Cholesterol Was Healthy in the End

Simopoulos AP, De Meester F (eds): A Balanced Omega-6/ Omega-3 Fatty Acid Ratio, Cholesterol and Coronary
Heart Disease. World Rev Nutr Diet. Basel, Karger, 2009, vol 100, pp 90–109

Cholesterol Was Healthy in the End

Uffe Ravnskov
Magle Stora Kyrkogata 9, Lund, Sweden

ravnskov@tele2.se

The idea that a high intake of saturated fat and a high cholesterol concentration in the
blood lead to atherosclerosis and cardiovascular disease emanates from a variety of
sources. When considered together, it is understandable that a whole world of doctors
and medical scientists have embraced the diet-heart idea and the cholesterol hypothesis,
in particular because two of the main supporters, Joseph Brown and Michael
Goldstein, have been honored with the Nobel Prize.
According to Karl Popper, a scientific theory is genuinely scientific only if it is possible
to create falsifiable predictions; no number of positive outcomes is able to prove
a scientific theory, whereas contradictory and reproducible observations show that
the theory is false. By this definition, the diet-heart idea and the cholesterol hypothesis
indeed satisfy Popper’s criteria, because there are a large number of observations
that are falsifiable, and indeed have been falsified again and again, as will be shown
in the following:

1 A high intake of saturated fat raises blood cholesterol.
2 Cholesterol is a main constituent of atherosclerotic plaques.
3 Premature atherosclerosis is seen in animals with hereditary, or dietary induced,
hypercholesterolemia.
4 High cholesterol is a risk marker for coronary heart disease (CHD).
5 People with familial hypercholesterolemia (FH) run a greater risk of dying from
CHD.
6 Cholesterol lowering prevents cardiovascular disease.

But it is also obvious that as soon as we start analyzing them, we find that all of
them have been falsified again and again. Great problems also arise when we try to
explain the pathogenic mechanisms, but let me start by reviewing the most striking
falsifications.


The Effect of Saturated Fat on Blood Cholesterol

The idea that saturated fat raises blood cholesterol was originally based on a number
of short-term laboratory studies. In a review from 1973, Reiser pointed at several
types of methodological and interpretational errors [1]. Instead of natural saturated
fat, many authors had used vegetable oils saturated by hydrogenation, and effects on
cholesterol were attributed to increased or decreased intakes of saturated fat when it
could be due to opposite changes of the intake of polyunsaturated fat.
In spite of these flaws, most authors maintain that saturated fat raises cholesterol,
whereas monounsaturated and in particular polyunsaturated fat lowers it, and some
saturated fatty acids are neutral [2–7]. These conclusions have been based mainly on
mathematical formulas using data from a large number of trials. But as most trial
directors have introduced similar types of bias, for instance by changing the intake of
several fats at the same time without controlling for intake of trans fat, it is obviously
difficult to rule out the effect of each type of fat.
No association has been found either in cross-sectional studies between total cholesterol
(tC) or low-density lipoprotein cholesterol (LDL-C) and the intake of saturated
fat, determined by questionnaires and interviews [9]. Also contradictory is that
populations who live almost entirely on animal food have the lowest cholesterol ever
measured in healthy people [10–12].
The strongest falsifications come from the controlled, randomized dietary trials. In
a review of eight such trials, where the intake of saturated fat was reduced by 30–40%,
the net reduction of tC was only 0–4% [13], and in more recent trials, where carbohydrates
were substituted with saturated fat, not even intakes between 20 and 50% of
calories influenced tC or LDL-C [14–23].
As cardiovascular disease is strongly associated with the concentration of small,
dense LDL than with other lipid fractions, it is also contradictory that the intake of
saturated fat is inversely associated with LDL size [24].
The Effect of Saturated Fat on Cardiovascular Disease

In a study considered as the strongest argument for the diet-heart idea, Keys selected
16 cohorts in seven countries and found a weak association between intake of saturated
fat and the prevalence and 5-year incidence of coronary mortality. But within
each country, there were great differences although intake of saturated fat was similar.
Coronary mortality for instance was three times higher in Karelia than in West
Finland and more than sixteen times higher on Corfu than on Crete [25].
Other epidemiological observations have been just as contradictory. More than
twenty cohort studies found no difference between the intake of saturated fat in
patients with CHD and healthy controls [9, 26]; and in seven of ten such studies
stroke patients had eaten less [27–36].
A relevant objection against such studies is that dietary information is inaccurate.
More reliable are analyses of fat tissue, because intake of saturated fat during the last
weeks or months is reflected by the concentration of the short chain fatty acids 12:0–
15:0 in fat cells [37–41]. Using this method, no difference was found between patients
with CHD and healthy controls; in two studies, the content of the short-chain fatty
acids was even significantly lower in the patients [42–46], and no association was
found with degree of atherosclerosis, determined either by autopsy [47] or by coronary
angiography [48]. These studies concerned only patients with first myocardial
infarction or patients who were not on a diet, and a diet bias is therefore unlikely.
The most important argument for causality is improvement or disappearance of the
disease after a decrease or discontinuation of the exposure to the suspected causal factor.
Two meta-analyses of the clinical trials where the only intervention was a change
of dietary fat found no effect, either on cardiovascular or total mortality [9, 49, 50].
Angiographic trials have given disparate results. In two studies, progress of the vascular
changes was associated with the intake of saturated fat, but both of them were
multifactorial, because in addition to lowering the intake of saturated fat, patients in
the treatment group were also instructed to eat more fish, fruit and vegetables [51,
52]. In contrast, a highly significant inverse association was found between intake of
saturated fat and progress of angiographic lesions in a 3-year follow-up study of 235
postmenopausal women with CHD [53]. No dietary advice was given in that study;
instead, the participants’ diets were recorded meticulously.
Two randomized, controlled dietary trials have succeeded in lowering both heart
and total mortality by changing dietary fat. However, the effect was most likely due to
an increased intake of omega–3 polyunsaturated fat, and it concerned sudden death
only, not coronary death, and the effect was not due to cholesterol lowering, because
no lowering was achieved [54, 55].
The Effect of High Cholesterol on Atherosclerosis 

It is true that hypercholesterolemic rodents develop atherosclerosis, but it is also true
that although these experimental models have been used for about 100 years, no one
has ever been able to produce an occluding thrombus or a myocardial infarction by
this method, and there is little evidence, if any at all, that high cholesterol causes
atherosclerosis in man. If too much cholesterol should cause atherosclerosis, people with
high cholesterol should be more atherosclerotic than people with low cholesterol, and
the progress of atherosclerosis in a cholesterol-lowering trial should depend on the
degree of cholesterol lowering, but this is not so.
Already in 1936, Landé and Sperry falsified the first prediction. In a study of a
large number of healthy people who had died violently, they found that on average
those with low cholesterol were just as atherosclerotic as those with high cholesterol
[56], and their result has been confirmed by others [57–61]. Weak associations have
been present between blood cholesterol and degree of atherosclerosis in studies of
selected patient groups. As these groups mainly included patients with cardiovascular
diseases, individuals with FH must have been much more frequent than in unselected
cohorts. In accordance, the associations disappeared after exclusion of people with
extremely high cholesterol, or the association was inconsistent and present in small
subgroups only [62–72] (table 1).
The second prediction is false as well. With a single exception, dose-response
between degree of cholesterol lowering and the angiographic changes has not been
found in any cholesterol-lowering trial [73]. In observational angiographic studies,
no or even an inverse association was found between the spontaneous changes of
cholesterol and the degree of progress [74–77].


The Effect of High Cholesterol on Cardiovascular Disease 

If high cholesterol leads to CHD or ischemic stroke, people with these diseases should
have higher cholesterol than others before the arrival of their disease, and the outcome
of a trial should depend on the degree of cholesterol lowering. Also these predictions
have been falsified in countless studies.
High cholesterol was found to be a risk factor for CHD for the first time in the
Framingham project. However, at the 30-year follow-up, it appeared that high cholesterol
was not a risk factor after age 47. Even more contradictory was that both coronary
and total mortality was higher in those whose cholesterol had decreased during
these years than in those whose cholesterol had increased. ‘For each 1% mg/dl drop of
cholesterol there was an 11 percent increase in coronary and total mortality’ [78]. It is
not too farfetched to assume that, being taken care of by the Framingham researchers,
most of these people had been on cholesterol lowering treatment, which adds further
strength to this falsification.
Since then, numerous studies have shown that for most populations high cholesterol
is not a risk factor. They included Canadian men [79], diabetics [80–93], patients with
renal failure [94, 95], patients who already had CHD [96–101], and almost all studies
have found that it is not a risk factor for women [102] or for old people either [103].
Indeed, old people with high cholesterol live longer than old people with low cholesterol
[104–120]. The two last-mentioned falsifications are particularly strong, because
at least in Sweden more than 90% of all cardiovascular deaths occur after age 65.
The Effect of High Cholesterol in Familial Hypercholesterolemia 

If high cholesterol is the cause of atherosclerosis and early CHD in FH, those with the
highest values should of course be at greater risk than those whose cholesterol is only
a little higher than normal. This is not so, however.

 

Table 1. Studies of the association between the concentration of cholesterol in the blood and degree of
atherosclerosis at autopsy

Study Type of investigated individuals Association between blood cholesterol and degree of atherosclerosis
Landé and Sperry [56] Healthy people who have died none
Paterson et al. [57] unselected group of war veteran s none
Mathur et al. [58) healthy people who have died
from accidents
none
Marek et al. [59] healthy people who have died
from accidents
none if those with very high cholesterol were excluded
Schwartz et al. [60] unselected hospital patients none for women, weak for men
Méndez and Tejada [61 healthy people who have died from accidents none
Rhoads et al. [62] a selection of hospital patients very weak
Feinleib et al (63) a selection of dead people men: very weak; women: none
Sadoshima et al. (64) a selection of dead people
Oalman et al. [65] a selection of dead people Black people: none
White people: none if those with very high
cholesterol were excluded
Sorlie et al. [66] a selection of dead people coronary arteries: weak
aorta: very weak
Solberg et al. [67, 68] a selection of dead people weak
Okumiya et al. [69] a selection of dead people weak
Reed et al. [70] a selection of dead people large arteries: weak
small arteries: none
Reed et al. [71] a selection of dead people coronary arteries: weak (tC)
cerebral arteries: none (tC)
none anywhere for LDL-C

At least eight studies have shown that neither the incidence nor the prevalence of
cardiovascular disease is associated with the lipid levels [121–128]; in one of these
studies, mean cholesterol was even lower in those who died from CHD [125]. A striking
fact is also that in people with FH and severe atherosclerotic changes in their
coronary arteries, no changes were seen in the cerebral arteries [129, 130].
That the vascular changes in FH are independent of blood cholesterol was noted even
by Brown and Goldstein. In a 1983 paper, they wrote the following: ‘Among patients
with FH (both heterozygous and homozygous), there is considerable variation in the
rate of progression of atherosclerosis, despite uniformly elevated LDL levels’ [132].
The number of those who die at a young age from CHD is not very large either.
In the Simon Bromee study, the authors followed almost 3,000 individuals with FH
for many years and found that their mean life span was similar as for normal British
citizens of the same age and sex; more died from heart disease, but fewer died from
cancer and other diseases [133].
A possible cause of cardiovascular disease in FH may be inborn errors of the coagulation
system. In cohort studies of people with FH, plasma fibrinogen and factor VIII
were significantly higher in those with CHD than in those without [134], whereas tC
and LDL-C did not differ significantly. Recently, Kastelein’s group found that polymorphism
in the prothrombin gene is strongly associated with cardiovascular risk in
people with this disorder [128]. The reason why statin treatment is of benefit in FH
may therefore be their antithrombotic effects, not their effect on cholesterol.


The Effect of Cholesterol Lowering
 

If high cholesterol causes cardiovascular disease, the most important prediction is
that its lowering alone should reduce that risk. Most studies used as support before
the statins were introduced were multifactorial. But a meta-analysis of all controlled
and randomized cholesterol lowering trials performed before the advent of the statins
found no effect on coronary mortality, and total mortality was increased [135].
That cholesterol lowering by the HMG coenzyme A inhibitors is able to lower the
risk of cardiovascular disease in high-risk patients is seen as evidence of the cholesterol
hypothesis. However, and as mentioned above, no trial has found any association
between the degree of cholesterol lowering and the clinical or angiographic outcome;
those whose cholesterol was lowered a little only had the same small benefit as those
whose cholesterol was lowered by more than 50%. Lack of exposure response means
that the statins must have other effects that are more beneficial than cholesterol lowering,
as suggested already after the publication of one of the first clinical statin trials
[136], and several such effects have indeed been documented.
But even if the lowering of cholesterol by these drugs were unimportant, there
should have been exposure response between cholesterol and outcome, because both
the pleiotropic effects and cholesterol lowering are caused by the same drug. A more
complete blockage of the mevalonate pathway should result in stronger pleiotropic
effects and a more pronounced lowering of cholesterol, and vice versa. As this was
not the case, the findings imply that high cholesterol is protective and that its lowering
therefore counteracts exposure response. There is indeed much support to that
interpretation.

Table 2. Studies of the lipoprotein immune system 

Study Microbial product Source of lipoprotein Methods used to demonstrate inactivation and/or binding of the microbial products by the lipoproteins

 

Stollerman et al. [140] Streptolysin S man inhibition of streptolysin S
Stollerman et al. [140] Streptolysin S man

same

Skarnes (141) (S- enteritides) rodents immunodiffusion
Shortridge et al. [142] Togaviruses man inhibition of hemagglutination
Whitelaw et al. [143] S. aureus δ-hemolysin man  inhibition of δ-hemol ysin
Freudenberg et al. [144] S. abortus equi;
S. minnesota
rat crossed immunoelectrophoresis
Ulevitch et al. [145] LPS (S. minnesota) rabbit binding of LPS to apoA1
Bhakdi et al. [146 S. aureus α-toxin man hemolytic titration; EM
Seganti et al. [147] Rhabdovirus man inhibition of hemagglutination
van Lenten et al. [148] LPS (Escherichia coli) man, rabbit inhibition of scavenger receptor
Huemer et al. [149] herpes simplex man E M
Flegel et al. [150] LPS (E. coli) man inhibition of endotoxin activation
Cavaillon et al. [151] LPS (E. coli) rabbit inhibition of cytokine respon se
Northoff et al. [152] LPS (?) man inhibition of cytokine response
Superti et al. [153] SA rotavirus man inhibition of viral hemagglutina- tion and replication; EM
Weinstock et al. [154] LPS (S. typhi) man inhibition of endotoxin production
Flegel et al. [155] LPS (S. typhi) man inhibition of endotoxin production
Feingold et al. [156] LPS (E. coli) man endotoxin sensitivity
Netea et al. [157] (LPS) E. coli) mouse LD50 after experimental infection

 

The Lipoprotein Immune System 

It is little known that the lipoproteins partake in the immune system. For many years,
a normal serum factor, named antistreptolysin S because it was able to neutralize the
hemolytic effects of streptolysin S, was considered to be an antibody. In 1937, Todd et
al. [137] found that it did not behave as a normal antibody because its titer fell below
normal values in patients with rheumatic fever at the peak of the clinical symptoms,
and a few years later, Stollerman and Bernheimer noted that, in contrast to the anti-
streptococcal antibodies, the antistreptolysin S titer did not rise above its normal level
during convalescence [138]. Humphrey discovered that antistreptolysin S was located
within the lipid fraction of the blood [139], and Stollerman et al. [140] identified it
as a phospholipoprotein complex. Since then, at least a dozen research groups have
established that antistreptolysin S is identical with the lipoproteins and constitutes a
nonspecific host defense system able to bind and inactivate not only streptolysin S but
also other endotoxins and several virus species as well (table 2) [139–157]. In rodents,
the main bulk of cholesterol is transported by high-density lipoprotein (HDL), and
in these species HDL has the main protective effect [144, 145], whereas most human
studies have found that all lipoproteins participate in the nonspecific defense system.
The immunoprotective role of the lipoproteins has been shown by their inhibition
of the biological effects of various microorganisms and endotoxins, such as hemagglutination,
hemolysis, the cytokine response of human monocytes, and virus replication
(table 2).
That lipoproteins also form complexes with microbial products was shown first
by Skarnes [141]. By using immunodiffusion with anti-endotoxin and serum from
various rodents that had been injected with Salmonella enteridis endotoxin, he demonstrated
lipoprotein-positive staining and esterase activity on the precipitation lines.
Using crossed immunoelectrophoresis, Freudenberg and Galanos [144] found that
the HDL peak of rat plasma changed position after injection with various lipopolysaccharides
(LPS), and Ulevitch et al. [145] found evidence of complex formation
between LPS from Salmonella minnesota and apoprotein A1, the major protein of
rabbit HDL.
Bhakdi et al. [146] have documented that human lipoproteins complex with microbial
components as well. By electron microscopy (EM), they found that the inactivation
of Staphylococcus aureus α-toxin by purified human LDL led to oligomerization
of 3S native toxin molecules into ring structures of 11S hexamers that adhered to the
LDL molecules.
Lipoproteins also form complexes with viruses. Thus, using various techniques
Huemer et al. [149] found that all lipoprotein subclasses were able to bind purified
herpes simplex virus, as demonstrated by EM, enzyme-linked immunoabsorbance
assay technique, and column chromatography. Superti et al. [153] confirmed that all
human subclasses of lipoproteins were able to inhibit the infectivity and hemagglutination
by SA-11 rotavirus, and complex formation was visualized by EM.
The lipoprotein immune system may be particularly important in early childhood
as, in contrast to antibody-producing cells, this system works immediately and with
high efficiency. For instance, human LDL inactivated up to 90% of S. aureus α-toxin
[146], and it inactivated an even larger fraction of bacterial LPS [150]. In agreement
with these findings, hypocholesterolemic rats injected with LPS had a markedly
increased mortality compared with normal rats, which could be ameliorated by
injecting purified human LDL [156]. On the other hand, hypercholesterolemic mice
challenged with LPS or live bacteria had an 8-fold increased LD50, compared with
normal mice [157]. That high levels of lipoproteins protect against infectious diseases
is also evident from clinical and epidemiological studies.


The Benefits of High Cholesterol 

If the lipoproteins have an immunoprotective role, high cholesterol should be an
advantage, not a risk factor, and there is indeed many observations in support. Thus,
a meta-analysis of 19 cohort studies including almost 70,000 deaths found an inverse
association between tC and mortality from respiratory and gastrointestinal diseases,
most of which are of an infectious origin [102]. It has been argued that low cholesterol
was secondary, but this explanation was disproved by Iribarren et al. [158, 159]. They
followed more than 20,000 healthy individuals for 15 years and found a strong inverse
association between tC and the risk of being admitted to hospital because of an infectious
disease. The association included all types of infection, and it was statistically
significant for most of them. As regards respiratory diseases, the association was significant
for pneumonia and influenza, but not for asthma. As all of the participants
were healthy at the start, it is obvious that their low cholesterol could not be secondary
to a disease they had not yet manifested.
There is evidence that subclinical infections participate in chronic heart failure. In
accordance, patients with heart failure and low cholesterol run a greater risk of premature
death than patients with high cholesterol [160]. Low cholesterol is also a risk
factor for HIV and AIDS [161, 162], hepatitis B [163], and for death due to an infectious
disease in patients with chemotherapy-induced neutropenia [164].
The protective role of high cholesterol is also evident from observations in people
with inborn errors of cholesterol metabolism. For instance, the frequent and severe
infections in children with extremely low cholesterol that are found in the Smith-
Lemli-Opitz syndrome are alleviated by the addition of cholesterol to their diet
[165].
Even in FH, a high cholesterol seems to protect against infections. Thus, before the
year 1900, when infectious diseases were the commonest cause of death, the life span
of people with a 50% risk of having FH was longer than for other people [166].
Cholesterol and Cancer
Many cohort studies have found that low cholesterol is a risk factor for cancer. The
usual explanation is that cancer causes low cholesterol because cholesterol is consumed
by the cancer cells. However, in the Framingham project low cholesterol was
a risk factor for cancer even after 18 years of follow-up [167], and as mentioned,
cancer mortality in people with FH is lower than in the general population. Many
observations are also in better accord with the opposite interpretation that low cholesterol
predisposes to cancer.
First, in a review of cholesterol-lowering experiments in laboratory animals, the
authors concluded that most studies produced cancer [168]. As this effect was seen
also after nonstatin drugs, and as no chromosomal aberrations were noted in the animals,
there is reason to suspect that the culprit was not the drugs, but rather their
effect, the lower concentration of cholesterol; an interpretation that is supported by
epidemiological observations and human experiments.
In 4S, the Scandinavian Simvastatin Survival Study [169], and in HPS, the Heart
Protection Study [170], the two first simvastatin trials, nonmelanoma skin cancer was
observed more often in the treatment groups. The difference was statistically significant
when the results from both studies were combined (in the simvastatin groups,
256 of the 12,490 participants, and in the control groups, 208 of the 12,490 participants;
p = 0.028). For unknown reasons, the number of nonmelanoma skin cancers
has not been reported in any of the trial reports that followed.
The clinical appearance of a cancer depends on its location. Lung cancer, for
instance, is not diagnosed until after decades of smoking, whereas superficial nonmelanoma
cancers may be observed much earlier. An increased number of patients
with skin cancer in a trial is therefore alarming because this is the first cancer type
that we should expect to find under conditions of general carcinogenicity.
In CARE, the Cholesterol and Recurrent Events Trial [171], 12 of the 286 women
in the pravastatin group but only 1 of the 290 in the placebo group had breast cancer
at follow-up (p = 0.002). Again, breast cancer is a superficial malignancy that is easier
to observe and should therefore occur much earlier than for instance a cancer located
in the pancreas. Furthermore, several of these breast cancers were recurrences, and
recurrences may appear earlier than primary cancers. However, it is not possible to
test the hypothesis that cholesterol lowering by statin treatment may provoke recurrences
because after the publication of the CARE report, previous cancer has become
an exclusion criterion in all trials.
Dormant cancer is a common finding in elderly people, and a carcinogenic effect
should therefore appear earlier in that patient group. Indeed, in the PROSPER trial [172],
which included elderly people only, 245 of the 2,891 participants in the pravastatin group
but only 199 of the 2,913 in the placebo group had new cancer. The difference was already
obvious after 1 year, and it increased steadily during the trial period to become statistically
significant (p = 0.02) after 4 years. The authors claimed that a meta-analysis of all
pravastatin trials did not confirm a carcinogenic effect. This is not reassuring because
the mean age in these trials was about 25 years lower than in the PROSPER trial.
In a cohort study of 47,294 Japanese patients treated with low-dose simvastatin
and followed for 6 years, the authors found that the number of cancer deaths was
significantly higher in patients whose tC at follow-up was less than 160 mg/dl than
in those whose cholesterol was 200–219 mg/dl (relative risk = 3.16; 95% CI = 1.72 to
5.81; p = 0.001) [173].
Another argument in support of carcinogenicity is a report by Iwata et al. [174],
who found that recent or previous statin treatment was seen twice as often in patients
with lymphoid cancer compared with patients admitted to the hospital for noncancer
diseases. Again, lymphoid cancer belongs to the types of malignancies that are easy to
diagnose at an early stage.
Several authors have claimed that statin treatment prevents cancer. However, a bias
is introduced by the method used in these studies because in all of them patients on
statin treatment were compared with untreated individuals. The first group is a selection
of people who initially had high cholesterol, which has been lowered for a few
years only; the second is dominated by people who might have had low cholesterol for
most of their life and are therefore at an increased risk of cancer.


The Pathogenic Mechanism 

Today, most researchers agree that atherosclerosis starts as an inflammation in the
arterial wall. What is also common knowledge is that the starting point of the occluding
thrombosis is the vulnerable plaque. Therefore, any credible hypothesis must be
able to explain how and why the inflammation starts and how a vulnerable plaque is
created.
According to the current view, the first step is endothelial dysfunction or damage
caused by hypercholesterolemia or other toxic factors in the circulation allowing the
migration of LDL-C and monocytes into the arterial wall. Here, LDL is said to be
modified by oxidation leading to an accumulation of T cells and the production of
LDL autoantibodies. Modified LDL is taken up by macrophages that are converted to
lipid-laden foam cells, considered as the early lesion of atherosclerosis. The inflammatory
processes, probably aggravated by antigens from microbes such as chlamydia,
herpes simplex and cytomegalovirus, are followed by smooth muscle cell proliferation
and the synthesis of extracellular matrix. The macrophages may become overloaded
and die resulting in the creation of a vulnerable plaque that by bursting initiates the
formation of an occluding thrombus [175]. There are a number of contradictions to
this hypothesis, however.
There is no association between the concentration of LDL-C in the blood and the
degree of endothelial dysfunction [176]; the atherosclerotic plaques seen in extreme
hyperhomocysteinemia due to inborn errors of methionine metabolism do not contain
any lipids in spite of pronounced endothelial damage [177].
A more likely mechanism is that aggregated complexes formed by lipoproteins
and microorganisms or their toxins may occlude vasa vasorum of the major arteries
because of the high extracapillary pressure, resulting in local ischemia, liberation of
microorganisms that are attached to the complexes and the formation of a microabscess,
the vulnerable plaque. Rupture of the latter may result not only in local clot
formation around the rupture, but also in an emptying of the microbial content of the
vulnerable plaque into the circulation [178].
Extensive aggregation may occur in severe infections, and it may be furthered by
hyperhomocysteinemia, because homocysteine thiolactone, the reactive cyclic anhydride
of homocysteine, reacts with free amino groups of protein to form peptidebound
homocysteine [179]. In accordance, in vitro experiments have shown that
thiolated LDL becomes aggregated and subject to spontaneous precipitation in vitro
[180]. Thiolated and oxidized LDL may also stimulate the formation of anti-LDL
autoantibodies [181], furthering complex formation and aggregation.
As macrophages take up aggregated LDL by phagocytosis after modification by
vortexing or by digestion with phospholipase C [182], they may do it with LDL molecules
modified by complex formation, oxidation or thiolation as well and in this way
be converted to foam cells. In support of that, in vitro experiments have shown that
LPS from Chlamydia pneumoniae [181] and also from several periodontal pathogens
[182] is able to convert macrophages to foam cells in the presence of human LDL.
The suggested mechanism explains how cholesterol enters the arterial wall, the
many associations between cardiovascular and infectious diseases and the similarities
between their clinical and laboratory symptoms and signs, why many bacterial and
viral remnants are present in atherosclerotic lesions [183, 184], why neutrophils are
found in the vulnerable plaques but not in the stable, fibrous plaques [185], why the
temperature of vulnerable plaques is higher than that of its surroundings [186], why
leucocytes are found preferably around vasa vasorum [187], and why bacteremia and
sepsis are often seen in myocardial infarction complicated with cardiogenic shock
[188].
The hypothesis is open for falsification as well. Viable microorganisms and endotoxins
in the arterial wall should be located within developing vulnerable plaques.
Arteries of germ-free animals should have fewer foam cells and fatty streaks than
their conventionally reared littermates.
A blood culture should be taken in all patients with unstable angina or myocardial
infarction, and we anticipate that if it is positive, the course of the disease should be
improved with an appropriate antibiotic.
 

References 

1 Reiser R: Saturated fat in the diet and serum cholesterol concentration: a critical examination of the literature. Am J Clin Nutr 1973;26:524–555.

2 Mensink RP, Katan MB: Effect of dietary fatty acids on serum lipids and lipoproteins. A meta- analysis of 27 trials. Arterioscler Thromb Vasc Dis 1992;12:911–919.

3 Hegsted DM, Ausman LM, Johnson JA, Dallal GE: Dietary fat and serum lipids: an evaluation of the experimental data. Am J Clin Nutr 1993; 57:875–883.

4 Woodside JV, Kromhout D: Fatty acids and CHD. Proc Nutr Soc 2005;64:554–564.

5 Kuller LH: Nutrition, lipids, and cardiovascular disease. Nutr Rev 2006;64:S15–S26.

6 Lapointe A, Balk EM, Lichtenstein AH: Gender differences in plasma lipid response to dietary fat. Nutr Rev 006;64:234–249.

7 Jansen AC, van Aalst-Cohen ES, Tanck MW, Trip MD, Lansberg FJ, Liem AH, et al: The contribution of classical risk factors to cardiovascular disease in familial hyperchole-sterolaemia: data in 2400 patients. J Intern Med 004;256:482–490.

8 Mensink RP, Zock PL, Kester AD, Katan MB: Effects of dietary fatty acids and carbohydrates on the ratio of serum total to HDL cholesterol and on serum lipids and apolipo-proteins: a meta-analysis of 60 controlled trials. Am J Clin Nutr 2003;77:1146–1155.

9 Ravnskov U: The questionable role of saturated and poly-unsaturated fatty acids in cardiovascular disease. J Clin Epidemiol 1998;51:443–460.

10 Shaper AG: Cardiovascular studies in the Samburu tribe of northern Kenya. Am Heart J 1962;63:437–442.

11 Lapiccirella V, Lapiccirella R, Abboni F, Liotta S: Enquete clinique, biologique et cardiographique parmi les tribus nomades de la Somalie qui se nourissent seulement de lait. Bull World Hlth Org 1962;27:681–697.

12 Mann GV, Shaffer RD, Sandstead HH: Cardiovascular disease in the Masai. J Atheroscl Res 1964;4:289–312.

13 Ramsay LE, Yeo WW, Jackson PR: Dietary reduction of serum cholesterol concentration: time to think again. BMJ 1991;303:953–957.

14 Noakes M, Foster PR, Keogh JB, James AP, Mamo JC, Clifton PM: Comparison of isocaloric very low carbohydrate/ high saturated fat and high carbohydrate/low saturated fat diets on body composition and cardiovascular risk. Nutr Metab 2006;3:7.

15 Meckling KA, O’Sullivan C, Saari D: Comparison of a low-fat diet to a lowcarbohy- drate diet on weight loss, body composition, and risk factors for diabetes and cardiovascular dis- ease in free-living, overweight men and women. J Clin Endocrinol Metab 2004;89:2717–2723.

16 Sondike SB, Copperman N, Jacobson MS: Effects of a low-carbohydrate diet on weight loss and cardiovascular risk factor in overweight adolescents. J Pediatr 2003;142:253–258.

17 Sharman MJ, Gomez AL, Kraemer WJ, Volek JS: Very low-carbohydrate and low-fat diets affect fasting lipids and postprandial lipemia differently in overweight men. J Nutr 2004;134:880–885.

18 Hays JH, DiSabatino A, Gorman RT, Vincent S, Stillabower ME: Effect of a high saturated fat and no-starch diet on serum lipid subfractions in patients with documented atherosclerotic cardio- vascular disease. Mayo Clin Proc 2003;78: 1331–1336.

19 Westman EC, Yancy WS, Edman JS, Tomlin KF, Perkins CE: Effect of 6-month adherence to a very low carbohydrate diet program. Am J Med 2002;113:30–36.

20 Foster GD, Wyatt HR, Hill JO, McGuckin BG, Brill C, Mohammed BS, Szapary BG, Rader DJ, Edman JS, Klein S: A randomized trial of a low-carbohydrate diet for obesity. N Engl J Med 2003;348:2082–2090.

21 Yancy WS Jr, Foy M, Chalecki AM, Vernon MC, Westman EC: A lowcarbohydrate, keto- genic diet to treat type 2 diabetes. Nutr Metab 2005;2:34–40.

22 Seshadri P, Iqbal N, Stern L, Williams M, Chicano KL, Daily DA, McGrory J, Gracely EJ, Rader DJ, Samaha FF: A randomized study comparing the effects of a low-carbo- hydrate diet and a conventional diet on lipoprotein sub- fractions and C-reactive protein levels in patients with severe obesity. Am J Med 2004;117: 398–405.

23 Brehm BJ, Seeley RJ, Daniels SR, D’Alessio DA: A randomized trial comparing a very low carbohydrate diet and a calorie-restricted low fat diet on body weight and cardiovascular risk factors in healthy women. J Clin Endocrinol Metab 2003;88:1617–1623.

24 Krauss RM, Blanche PJ, Rawlings RS, Fernstrom HS, Williams PT: Separate effects of reduced carbohydrate intake and weight loss on atherogenic dyslipidemia. Am J Clin Nutr 2006; 83:1025–1031.

25 Keys A: Coronary heart disease in seven countries. Circulation 1970;41(suppl 1):1–211.

26 Leosdottir M, Nilsson PM, Nilsson JA, M.nsson H, Berglund G: Dietary fat intake and early mortality patterns – data from The Malmö Diet and Cancer Study. J Intern Med 2005;258: 153–165.

27 Stemmermann GN, Hayashi T, Resch JA, Chung CS, Reed DM, Rhoads GG: Risk factors related to ischemic and hemorrhagic cerebro- vascular disease at autopsy: the Honolulu Heart Study. Stroke 1984;15:23–28.

28 Takeya Y, Popper JS, Shimizu Y, Kato H, Rhoads GG, Kagan A: Epidemiologic studies of coronary heart disease and stroke in Japanese men living in Japan, Hawaii and California: inci- dence of stroke in Japan and Hawaii. Stroke 1984;15:15–23.

29 McGee D, Reed D, Stemmerman G, Rhoads G, Yano K, Feinleib M: The relationship of dietary fat and cholesterol to mortality in 10 years: the Honolulu Heart Program. Int J Epidemiol 1985;14:97–105.

30 Gillman MW, Cupples LA, Millen BE, Ellison RC, Wolf PA: Inverse association of dietary fat with development of ischemic stroke in men. JAMA 1997;278:2145–2150

31 Ross RK, Yuan JM, Henderson BE, Park J, Gao YT, Yu MC: Prospective evaluation of dietary and other predictors of fatal stroke in Shanghai, China. Circulation 1997;96:50–55.

32 Seino F, Date C, Nakayama T, Yoshiike N, Yokoyama T, Yamaguchi M, Tanaka H: Dietary lipids and Incidence of cerebral infarction in a Japanese rural community. J Nutr Sci Vitaminol 1997;43:83–99.

33 Iso H, Stampfer MJ, Manson JE, Rexrode K, Hu F, Hennekens CH, Colditz GA, Speizer FE, Willett WC: Prospective study of fat and protein intake and risk of intraparenchymal hemorrhage in women. Circulation 2001;103:856–863.

34 He K, Merchant A, Rimm EB, Rosner BA, Stampfer MJ, Willett WC, Ascherio A: Dietary fat intake and risk of stroke in male US health- care professionals: 14 year prospective cohort study. BMJ 2003;327:777–782.

35 Iso H, Sato S, Kitamura A, Naito Y, Shima- moto T, Komachi Y: Fat and protein intakes and risk of intraparenchymal hemorrhage among middle-aged apanese. Am J Epidemiol 2003; 157:32–39.

36 Sauvaget C, Nagano J, Hayashi M, Yamada M: Animal protein, animal fat, and cholesterol intakes and risk of cerebral infarction mortality in the adult health study. Stroke 2004;35:1531–7.

37 Smedman AE, Gustafsson IB, Berglund LG, Vessby BO: Pentadecanoic acid in serum as a marker for intake of milk fat: relations between intake of milk fat and metabolic risk factors. Am J Clin Nutr 1999;69:22–29.

38 Wolk A, Furuheim M, Vessby B: Fatty acid composition of adipose tissue and serum lipids are valid biological markers of dairy fat intake in men. J Nutr 2001;131:828–833.

39 Rosell M, Johansson G, Berglund L, Vessby B, de Faire U, Hellenius ML: Associations bet- ween the intake of dairy fat and calcium and ab- dominal obesity. Int J Obes Relat Metab Disord 2004;28:1427–1434

40 Brevik A, Veierod MB, Drevon CA, Andersen LF: Evaluation of the odd fatty acids 15:0 and 17:0 in serum and adipose tissue as markers of intake of milk and dairy fat. Eur J Clin Nutr 2005; 59:1417–1422.

41 Hudgins LC, Hellerstein MK, Seidman CE, Neese RA, Tremaroli JD, Hirsch J: Relationship between carbohydrate-induced hypertriglyce- ridemia and fatty acid synthesis in lean and obese subjects. J Lipid Res 2000;41:595–604.

42 Kirkeby K, Ingvaldsen P, Bjerkedal I: Fatty acid composition of serum lipids in men with myocardial infarction. Acta Med Scand 1972; 192:513–519.

43 Wood DA, Butler S, Riemersma RA, Thomson M, Oliver MF, Fulton M, Birtwhistle A, Elton R: Adipose tissue and platelet fatty acids and coronary heart disease in Scottish men. Lancet 1984;2:117–121.

44 Kark JD, Kaufmann NA, Binka F, Goldberger N, Berry EM: Adipose tissue n-6 fatty acids and acute myocardial infarction in a population con- suming a diet high in polyunsaturated fatty acids. Am J Clin Nutr 2003;77:796–802.

45 Clifton PM, Keogh JB, Noakes M: Trans fatty acids in adipose tissue and the food supply are associated with myocardial infarction. J Nutr 2004;134:874–879.

46 Pedersen JI, Ringstad J, Almendingen K, Haugen TS, Stensvold I, Thelle DS: Adipose tissue fatty acids and risk of myocardial infarction–a case-control study. Eur J Clin Nutr 2000;54:618–25.

47 Lang PD, Degott M, Heuck CC, Opherk D, Vollmar J: Fatty acid composition of adipose tissue, blood, lipids, and glucose tolerance in patients with different degrees of angiographi- cally documented coronary arteriosclerosis. Res Exp Med 1982;180:161–168.

48 Reed DM, Resch JA, Hayashi T, MacLean C, Yano K: A prospective study of cerebral artery atherosclerosis. Stroke 1988;19:820–825.

49 Hooper L, Summerbell CD, Higgins JP, Thompson RL, Capps NE, Smith GD, Riemersma RA, Ebrahim S: Dietary fat intake and prevention of cardiovascular disease: systematic review. BMJ 2000;322:757–763.

50 Ravnskov U: Diet-heart disease hypothesis is wishful thinking. BMJ 2002;324:238.

51 Watts GF, Jackson P, Burke V, Lewis B: Dietary fatty acids and progression of coronary artery disease in men. Am J Clin Nutr 1996; 64:202–209.

52 Bemelmans WJ, Lefrandt JD, Feskens EJ, Broer J, Tervaert JW, May JF, Smit AJ: Effect of an increased intake of alpha-linolenic acid and group nutritional education on cardiovascular risk factors: the Mediterranean Alpha-linolenic Enriched Groningen Dietary Intervention (MAR- GARIN) study. Am J Clin Nutr 2002;75:221–227

53 Mozaffarian D, Rimm EB, Herrington DM: Dietary fats, carbohydrate, and progression of coronary atherosclerosis in postmenopausal women. Am J Clin Nutr 2004;80:1175–1184.

54 de Lorgeril M, Renaud S, Mamelle N, Salen P, Martin JL, Monjaud I, Guidollet J, Touboul P, Delaye J: Mediterranean alpha-linolenic acid-rich diet in secondary prevention of coronary heart disease. Lancet 1994;343:1454–1459.

55 Gruppo Italiano per lo Studio della Sopravvivenza nell’Infarto miocardico: Dietary supplementation with n-3 polyunsaturated fatty acids and vitamin E after myocardial infarction: results of the GISSIPrevenzione trial. Gruppo Italiano per lo Studio della Sopravvivenza nell’Infarto miocardico. Lancet 1999;354: 447–455.

56 Landé KE, Sperry WM: Human athero- sclerosis in relation to the cholesterol content of the blood serum. Arch Pathol 1936;22:301–312.

57 Paterson JC, Dyer L, Armstrong EC: Serum cholesterol levels in human atherosclerosis. Can Med Ass J 1960;82:6–11.

58 Mathur KS, Patney NL, Kumar V, Sharma RD: Serum cholesterol and atherosclerosis in man. Circulation 1961;23: 847–852.

59 Marek Z, Jaegermann K, Ciba T: Athero- sclerosis and levels of serum cholesterol in postmortem investigations. Am Heart J 1962; 63:768–774.

60 Schwartz CJ, Stenhouse NS, Taylor AE, White TA: Coro-nary disease severity at necropsy. Br Heart J 1965;27;731–739.

61 Méndez J, Tejada C: Relationship between serum lipids and aortic atherosclerotic lesions in sudden accidental deaths in Guatemala City. Am J Clin Nutr 1967;20:1113–1117.

62 Rhoads GG, Blackwelder WC, Stemmer- mann GN, Hayashi T, Kagan A: Coronary risk factors and autopsy findings in Japanese- American men. Lab Invest 1978;38:304–311.

63 Feinleib M, Kannel WB, Tedeschi CG, Landau TK, Garrison RJ: The relation of antemortem characteristics to cardiovascular findings at necropsy. The Framingham Study. Atherosclerosis 1979;34:145–157.

64 Sadoshima S, Kurozumi T, Tanaka K, et al: Cerebral and aortic atherosclerosis in Hisa- yama, Japan. Atherosclerosis 1980;36:117–126

65 Oalmann MC, Malcom GT, Toca VT, Guzman MA, Strong JP: Community pathology of atherosclerosis and coronary heart disease: post mortem serum cholesterol and extent of coronary atherosclerosis. Am J Epidemiol 1981;113:396–403.

66 Sorlie PD, Garcia-Palmieri MR, Castillo- Staab MI, Costas R, Oalmann MC, Havlik R: The relation of antemortem factors to athero- sclerosis at autopsy. The Puerto Rico Heart Health Program. Am J Pathol 1981;103:345–352.

67 Cabin HS, Roberts WC: Relation of serum total cholesterol and triglyceride levels to the amount and extent of coronary artery narrowing by atherosclerotic plaque in coronary heart disease. Am J Med 1982;73:227–234.

68 Solberg LA, Strong JP, Holme I, Helgeland A, Hjerman I, Leren P, Mogensen SB: Stenoses in the coronary arteries. Relation to atheroscle- rotic lesions, coronary heart disease, and risk factors. The Oslo Study. Lab Invest 1985;53: 648–655.

69 Solberg LA, Hjermann I, Helgeland A, Holme I, Leren PA, Strong JP: Association between risk factors and atherosclerotic lesions based on autopsy findings in the Oslo study: a preliminary report; in Schettler G, Goto Y, Hata Y, Klose G (eds): Atherosclerosis IV. Proc 4 Int Symp. Berlin, Springer, 1977, pp 98–100.

70 Okumiya N, Tanaka K, Ueda K, Omae T: Coronary atherosclerosis and antecedent risk factors: pathologic and epidemiologic study in Hisayama, Japan. Am J Cardiol 1985;56:62–66.

71 Reed DM, Resch JA, Hayashi T, MacLean C, Yano K: A prospective study of cerebral artery atherosclerosis. Stroke 1988;19:820–825.

72 Reed DM, Strong JP, Resch J, Hayashi T: Serum lipids and lipoproteins as predictors of atherosclerosis. An autopsy study. Arterio- sclerosis 1989;9:560–564.

73 Ravnskov U: Is atherosclerosis caused by high cholesterol?QJM 2002;95:397–403.

74 Kramer JR, Kitazume H, Proudfit WL, Matsuda Y, Williams GW, Sones FM: Pro- gression and regression of coronary athero- sclerosis: relation to risk factors. Am Heart J 1983;105:134–144.

75 Bemis CE, Gorlin R, Kemp HG, Herman MV: Progression of coronary artery disease: a clini- cal arteriographic study. Circulation 1973;47: 455–464.

76 Kimbiris D, Lavine P, Van Den Broek H, Najmi M, Likoff W: Devolutionary pattern of coro- nary atherosclerosis in patients with angina pectoris. Coronary arteriographic studies. Am J Cardiol 1974;33:7–11.

77 Shub C, Vlietstra RE, Smith HC, Fulton RE, Elveback LR: The unpredictable progression of symptomatic coronary artery disease: a serial clinical-angiographic analysis. Mayo Clin Proc 1981;56:155–160.

78 Anderson KM, Castelli WP, Levy D: Chole- sterol and mortality. 30 years of follow-up from the Framingham study. JAMA 1987;257: 2176–2180.

79 Dagenais GR, Ahmed Z, Robitaille NM, Gingras S, Lupien PJ, Christen A, Meyer F, Rochon J: Total and coronary heart disease mortality in relation to major risk factors. Quebec Cardiovascular Study. Can J Cardiol 1990;6:59–65.

80 Fontbonne A, Eschwège E, Cambien F, Richard JL, Ducimetière P, Thibult N, Warnet JM, Claude JR, Rosselin GE: Hypertriglycerid- aemia as a risk factor of coronary heart disease mortality in subjects with impaired glucose tole- rance or diabetes. Results from the 11-year follow-up of the Paris Prospective Study. Diabe- tologia 1989;32:300–304.

81 Uusitupa MI, Laakso M, Sarlund H, Majander H, Takala J, Penttilä I: Effects of a very-low- calorie diet on metabolic control and cardiovas- cular risk factors in the treatment of obese non- insulin-dependent diabetics. Am J Clin Nutr 1990;51:768–773.

82 Fitzgerald AP, Jarrett RJ: Are conventional risk factors for mortality relevant in type 2 diabetes? Diabet Med 1991;8:475–480.

83 Ford ES, DeStefano F: Risk factors for mortality from all causes and from coronary heart disease among persons with diabetes. Findings from the National Health and Nutrition Examination Survey I Epidemiologic Follow-up Study. Am J Epidemiol 1991;133:1220–1230.

84 Laakso M, Lehto S, Penttilä I, Pyörälä K: Lipids and lipoproteins predicting coronary heart disease mortality and morbidity in patients with non-insulin-dependent diabetes. Circulation 1993;88:1421–1430.

85 Janghorbani M, Jones RB, Gilmour WH, Hedley AJ, Zhianpour M: A prospective popu- lation based study of gender differential in mortality from cardiovascular disease and ‘all causes’ in asymptomatic hyperglycaemics. J Clin Epidemiol 1994;47:397–405.

86 Collins VR, Dowse GK, Ram P, Cabealawa S, Zimmet PZ: Non-insulin-dependent diabetes and 11-year mortality in Asian Indian and Mela- nesian Fijians. Diabet Med 1996;13: 125–132.

87 Muggeo M, Verlato G, Bonora E, Zoppini G, Corbellini M, de Marco R: Long-term instability of fasting plasma glucose, a novel predictor of cardiovascular mortality in elderly patients with noninsulin-dependent diabetes mellitus: the Verona Diabetes Study. Circulation 1997;96: 1750–1754.

88 Niskanen L, Turpeinen A, Penttilä I, Uusitupa MI: Hyper-glycemia and compositional lipopro- tein abnormalities as predictors of cardiovascu- lar mortality in type 2 diabetes: a 15-year follow- up from the time of diagnosis. Diabetes Care 1998;21:1861–1869.

89 Hänninen J, Takala J, Keinänen- Kiukaanniemi S: Albuminuria and other risk factors for mortality in patients with non-insulin- dependent diabetes mellitus aged under 65 years: a population-based prospective 5-year study. Diabetes Res Clin Pract 1999;43: 121–126.

90 Forrest KY, Becker DJ, Kuller LH, Wolfson SK, Orchard TJ: Are predictors of coronary heart disease and lower-extremity arterial disease in type 1 diabetes the same? A prospective study. Atherosclerosis 2000;148:159–169.

91 Ostgren CJ, Lindblad U, Melander A, Råstam L: Survival in patients with type 2 diabetes in a Swedish community: Skaraborg hypertension and diabetes project. Diabetes Care 2002;25: 1297–302.

92 Roselli della Rovere G, Lapolla A, Sartore G, Rossetti C, Zambon S, Minicuci N, Crepaldi G, Fedele D, Manzato E: Plasma lipoproteins, apoproteins and cardiovascular disease in type 2 diabetic patients. A nine-year follow-up study. Nutr Metab Cardiovasc Dis 2003;13:46–51.

93 Chan WB, Tong PC, Chow CC, So WY, Ng MC, Ma RC, Osaki R, Cockram CS, Chan JC: Triglyceride predicts cardiovascular mortality and its relationship with glycaemia and obesity in Chinese type 2 diabetic patients. Diabetes Metab Res Rev 2005;21:183–188.

94 Zoccali C, Bode-Böger S, Mallamaci F, Benedetto F, Tripepi G, Malatino L, Cataliotti A, Bellanuova I, Fermo I, Frölich J, Böger R: Plasma concentration of asymmetrical dimethyl- arginine and mortality in patients with end-stage renal disease: a prospective study. Lancet 2001;358:2113–2117.

95 Bellomo G, Lippi G, Saronio P, Reboldi G, Verdura C, Timio F, Timio M: Inflammation, infection and cardio-vascular events in chronic hemodialysis atients: a prospective study. J Nephrol 2003;16:245–251.

96 Gertler MM, et al: Long-term follow-up study of young coronary patients. Am J Med Sci 1964;247:145–155.

97 Frank CW, et al: Angina pectoris in men. Prognostic significance of selected medical factors. Circulation 1973;47:509–517.

98 Mulcahy R, et al: Factors influencing long-term prognosis in male patients surviving a first coronary attack. Br Heart J 1975;37: 158–165.

99 Khaw KT, Barrett-Connor E: Prognostic factors for mortality in a population-based study of men and women with a history of heart disease. J Cardiopulm Rehab 1986;6:474–480.

100 Chester M, et al: Identification of patients at high risk for adverse coronary events while awaiting routine coronary angioplasty. Br Heart J 1995;73:216–222.

101 Behar S, et al: Low total cholesterol is associated with high total mortality in patients with coronary heart disease. The Bezafibrate Infarction Prevention (BIP) Study Group. Eur Heart J 1997;18:52–59.

102 Jacobs D, Blackburn H, Higgins M, et al: Report of the conference on low blood chole- sterol: mortality associations. Circulation 1992;86:1046–1060.

103 Ravnskov U: High cholesterol may protect against infections and atherosclerosis. QJM 2003;96:927–934.

104 Behar S, Graff E, Reicher-Reiss H, Boyko V, Benderly M, Shotan A, Brunner D: Low total cholesterol is associated with high total mor- tality in patients withcoronary heart disease. The Bezafibrate Infarction Prevention (BIP) Study Group. Eur Heart J 1997;18: 52–59.

105 Casiglia E, Spolaore P, Ginocchio G, Colangeli G, Di Menza G, Marchioro M, Mazza A, Ambrosio GB: Predictors of mortality in very old subjects aged 80 years or over. Eur J Epidemiol 1993;9:577–586.

106 Chyou PH, Eaker ED: Serum cholesterol concentrations and all-cause mortality in older people. Age Ageing 2000;29:69–74

107 Forette B, Tortrat D, Wolmark Y: Chole- sterol as risk factor for mortality in elderly women. Lancet 1989;1:868–870.

108 Fried LP, Kronmal RA, Newman AB, Bild DE, Mittelmark MB, Polak JF, Robbins JA, Gardin JM: Risk factors for 5-year mortality in older adults: the Cardiovascular Health Study. JAMA 1998;279:585–592.

109 Harris T, Feldman JJ, Kleinman JC, Ettinger WH Jr,Makuc DM, Schatzkin AG: The low cholesterol mortality association in a national cohort. J Clin Epidemiol 1992;45: 595–601.

110 Jonsson A, Sigvaldason H, Sigfusson N: Total cholesterol and mortality after age 80 years. Lancet 1997;350:1778–1779.

111 Krumholz HM, Seeman TE, Merrill SS, Mendes de Leon CF, Vaccarino V, Silverman DI, Tsukahara R, Ostfeld AM, Berkman LF: Lack of association between cholesterol and coronary heart disease mortality and morbidity and all-cause mortality in persons older than 70 years. JAMA 1994;272:1335–1340.

112 Räihä I, Marniemi J, Puukka P, Toikka T, Ehnholm C, Sourander L: Effect of serum lipids, lipoproteins, and apolipoproteins on vascular and nonvascular mortality in the elderly. Arterioscler Thromb Vasc Biol 1997;17: 1224–1232.

113 Rudman D, Mattson DE, Nagraj HS, Caindec N, Rudman IW, Jackson DL: Ante- cedents of death inthe men of a Veterans Administration nursing home. J Am Geriatr Soc 1987;35:496–502.

114 Schatz IJ, Masaki K, Yano K, Chen R, Rodriguez BL, Curb JD: Cholesterol and all-cause mortality in elderly people from the Honolulu Heart Program: a cohort study. Lancet 2001;358:351–355.

115 Schupf N, Costa R, Luchsinger J, Tang MX, Lee JH, Mayeux R: Relationship between plasma lipids and all-cause mortality in non- demented elderly. J Am Geriatr Soc 2005;53: 219–226.

116 Siegel D, Kuller L, Lazarus NB, Black D, Feigal D, Hughes G, Schoenberger JA, Hulley SB: Predictors of cardiovascular events and mortality in the Systolic Hypertension in the Elderly Program pilot project. Am J Epidemiol 1987;126:385–399.

117 Staessen J, Amery A, Birkenhager W, Bulpitt C, Clement D, de Leeuw P, Deruyttere M, De Schaepdryver A, Dollery C, Fagard R, et al: Is a high serum cholesterol level associated with longer survivalin elderly hypertensives? J Hypertens 1990;8:755–761.

118 Ulmer H, Kelleher C, Diem G, Concin H: Why Eve is not Adam: prospective follow-up in 149650 women and men of cholesterol and other risk factors related to cardiovascular and all-cause mortality. J Womens Health 2004;13: 41–53.

119 Weverling-Rijnsburger AW, Blauw GJ, Lagaay AM, Knook DL, Meinders AE, Westendorp RG: Total cholesterol and risk of mortality in the oldest old. Lancet 1997;350: 1119–1123.

120 Akerblom JL, Costa R, Luchsinger JA, Manly JJ, Tang MX, Lee JH, Mayeux R, Schupf N: Relation of plasma lipids to all-cause morta- lity in Caucasian, African- American and Hispanic elders. Age Ageing 2008;37:207–213.

121 Miettinen TA, Gylling H: -Mortality and cholesterol metabolism in familial hyper- cholesterolemia. Longterm follow-up of 96 patients. Arteriosclerosis 1988;8:163–167.

122 Hill JS, Hayden MR, Frohlich J, Pritchard PH: Genetic and environmental factors affecting the incidence of coronary artery disease in heterozygous familial hyper- cholesterolemia. Arterioscler Thromb 1991; 11:290– 7.

123 Ferrieres J, Lambert J, Lussier-Cacan S, Davignon J: Coronary artery disease in hetero- zygous familial hypercholesterolemia patients with the same LDL receptor gene mutation. Circulation 1995;92:290–295.

124 Kroon AA, et al: The prevalence of peri- pheral vascular disease in familial hyperchole- sterolaemia. J Intern Med 1995;238:451–459.

125 Hopkins PN, Stephenson S, Wu LL, Riley WA, Xin Y, Hunt SE: Evaluation of coronary risk factors in patients with heterozygous famlial hypercholesterolema. Am J Cardiol 2001;87:47–53.

126 de Sauvage Nolting PR, Defesche JC, Buirma RJ, Hutten BA, Lansberg PJ, Kastelein JJ: Prevalence and significance of cardiovas- cular risk factors in a large cohort of patients with familial hypercholesterolaemia. J Intern Med 2003;253:161–168.

127 Neil HA, Seagroatt V, Betteridge DJ, Cooper MP, Durrington PN, Miller JP, et al: Established and emerging coronary risk factors in patients with heterozygous familial hyper- cholesterolaemia. Heart 2004;90:1431–1437.

128 Jansen AC, van Aalst-Cohen ES, Tanck MW, ChengS, Fontecha MR, Li J, et al: Genetic determinants of cardiovascular disease risk in familial hyperchole sterol- emia.Vasc Arterioscler Thromb Biol 2005;25:1475–1481.

129 Skoumas I, Masoura C, Pitsavos C, Tousoulis D,Papadimitriou L, Aznaouridis K, Chrysohoou C,Giotsas -N, Toutouza M, Tentolouris C, Antoniades C, Stefanadis C: Evidence that non-lipid cardiovascular risk factors are associated with high prevalence of coronary artery disease in patients with hetero- zygous familial hypercholesterolemia or familial combined hyperlipidemia. Int J Cardiol 2007; 121:178–183.

130 Postiglione A, Nappi A, Brunetti A, Soricelli A, Rubba P, Gnasso A, Cammisa M, Frusciante V, Cortese C, Salvatore M, et al: Relative protection from cerebral atheroscle- rosis of young patients with homozygous fa- milial hypercholesterolemia. Atherosclerosis 1991;90:23–30.

131 Rodriguez G, Bertolini S, Nobili F, Arrigo A, Masturzo P, Elicio N, Gambaro M, Rosadini G: Regional cerebral blood flow in familial hypercholesterolemia. Stroke 1994;25: 831–836.

132 Brown MS, Goldstein JL: Lipoprotein metabolism in the macrophage: implications for cholesterol deposition in atherosclerosis. Annu Rev Biochem 1983;52:223–261.

133 Neil HA, Hawkins MM, Durrington PN, Betteridge DJ, Capps NE, Humphries SE, Simon Broome Familial Hyperlipidaemia Re- gister Group and Scientific Steering Commit- tee: Non-coronary heart disease mortality and risk of fatal cancer in patients with treated heterozygous familial hypercholesterolaemia: a prospective registry study. Atherosclerosis 2005;179:293–297.

134 Sugrue DD, Trayner I, Thompson GR, Vere TV, Dimeson J, Stirling Y, et al: Coronary artery disease and haemostatic variables in heterozygous familial hyper-cholesterolaemia. Br Heart J 1985;53:265–268.

135 Ravnskov U: Cholesterol lowering trials in coronary heart disease: frequency of citation and outcome. BMJ 1992;305:15–19.

136 Ravnskov U: Implications of 4S evidence on baseline lipid levels. Lancet 1995;346:181.

137 Todd EW, Coburn AF, Hill AB: Antistreptolysin S titres in rheumatic fever. Lancet 1939;2:1213–1217.

138 Stollerman GH, Bernheimer AW: Inhibition of strepto-lysin S by the serum of patients with rheumatic fever and acute streptococcal pharyngitis. J Clin Invest 1950;29:1147–1155.

139 Humphrey JH: The nature of antistrepto- lysin S in the sera of man and of other species; the lipoprotein properties of antistreptolysin S. Br J Exp Pathol 1949;30: 365–375.

140 Stollerman GH, Bernheimner AW, MacLeod CM: The association of lipoproteins with the inhibition of streptolysin S by serum. J Clin Invest 1950;29:1636–1645.

141 Skarnes RC: In vivo interaction of endo- toxin with a plasma lipoprotein having esterase activity. J Bacteriol 1968;95:2031–2034.

142 Shortridge KF, Ho WK, Oya A, Kobayashi M: Studies on the inhibitory activities of human serum lipoproteins for Japanese encephalitis virus. Southeast Asian J Trop Med Public Health 1975;6:461–466.

143 Whitelaw DD, Birkbeck TH: Inhibition of staphylo-coccal delta-haemolysin by human serum lipoproteins. FEMS Microbiology Letters 1978;3:335–338.

144 Freudenberg MA, Galanos C: Interaction of lipopoly-saccharides and lipid A with comple- ment in rats and its relation to endotoxicity. Infect Immun 1978;19:875–882.

145 Ulevitch RJ, Johnston AR, Weinstein DB: New function for high density lipoproteins. Isolation and charac-terization of a bacterial lipopolysaccharidehigh density lipoprotein complex formed in rabbit plasma. J Clin Invest 1981;67:827–837.

146 Bhakdi S, Tranum-Jensen J, Utermann G, Fussle R: Binding and partial inactivation of Staphylococcus aureus alpha-toxin by human plasma low density lipoprotein. J Biol Chem 1983;258:5899–5904.

147 Seganti L, Grassi M, Mastromarino P, Pana A, Superti F, Orsi N: Activity of human serum lipoproteins on the infectivity of rhabdo- viruses. Microbiologica 1983;6:91–99.

148 Van Lenten BJ, Fogelman AM, Haberland ME, Edwards PA: The role of lipoproteins and receptormediated endocytosis in the transport of bacterial lipopolysaccharide. Proc Natl Acad Sci USA 1986;83:2704–2708.

149 Huemer HP, Menzel HJ, Potratz D, Brake B, Falke D, Utermann G, et al: Herpes simplex virus binds to human serum lipoprotein. Inter- virology 1988;29:68–76.

150 Flegel WA, Wölpl A, Männel DN, Northoff H: Inhi-bition of endotoxin-induced activation of human monocytes by human lipoproteins. Infect Immun 1989;57:2237–2245.

151 Cavaillon JM, Fitting C, Haeffner-Cavaillon N, Kirsch SJ, Warren HS: Cytokine response by monocytes and macrophages to free and lipoproteinbound lipopoly-saccharide. Infect Immun 1990;58:2375–2382.

152 Northoff H, Flegel WA, Yurttas R, Wein- stock C: The role of lipoproteins in inactivation of endotoxin by serum. Beitr Infusionsther 1992;30:195–197.

153 Superti F, Seganti L, Marchetti M, Marziano ML, Orsi N: SA-11 rotavirus binding to human serum lipoproteins. Med Microbiol Immunol 1992;181:77–86.

154 Weinstock C, Ullrich H, Hohe R, Berg A, Baumstark MW, Frey I, et al: Low density lipoproteins inhibit endo-toxin activation of monocytes. Arterioscler Thromb 1992;12: 341–347.

155 Flegel WA, Baumstark MW, Weinstock C, Berg A, Northoff H: Prevention of endotoxin- induced monokine release by human low- and high-density lipoproteins and by apolipoprotein A-I. Infect Immun 1993;61:5140–5146.

156 Feingold KR, Funk JL, Moser AH, Shige- naga JK, Rapp JH, Grunfeld C: Role for cir- culating lipoproteins in protection from endo- toxin toxicity. Infect Immun 1995;63: 2041–2046.

157 Netea MG, Demacker PNM, Kullberg BJ, Boerman OC, Verschueren I, Stalenhoef AFH, et al: Lowdensity lipoprotein receptor-deficient mice are protected against lethal endotoxemia and severe Gram-negative infections. J Clin Invest 1996;97:1366–1372.

158 Iribarren C, Jacobs DR Jr, Sidney S, Claxton AJ, Gross MD, Sadler M, Blackburn H: Serum total cholesterol and risk of hospitali- zation, and death from respiratory disease. Int J Epidemiol 1997;26:1191–1202.

159 Iribarren C, Jacobs DR Jr, Sidney S, Claxton AJ, Feingold KR: Cohort study of serum total cholesterol and in-hospital incidence of infectious diseases. Epidemiol Infect 1998;121:335–347.

160 Rauchhaus M, Coats AJ, Anker SD: The endotoxin- lipoprotein hypothesis. Lancet 2000;356:930–933.

161 Claxton AJ, Jacobs DR Jr, Iribarren C, Welles SL, Sidney S, Feingold KR: Asso- ciation between serum total cholesterol and HIV infection in a high-risk cohort of young men. J Acquir Immune Defic Syndr Hum Retrovirol 1998;17:51–57.

162 Neaton JD, Wentworth DN: Low serum cholesterol and risk of death from AIDS. AIDS 1997;11:929–930

163 Chen Z, Keech A, Collins R, Slavin B, Chen J, Campbell TC, Peto R: Prolonged infection with hepatitis B virus and association between low blood cholesterol concentration and liver cancer. BMJ 1993;306:890–894.

164 Fraunberger P, Hahn J, Holler E, Walli AK, Seidel D: Serum cholesterol levels in neutro- penic patients with fever. Clin Chem Lab Med 2002;40:304–307.

165 Elias ER, Irons MB, Hurley AD, Tint GS, Salen G: Clinical effects of cholesterol supplementation in six patients with the Smith-Lemli-Opitz syndrome (SLOS). Am J Med Genet 1997;68:305–310.

166 Sijbrands EJ, Westendorp RG, Defesche JD, de Meier PH, Smelt AH, Kastelein JJP: Mortality over two centuries in large pedigree with familial hypercholeste-rolaemia: family tree mortality study. BMJ 2001;322: 1019–1023.

167 Williams RR, Sorlie PD, Feinleib M, McNamara PM, Kannel WB, Dawber TR: Cancer incidence by levels of cholesterol. JAMA 1981;245:247–252.

168 Newman TB, Hulley SB: Carcinogenicity of  lipid-lowering drugs. JAMA 1996;275:55–60.

169 Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease the Scandinavian Simvastatin Survival Study (4S). Lancet 1994;344:1383–1389.

170 Heart Protection Study Collaborative Group: MRC/BHF heart protection study of cholesterol lowering in 20536 high-risk individuals: a randomised placebo-controlled trial. Lancet 2002;360:7–22.

171 Sacks FM, Pfeffer MA, Moye LA, et al, for the Cholesterol and Recurrent Events Trial investigators: The effect of pravastatin on coronary events after myocardial infarction in patients with average cholesterol levels. N Engl J Med 1996;335:1001–1009.

172 Shepherd J, Blauw GJ , Murphy MB, et al: Pravastatin in elderly individuals at risk of vas cular disease (PROSPER): a randomised controlled trial. Lancet 2002; 360:1623–1630.

173 Matsuzaki M, Kita T, Mabuchi H, et al: Large scale cohort study of the relationship between serum cholesterol concentration and coronary events with low-dose simvastatin therapy in Japanese patients with hyper- cholesterolemia. Circ J 2002;66:1087–1095.

174 Iwata H, Matsuo K, Hara S, et al: Use of  hydroxymethyl-glutaryl coenzyme A reductase inhibitors is associated with risk of lymphoid malignancies. Cancer Sci 2006;97:133–138.

175 Hansson GK: Inflammation, atheroscle- rosis, and coro-nary artery disease. N Engl J Med 2005;352:1685–1695.

176 Reis SE, Holubkov R, Conrad Smith AJ, Kelsey SF, Sharaf BL, Reichek N, Rogers WJ, Merz CN, Sopko G, Pepine CJ: Coronary microvascular dysfunction is highly prevalent in women with chest pain in the absence of coro- nary artery disease: results from the NHLBI WISE study. Am Heart J 2001;141:735–741.

177 McCully KS: Vascular pathology of homo- cysteinemia: implications for the pathogenesis of arteriosclerosis. Am J Pathol 1969;56: 111–128.

178 Ravnskov U, McCully KS: Vulnerable plaque  formation from obstruction of vasa vasorum by homo-cysteinylated and oxidized lipoprotein aggregates complexed with micro- bial remnants and LDL autoanti-bodies. Ann Clin Lab Sci 2009;39:3–16.

179 Benesch R, Benesch RE: Thiolation of proteins. Proc Natl Acad Sci USA 1958;44: 848–853.

180 Naruszewicz M, Mirkiewicz E, Olszewski AJ, McCully KS: Thiolation of low-density lipoprotein by homo- cysteine thiolactone causes increased aggregation and altered interaction with cultured macrophages. Nutr Metab Cardiovas Dis 1994;64:70–77.

181 Ferguson E, Parthasarathy S, Joseph J, Kalyanaraman B: Generation and initial cha- racterization of a novel poly-clonal antibody directed against homocysteine thiolactone- modified low density lipoprotein. J Lipid Res 1998;39:925–933.

182 Heinecke JW, Suits AG, Aviram M, Chait A: Phago-cytosis of lipase-aggregated low density lipoprotein promotes macrophage foam cell formation. Sequential morphological and biochemical events. Arterioscler Thromb 1991; 11:1643–1651.

183 Kalayoglu MV, Indrawati, Morrison RP, Morrison SG, Yuan Y, Byrne GI: Chlamydial virulence determinants in atherogenesis: the role of chlamydial lipopolysaccharide and heat shock protein 60 in macrophage-lipoprotein interactions. J Infect Dis 2000;181(suppl 3): S483–S489.

184 Qi M, Miyakawa H, Kuramitsu HK: Por- phyromonas gingivalis induces murine macro- phage foam cell formation. Microb Pathog 2003;35:259–267.

185 Ott SJ, El Mokhtari NE, Musfeldt M, Hellmig S, Freitag S, Rehman A, Kuhbacher T, Nikolaus S, Namsolleck P, Blaut M, Hampe J, Sahly H, Reinecke A, Haake N, Gunther R, Kruger D, Lins M, Herrmann G, Folsch UR, Simon R, Schreiber S: Detection of diverse bacterial signatures in atherosclerotic lesions of patients with coronary heart disease. Circulation 2006;113:29–37.

186 Melnick JL, Petrie BL, Dreesman GR, Burek J, McCollum CH, DeBakey ME: Cyto- megalovirus antigen within human arterial smooth muscle cells. Lancet 1983;2:644–647.

187 Pampou SY, Gnedoy SN, Bystrevskaya VB, Smirnov VN, Chazov EI, Melnick JL, DeBakey ME: Cytomegalo-virus genome and the immediate-early antigen in cells of different layers of human aorta. Virchows Arch 2000;436:539–552.

188 Naruko T, Ueda M, Haze K, van der Wal AC, van der Loos CM, Itoh A, Komatsu R, Ikura Y, Ogami M, Shimada Y, Ehara S, Yoshiyama M, Takeuchi K, Yoshikawa J, Becker AE: Neutrophil infiltration of culprit lesions in acute coronary syndromes. Circulation 2002;106:2894–2900.

189 Madjid M, Naghavi M, Malik BA, Litovsky S Willerson JT, Casscells W: Thermal detec- tion of vulnerable plaque. Am J Cardiol 2002; 90:36L–39L.

190 Guyton JR, Klemp KF: Transitional features in human atherosclerosis. Intimal thickening, cholesterol clefts, and cell loss in human aortic fatty streaks. Am J Pathol 1993;143:1444–1457.

191 Kohsaka S, Menon V, Lowe AM, Lange M, Dzavik V, Sleeper LA, Hochman JS, SHOCK Investigators: Systemic inflammatory response syndrome after acute myocardial infarction complicated by cardiogenic shock. Arch Intern Med 2005;165:1643–1650

.

I am an independent medical researcher