: 557-561.
192. Le Page E, Leray E, Edan G. (2011) Long-term safety prole of mitoxantrone
in a French cohort of 802 multiple sclerosis patients: a 5-year prospective
study. Mult Scler 17: 867-875.
193. Kappos L, Radue EW, O’Connor P, Polman C, Hohlfeld R, et al. (2010) A
placebo-controlled trial of oral ngolimod in relapsing multiple sclerosis. N
Engl J Med 362: 387-401.
194. Pelletier D, Haer DA (2012) Fingolimod for multiple sclerosis. N Engl J Med
366: 339-347.
195. Schwarz A, Korporal M, Hosch W, Max R, Wildemann B (2010) Critical
vasospasm during ngolimod (FTY720) treatment in a patient with multiple
sclerosis. Neurology 74: 2022-2024.
196. Sheremata WA, Taylor JR, Elgart GW (1995) Severe necrotizing cutaneous
lesions complicating treatment with interferon beta-1b. N Engl J Med 332:
1584.
197. Elgart GW, Sheremata W, Ahn YS (1997) Cutaneous reactions to recombinant
human interferon beta-1b: the clinical and histologic spectrum. J Am Acad
Dermatol 37: 553-558.
198. Samuel L, Lowenstein EJ (2006) Recurrent injection site reactions from
interferon beta 1-b. J Drugs Dermatol 5: 366-367.
Citation: Caprio MG, Russo C, Giugliano A, Ragucci M, Mancini M (2016)
Vascular Disease in Patients with Multiple Sclerosis: A Review. J Vasc Med
Surg 4: 259. doi:10.4172/2329-6925.1000259
OMICS International: Publication Benefits & Features
Unique features:
• Increasedglobalvisibilityofarticlesthroughworldwidedistributionandindexing
• Showcasingrecentresearchoutputinatimelyandupdatedmanner
• Specialissuesonthecurrenttrendsofscienticresearch
Special features:
• 700+OpenAccessJournals
• 50,000+Editorialteam
• Rapidreviewprocess
• Qualityandquickeditorial,reviewandpublicationprocessing
• Indexingatmajorindexingservices
• SharingOption:SocialNetworkingEnabled
• Authors,ReviewersandEditorsrewardedwithonlineScienticCredits
• Betterdiscountforyoursubsequentarticles
Submityourmanuscriptat:www.editorialmanager.com/lifesciences
... The cellular and molecular mechanisms behind this plasticity are still largely unknown. However, a large body of literature describes an alteration of the vascular system and of the neurovascular unit in MS ( Caprio & Russo, 2016), opening the debate on whether vascular events may be the primary cause of neurological diseases or rather a mere participant recruited from a primary neuronal origin. Our findings suggest that demyelination itself, caused by oligodendrocyte disruption, can promote neurovascular coupling alterations. ...
... Given that these changes closely mimic those observed in the early phases of multiple sclerosis (MS) ( Rocca et al., 2022), we hypothesize that this measurement is a critical indicator of the vascular dysfunction inherent to the model. As highlighted by various studies ( Caprio & Russo, 2016;Cashion et al., 2023;Sweeney et al., 2018), MS patients frequently experience vascular complications, such as blood-brain barrier leakage ( Sweeney et al., 2019), microbleeds, and reduced cerebral blood flow. A growing body of evidence suggests that the dysfunction of various cells within the neurovascular unit-such as endothelial cells ( Zlokovic, 2008), astrocytes ( Correale & Farez, 2015), pericytes ( Zhang et al., 2020), and microglial cells ( Lassmann et al., 2012)-plays a significant role in MS pathology ( Ballabh et al., 2004;Cashion et al., 2023;Sweeney et al., 2019). ...
Article
Full-text available
Multiple Sclerosis (MS) is an autoimmune disease of the central nervous system (CNS), affecting 2.8 million people worldwide, that presents multiple features, one of which is demyelination. Although treatments exist to manage the condition, no cure has been found to stop the progression of neurodegeneration. To develop new treatments and investigate the multiple systems impacted by MS, new imaging technologies are needed at the preclinical stage. Functional ultrasound imaging (fUS) has recently emerged as a robust method to measure brain cerebral blood volume (CBV) dynamics as an indirect indicator of neural activity. This study aimed to quantify the amplitude of alteration of evoked hemodynamic response in the somatosensory cortex, and its potential link with demyelination in a mouse model of CNS demyelination induced by cuprizone. We demonstrate that extended demyelination leads to an increased hemodynamic response in the primary sensory cortex, both spatially and temporally, aligning with fMRI findings in MS patients. Second, using descriptors of the evoked cortical hemodynamic response, we demonstrate that certain parameters (the number of active pixels and the rise time) correlate with the level of Myelin Basic Protein in the primary sensory cortex and the thalamus, when taken together. Interestingly, the increased CBV is not associated with demyelination but instead reflects the well-documented vascular alteration described in MS. Moreover, these changes were absent in the thalamus, and in focalized demyelinated lesions induced by lysolecithin injection, suggesting the involvement of specific cortical mechanisms driven by oligodendrocyte depletion. In conclusion, our study introduces a novel, non-invasive functional approach for investigating vascular dysfunction in the context of MS, addressing an important yet understudied aspect in both pre-clinical and clinical research.
... In the 1980s, Adams et al. pioneered the demonstration of vasculitis within the walls of the veins and small veins near active MS lesions (11). Recent evidence suggests that vascular components may be the initiating triggers for neuronal pathology and subsequent neurological manifestations of the disease (12). ...
... In the examination of deep medullary veins (DMVs), SWI is capable of noninvasively and visually imaging cerebral veins and assessing venous fluid oxygenation (12). SWI is now recognized as a sensitive method for imaging small intracranial veins and is able to visualize DMVs on 3-T MRI (17). ...
Article
Full-text available
Background: Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS). Recent studies have shown that different forms of vascular abnormalities may be related to the pathogenesis of MS. Susceptibility-weighted imaging (SWI) can directly image intracranial venules. The aim of this study was to investigate the association between deep medullary veins (DMVs) and the degree of neurodegeneration in patients with MS. Methods: In this prospective cross-sectional study, 34 patients with MS and 30 age-matched healthy controls (HCs) were recruited. The count and score of DMVs, which can reflect the visibility and continuity of DMVs were evaluated based on SWI. The differences between the group with a high DMV score (DMV >10) and the group with a low DMV score (DMV ≤10) were assessed. The association of DMV change with neurodegeneration neuroimaging markers [including amount and volume of white matter lesion (WML), degree of cortical atrophy, whole-brain atrophy, and deep gray matter (DGM) atrophy] and clinical Expanded Disability Status Scale (EDSS) were observed in patients with MS. Results: It was found that compared with controls, patients with MS (n=34) had a significantly lower DMV count (P<0.001) and a significantly higher DMV score (P<0.001). The low- and high-DMV score groups differed significantly in terms of EDSS (P=0.048) and neurodegeneration neuroimaging indicators, including WML volume (P=0.015), brain parenchymal fraction (BPF) (P=0.047), thalamic fraction (P=0.036), and caudate fraction (P=0.015). In the correlation analysis of the MS group, DMV count was negatively correlated with the number of WMLs (r=-0.535; P=0.001) and the WML volume (r=-0.416; P=0.014) but positively correlated with the neuroimaging measurements reflecting the degree of whole-brain atrophy and DGM atrophy. Furthermore, the DMV score was positively correlated with EDSS (r=0.450; P=0.008), number of WMLs (r=0.490; P=0.003), and WML volumes (r=0.635; P=0.001) but negatively correlated with the neuroimaging measurements reflecting the degree of whole-brain atrophy and DGM atrophy. Conclusions: Reduced DMV visibility and continuity could reflect the severity of neurodegeneration in patients with MS. DMV count and score may be imaging indicators for assessing the severity of MS.
... Considering modifiable risk factors for stroke, it is more frequent to smoke for patients diagnosed with MS and also be obese than the general population, however, there is a dearth of information on alcohol intake. 11,42,44,45 Even in the early stages of MS when disability is moderate, the reduced physical activity of MS patients might activate coagulation pathways, increasing the risk for venous thromboembolism (VTE) and subclinical atherosclerosis. 42 Low levels of vitamin D and infections are among the additional risk factors that exhibit independent associations with the progression and outcomes of MS or the risk for stroke caused by bacteria or viruses. ...
... 42 Low levels of vitamin D and infections are among the additional risk factors that exhibit independent associations with the progression and outcomes of MS or the risk for stroke caused by bacteria or viruses. 13,45 Patients with MS had greater rates of infection incidence, hospitalization associated with infection, and infection-related mortality. 46,47 F I G U R E 1 Depicts astrocytes, blood-brain barrier (BBB) failure, and M1/M2 polarization as common routes in the etiology of multiple sclerosis and ischemic stroke. ...
Article
Full-text available
Background Multiple sclerosis (MS) is a chronic immune‐mediated disorder characterized by the degradation of the myelin sheath in the central nervous system. Research indicates that individuals with MS exhibit a higher susceptibility to stroke compared to the general population. This association is rooted in shared underlying mechanisms, specifically involving neuroinflammatory processes. Methodology We performed an extensive search on PubMed, MEDLINE, Embase, Scopus, and Google Scholar using specific terms. The search terms included variations of “multiple sclerosis,” “stroke,” “cerebrovascular disease,” “vascular risk factors,” “disease‐modifying therapies,” and “neuroinflammation.” The search was limited to articles published from January 1, 2000, up to 31 May, 2023. Results and Discussion Stroke, a global health burden characterized by significant mortality and adult disability, underscores the critical importance of understanding the link between MS and stroke. Despite a growing body of research establishing an elevated risk of stroke in MS patients, notable information gaps persist. Limited prospective multicenter studies on stroke incidence in MS patients contribute to an incomplete understanding of the precise relationship between these two conditions. Conclusion In conclusion, this review underscores the critical need for a thorough understanding of the complex relationship between MS and stroke. The identified risk factors and the influence of MS DMTs on stroke risk necessitate further investigation to inform evidence‐based preventive and therapeutic strategies. Bridging the existing information gaps through prospective multicenter studies is imperative for a comprehensive understanding of this association. The development of targeted diagnostic and therapeutic approaches for acute stroke risk in MS patients is paramount to mitigate the impact of these debilitating conditions. Ultimately, this review serves as a foundation for future efforts to enhance preventative measures and therapeutic interventions, thereby improving the overall quality of life for individuals with MS susceptible to strokes.
... Current literature indicates that MS is a complex condition with causation likely linked to both genetic and environmental factors (Noseworthy 1999). The progression of MS is variable, manifesting in both progressive and relapsing-remitting forms (Caprio et al. 2016). A pathological component of the disease is the formation of areas of axonal demyelination, which form white matter lesions in the CNS (Wingerchuk et al. 2001). ...
Article
Full-text available
This study aimed to compare computational fluid dynamics (CFD) results to those acquired in vivo with 4D Flow magnetic resonance imaging (MRI) and in vitro with a 3D printed model using pressure catheter manometry. The goal was to investigate the haemodynamics of the cerebral venous system (CVS) and assess the accuracy of the methodologies, to highlight any discrepancies between the techniques. One participant living with multiple sclerosis (MS) and one healthy control were recruited for this study. MRI was performed to generate 3D geometries of the anatomy and to compute blood flow rates at the boundaries, with 4D Flow MRI velocity streamlines for the control participant. CFD models were created for the two participants and simulated using the patient-specific boundary conditions. A 3D printed geometry of the MS participant was created and a flow loop experiment was conducted to measure the cerebral venous pressures. The venous pressures were found to be comparable to that observed in the CFD simulation. 4D Flow MRI velocity streamlines of the CVS were found to correspond well to the CFD findings, except for a few regions, which were likely impacted by the low resolution of the MRI. The use of all three methods enabled the successful validation of the velocity, flow features and pressure, and ensured that the haemodynamics of the CVS as resolved using CFD, were accurate. This highlights the potential for increased efficacy of the clinical outcomes of future studies that utilise such methods. Graphical abstract
... J. Lechner-Scott is with the School of Medicine and Public Health, in the College of Health, Medicine and Wellbeing at the University of Newcastle, of MS has not been fully determined, with a restricted understanding of the factors which impact exactly how the disease progresses [2], although several contributing aspects have been identified including the presence of the Epstein-Barr virus (EBV) [3]. The evolution of the disorder is variable, with both progressive and relapsing-remitting forms [4]. The present literature shows that MS is a complex condition and appears to have a causation associated with genetic and environmental factors [2]. ...
Article
Full-text available
Objective: An investigation was performed to determine the relevant hemodynamic parameters which could help assess vascular pathology in human diseases. Using these parameters, this study aims to assess if there are any hemodynamic differences in the cerebral veins of multiple sclerosis (MS) patients and controls which could impact the etiology of MS. Methods: 40 MS participants and 20 controls were recruited for this study. Magnetic resonance imaging (MRI) was performed to enable 3D geometries of the anatomy and the blood flow rates at the boundaries to be computed. Computational fluid dynamics (CFD) models were created for each participant and simulated using patient-specific boundary conditions. Results: The pressure drop and vascular resistance did not significantly differ between the groups. The internal jugular vein (IJV) cross-sectional area was larger in the MS group (Right IJV: p = 0.04, Left IJV: p = 0.02) and the straight sinus (ST) flow rate was higher in MS across all ages (p = 0.005) compared to controls. Vascular resistance was shown to indicate regions in the cerebral veins which could correspond to increased venous pressure. Conclusion & Significance: This study shows that the pressure and vascular resistance of the cerebral veins are unlikely to be directly related to the etiology of MS. The finding of higher ST flow could correspond to increased inflammation in the deep venous system. Resistance as a measure of vascular pathology shows promise and could be useful to holistically investigate blood flow hemodynamics in a variety of other diseases of the circulatory system.
... The cellular and molecular mechanisms behind this plasticity are still largely unknown. There is, however, a large literature describing an alteration of the vascular system and of the neurovascular unit in MS [35], opening the debate on whether vascular events may be the primary cause of neurological diseases or rather a mere participant recruited from a primary neuronal origin. Today, our results suggest that demyelination, induced by oligodendrocyte disruption, is able by itself to promote such alteration of the neurovascular coupling. ...
Preprint
Full-text available
Multiple Sclerosis (MS) is an autoimmune disease of the central nervous system (CNS), affecting 2.8 million people worldwide, that presents multiple features, one of which is demyelination. Although treatments exist to manage the condition, no cure has been found to stop the progression of neurodegeneration. To develop new treatments and investigate the multiple systems impacted by MS, new imaging technologies are needed at the preclinical stage. Functional ultrasound imaging (fUS) has recently been demonstrated to robustly measure brain cerebral blood volume (CBV) dynamics as an indirect measure of neural activity. This study aimed at proposing a new biomarker of de- and/or re- myelination in a mouse model of MS induced by cuprizone. We demonstrate first that extended demyelination induces an increased hemodynamic response in the primary sensory cortex both spatially and temporally, which is consistent with fMRI data collected on MS patients. Second, using descriptors of the evoked hemodynamic response, we show that 3 of these descriptors allows the prediction of the level of myelin in the primary sensory cortex (p=5. 10-5) and the thalamus (p=6. 10-6). The development of such a non-invasive biomarker is crucial in the MS field as is provides an extremely useful tool for both disease follow-up and drug development.
... The cellular and molecular mechanisms behind this plasticity are still largely unknown. There is, however, a large literature describing an alteration of the vascular system and of the neurovascular unit in MS [35], opening the debate on whether vascular events may be the primary cause of neurological diseases or rather a mere participant recruited from a primary neuronal origin. Today, our results suggest that demyelination, induced by oligodendrocyte disruption, is able by itself to promote such alteration of the neurovascular coupling. ...
Preprint
Full-text available
In this study, using functional ultrasound imaging, we develop a biomarker of the extend of demyelination in a mouse model of multiple sclerosis.
... Moreover, different studies suggest that endothelin-1, a vasoconstrictor peptide produced intrathecally, could be responsible for the vascular abnormalities [22]. Endothelin-1 levels were measured in MS patients and results showed that the internal jugular vein blood had significantly higher levels in MS patients compared to a healthy control group. ...
Article
Full-text available
The prevalence of multiple sclerosis (MS) has been increasing among young people in developing countries over the last years. With the continuous development of new technology, the diagnosis and follow-up of these patients has received new parameters that physicians may use in their practice. This paper reviews the main biomarkers identified through Optical Coherence Tomography Angiography (OCT-A) involved in the development and progression of MS and investigates the role it may have in detecting changes to the central nervous system (CNS).
... The chronic condition, multiple sclerosis (MS), that affects the central nervous system (CNS) is characterized by inflammation and damage to the protective myelin sheath around nerve fibers. The autoimmune theory of the disease has been widely accepted due to a significant amount of evidence supporting it [1][2][3] . But, ...
Article
Full-text available
Dysfunction in the epithelium, breakdown of the blood-brain barrier, and consequent leukocyte and T-cell infiltration into the central nervous system define Vascular Multiple Sclerosis. Multiple Sclerosis (MS) affects around 2.5 million individuals worldwide, is the leading cause of neurological impairment in young adults, and can have a variety of progressions and consequences. Despite significant discoveries in immunology and molecular biology, the root cause of MS is still not fully understood, as do the immunological triggers and causative pathways. Recent research into vascular anomalies associated with MS suggests that a vascular component may be pivotal to the etiology of MS, and there can be actually a completely new entity in the already available classification of multiple sclerosis, that can be called ‘Vascular multiple sclerosis’. Unlike the usual other causes of MS, Vascular MS is not dependent on autoimmune pathophysiologic mechanisms, instead, it is caused due to the blood vessels pathology. This review aims to thoroughly analyze existing information and updates about the scattered available findings of genetics, pro-angiogenetic factors, and vascular abnormalities in this important spectrum, the vascular facets of multiple sclerosis.
Article
Full-text available
Thrombosis, the formation of blood clots in arteries or veins, poses a significant health risk by disrupting the blood flow. It can potentially lead to major cardiovascular complications such as acute myocardial infarction or ischemic stroke (arterial thrombosis) and deep vein thrombosis or pulmonary embolism (venous thrombosis). Nevertheless, over the course of several decades, researchers have observed an association between different cardiovascular events and neurodegenerative diseases, which progressively harm and impair parts of the nervous system, particularly the brain. Furthermore, thrombotic complications have been identified in numerous clinical instances of neurodegenerative diseases, particularly Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and Huntington’s disease. Substantial research indicates that endothelial dysfunction, vascular inflammation, coagulation abnormalities, and platelet hyperactivation are commonly observed in these conditions, collectively contributing to an increased risk of thrombosis. Thrombosis can, in turn, contribute to the onset, pathogenesis, and severity of these neurological disorders. Hence, this concise review comprehensively explores the correlation between cardiovascular diseases and neurodegenerative diseases, elucidating the cellular and molecular mechanisms of thrombosis in these neurodegenerative diseases. Additionally, a detailed discussion is provided on the commonly employed antithrombotic medications in the context of these neuronal diseases. Graphical Abstract
Article
Full-text available
Autoimmunity and macrophage recruitment into the central nervous system (CNS) are critical determinants of neuroinflammatory diseases. However, the mechanisms that drive immunological responses targeted to the CNS remain largely unknown. Here we show that fibrinogen, a central blood coagulation protein deposited in the CNS after blood–brain barrier disruption, induces encephalitogenic adaptive immune responses and peripheral macrophage recruitment into the CNS leading to demyelination. Fibrinogen stimulates a unique transcriptional signature in CD11b+ antigen-presenting cells inducing the recruitment and local CNS activation of myelin antigen-specific Th1 cells. Fibrinogen depletion reduces Th1 cells in the multiple sclerosis model, experimental autoimmune encephalomyelitis. Major histocompatibility complex (MHC) II-dependent antigen presentation, CXCL10- and CCL2-mediated recruitment of T cells and macrophages, respectively, are required for fibrinogen-induced encephalomyelitis. Inhibition of the fibrinogen receptor CD11b/CD18 protects from all immune and neuropathologic effects. Our results show that the final product of the coagulation cascade is a key determinant of CNS autoimmunity.
Article
Full-text available
Gut microbiota regulate intestinal function and health. However, mounting evidence indicates that they can also influence the immune and nervous systems and vice versa. This article reviews the bidirectional relationship between the gut microbiota and the brain, termed the microbiota-gut-brain (MGB) axis, and discusses how it contributes to the pathogenesis of certain disorders that may involve brain inflammation. Articles were identified with a search of Medline (starting in 1980) by using the key words anxiety, attention-deficit hypersensitivity disorder (ADHD), autism, cytokines, depression, gut, hypothalamic-pituitary-adrenal (HPA) axis, inflammation, immune system, microbiota, nervous system, neurologic, neurotransmitters, neuroimmune conditions, psychiatric, and stress. Various afferent or efferent pathways are involved in the MGB axis. Antibiotics, environmental and infectious agents, intestinal neurotransmitters/neuromodulators, sensory vagal fibers, cytokines, and essential metabolites all convey information to the central nervous system about the intestinal state. Conversely, the hypothalamic-pituitary-adrenal axis, the central nervous system regulatory areas of satiety, and neuropeptides released from sensory nerve fibers affect the gut microbiota composition directly or through nutrient availability. Such interactions seem to influence the pathogenesis of a number of disorders in which inflammation is implicated, such as mood disorder, autism-spectrum disorders, attention-deficit hypersensitivity disorder, multiple sclerosis, and obesity. Recognition of the relationship between the MGB axis and the neuroimmune systems provides a novel approach for better understanding and management of these disorders. Appropriate preventive measures early in life or corrective measures such as use of psychobiotics, fecal microbiota transplantation, and flavonoids are discussed. Copyright © 2015 Elsevier HS Journals, Inc. All rights reserved.
Article
Recent experimental work¹ has made it seem probable that the lesions of multiple sclerosis and also those of the forms of "disseminated encephalomyelitis" which seem to represent a more acute stage of the same process² are produced by a local circulatory disturbance, apparently of the nature of an obstruction on the venous side. According to this point of view, there must be a primary change in the contents of the vessels of the central nervous system, or possibly of the intimal lining, which leads to thrombosis of venules. This produces local passive congestion and a mild degenerative process which affects myelin sheaths more than other structures. The myelin degenerates and is phagocytosed. The "inflammatory" phenomena would then be regarded as secondary or symptomatic and the gliosis as reparative or reactive. This hypothesis obviously must remain largely a speculation as long as it rests on the results of experimentation
Article
Information about metabolic comorbidities in patients with multiple sclerosis (MS) is scarce. Our aim was to examine the prevalence of the metabolic syndrome (MetS) and its components in patients with long duration of MS and significant disability. Demographic and clinical data, weight, height, waist circumference, blood pressure, and levels of fasting glucose, triglycerides and high density lipoprotein cholesterol (HDL-C) were obtained from 130 MS patients with Extended Disability Status Scale (EDSS) score ≥3.0. Seventy-two percent were female, mean ± SD age 55.8 ± 6.0, range 45-65 years, disease duration 18.2 ± 10.1 years, EDSS 5.5 ± 1.0. Obesity [body mass index (BMI) ≥ 30 kg/m(2) ] was present in 18.5% and overweight (BMI 25.0-29.9 kg/m(2) ) in 34.6%. The prevalence of the MetS was 30% with no gender difference. Fifty-six percent had central obesity by waist circumference, 28% treated hypertension, 45.8% elevated blood pressure, 11% type 2 diabetes mellitus, 31.4% treated dyslipidemia, 28.8% elevated triglyceride levels and 31.4% had low HDL-C. MS patients with MetS were significantly older (59.0 ± 5.5 vs. 53.8 ± 5.5, P < 0.0001) and heavier (BMI 29.0 ± 6.9 vs. 25.1 ± 4.7, P = 0.0009). There were no differences between the groups in neurological disability by the EDSS (5.7 ± 1.0 vs. 5.4 ± 1.0), disease duration (18.4 ± 9.9 vs. 18.2 ± 10.2 years) and number of steroid courses received (6.6 ± 9.5 vs. 6.3 ± 8.4). Compared to the general population, adult disabled MS patients had lower rates of obesity and overweight, as assessed by BMI. Despite these reduced rates, the prevalence of the MetS was similar to the general population. Specifically higher rates of increased waist circumference were found, suggesting that the lower BMI may be misleading in terms of health risk. © 2015 EAN.
Article
Cardiovascular risk factors can increase the risk of multiple sclerosis (MS) and modify its course. However, such factors possibly interact, determining a global cardiovascular risk. Our aim was to compare the global cardiovascular risk of subjects with and without MS with the simplified 10-year Framingham General Cardiovascular Disease Risk Score (FR) and to evaluate its importance on MS-related outcomes. Age, gender, smoking status, body mass index, systolic blood pressure, type II diabetes and use of antihypertensive medications were recorded in subjects with and without MS to estimate the FR, an individualized percentage risk score estimating the 10-year likelihood of cardiovascular events. In total, 265 MS subjects were identified with 530 matched controls. A t test showed similar FR in cases and controls (P = 0.212). Secondary progressive MS presented significantly higher FR compared to relapsing-remitting MS (P < 0.001). Linear regression analysis showed a direct relationship between FR and Expanded Disability Status Scale (P < 0.001) and MS Severity Scale (P < 0.001). The FR, evaluating the global cardiovascular health by the interaction amongst different risk factors, relates to MS disability, severity and course. © 2015 EAN.