spiral.imperial.ac.uk:8443€¦  · web vieweffects of dyslipidaemia on monocyte production and...

63
Effects of dyslipidaemia on monocyte production and function in cardiovascular disease Shamim Rahman 1 , Andrew J. Murphy 2,3 and Kevin J. Woollard 1 1 Renal & Vascular Inflammation Section, Division of Immunology and Inflammation, Imperial College London, Du Cane Road, London W12 0NN, UK. 2 Haematopoiesis and leukocyte biology lab, Baker IDI Heart & Diabetes Research Institute, 75 Commercial Rd, Melbourne VIC 3004 Australia. 3 Department of Immunology, Monash University, Melbourne, Victoria, 3004, Australia. Correspondence to K.J.W. [email protected] Abstract | Monocytes are heterogeneous effector cells involved in the maintenance and restoration of tissue integrity. Hyperlipidaemia can accelerate cardiovascular disease progression. Monocytes and macrophages are involved in cardiovascular disease progression, and are associated with the development of unstable atherosclerotic plaques. However, monocyte responses to hyperlipidaemia are poorly understood. In the past decade, accumulating data describe the relationship between the dynamic, blood lipid environment and the heterogeneous circulating monocyte pool, which might have profound consequences for cardiovascular disease. In this Review, we explore the updated view of monocytes in cardiovascular disease and their relationship with macrophages 1

Upload: others

Post on 26-Sep-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Effects of dyslipidaemia on monocyte production and function in cardiovascular disease

Shamim Rahman1, Andrew J. Murphy2,3 and Kevin J. Woollard1

1Renal & Vascular Inflammation Section, Division of Immunology and Inflammation, Imperial

College London, Du Cane Road, London W12 0NN, UK.

2Haematopoiesis and leukocyte biology lab, Baker IDI Heart & Diabetes Research Institute,

75 Commercial Rd, Melbourne VIC 3004 Australia.

3Department of Immunology, Monash University, Melbourne, Victoria, 3004, Australia.

Correspondence to K.J.W. [email protected]

Abstract | Monocytes are heterogeneous effector cells involved in the maintenance and

restoration of tissue integrity. Hyperlipidaemia can accelerate cardiovascular disease

progression. Monocytes and macrophages are involved in cardiovascular disease

progression, and are associated with the development of unstable atherosclerotic plaques.

However, monocyte responses to hyperlipidaemia are poorly understood. In the past

decade, accumulating data describe the relationship between the dynamic, blood lipid

environment and the heterogeneous circulating monocyte pool, which might have profound

consequences for cardiovascular disease. In this Review, we explore the updated view of

monocytes in cardiovascular disease and their relationship with macrophages in promoting

the homeostatic and inflammatory responses related to atherosclerosis. We describe the

different definitions of dyslipidaemia, highlight current theories on the ontogeny of

monocyte heterogeneity, discuss how dyslipidaemia might alter monocyte production, and

explore the mechanistic interface linking dyslipidaemia with monocyte effector functions,

such as migration and the inflammatory response. Finally, we discuss the role of dietary and

endogenous lipid species in mediating dyslipidaemic responses, and the role of these lipids

in promoting the risk of cardiovascular disease through modulation of monocyte behaviour.

1

Page 2: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Investigation of monocytes and macrophages has had a resurgence of activity over the past

10 years. This increase in monocyte and macrophage research is a result of a refinement of

the definition and characterization of the mononuclear phagocyte system. These

phagocytes make up diverse populations of cells with tissue-specific specialization to

maintain homeostasis, such as Kupffer cells in the liver and alveolar macrophages in the

lung1. Some of these tissue-specific macrophages can maintain their population levels

through in situ proliferation, and do not require continuous replenishment via blood

monocytes2. Therefore, the role of blood monocytes as short-lived macrophage precursors

might need redefinition and deeper characterization. Moreover, monocytes populations are

heterogeneous and have subset specific effector functions3, which are altered as a result of

changes in the local environment and inflammatory response. This Review focuses on

dyslipidaemia and changes in blood lipid profiles that might alter the behaviour of

monocytes, thereby contributing to cardiovascular disease.

[H1] Steady-state production and function

[H2] Origin of tissue-resident macrophages

The identification of fetal-derived, self-maintaining, tissue-resident macrophages has shifted

our understanding of myeloid lineages away from the previously accepted dogma, which

postulated as the core definition of the mononuclear phagocyte system that short-lived,

bone marrow-derived monocytes are required to replenish tissue macrophages 4.

Monocytes have long been known to contribute to the tissue macrophage pool, particularly

during infection and inflammation5. However, the relative contribution of monocytes to

tissue macrophage populations in the steady state has been under scrutiny. Increasingly

sophisticated experiments with fate-mapping technology, adoptive transfer, and parabiosis

models have helped to improve our understanding of the complex contributions of different

cell lineages throughout embryological development to haematopoiesis and the seeding of

tissues with resident macrophage precursors. Data now indicate that monocytes have a

minimal contribution to most tissue-resident macrophage pools in the steady state6,7 or

even under certain inflammatory conditions. Many tissue-resident macrophage populations

have now been identified, but tissue-specific differences cannot yet be fully explained.

2

Page 3: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Tissue macrophage ontogeny can be divided into the early ‘primitive’ and subsequent

‘definitive’ haematopoietic waves1, with the bulk of experimental data derived from mouse

models. Primitive haematopoiesis refers to early formation of haematopoietic progenitors,

including macrophage progenitors (or ‘primitive macrophages’) and erythro-myeloid

progenitors, from blood islands of the yolk sac in mice8 and rats9 at embryonic day 8.5–9.0

(E8.5–E9.0), with subsequent colonization of the fetal liver at E10.0–E10.5 via the newly

established circulatory system10,11. Definitive haematopoiesis in the aorta–gonad–

mesonephros (AGM) results in the release of haematopoietic stem cells (HSCs) at E10.5,

which then colonize the fetal liver (E11.0)12. Thus, from E11.0 to E11.5 the fetal liver

becomes the dominant site of haematopoiesis1, before the initiation of bone marrow-

derived haematopoiesis.

Yolk sac-derived primitive macrophages, colonize tissues such as the brain, spleen, pancreas,

kidney, heart and lungs7,13-16. Fetal liver monocytes directly derived from cMyb+ erythro-

myeloid progenitors generated in dorsal aortic endothelial cells17, subsequently adds to the

tissue-resident macrophage population. Exceptions to this fetal monocytic infiltrate includes

the central nervous system, where fetal liver-derived monocytes do not contribute to the

microglial cell population14; the skin, where dual-origin macrophages contribute to the

Langerhans cell pool18; and the gut, which seems to remain dependent on constant renewal

from blood precursors, with data suggesting circulating blood monocytes contribute to the

pool of macrophages and dendritic cells in the intestine19,20.

[H2] Monocyte development

Monocyte development and survival depends on colony-stimulating factor 1 (CSF1; also

known as M-CSF) and its receptor CFS1R (also known as CD115)21-23. Embryonic and adult

haematopoiesis in the steady state results in monocyte development from myeloid

precursor cells found in primary lymphoid organs, including the fetal liver and bone

marrow1. In addition, in mice under some inflammatory conditions the spleen has been

shown also to contribute to monocyte production from precursor cells24.

Following the establishment of bone marrow progenitors, macrophage and dendritic cell

precursors (MDP) — defined as lineage-negative (Lin-), CD117+ (also known as KIT), CD135+

3

Page 4: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

(also known as FLT3), and CSF1R+ — give rise to monocytes, macrophages, and dendritic

cells25. However, the absolute requirement for MDP as a common precursor for monocyte

and macrophage lineages and resident dendritic cells is debated26.

A potential additional monocyte precursor, termed the common monocyte progenitor

(cMoP), has been identified27. cMoPs derive from, and also differ from, MDPs, and are

characterized by a reduction in the relative expression of CD135 (that is, cMoPs are Lin–

CD117+CD135–CSF1R+)27. cMoPs give rise to mature monocytes in the bone marrow. These

mature monocytes do not subsequently differentiate into either classical dendritic cells or

plasmacytoid dendritic cells 27. Thus in the adult, monocytes can arise from the bone

marrow via myelopoiesis. Myelopoiesis begins from haematopoietic stem and progenitor

cells (HSPCs), which give rise to common myeloid progenitors (CMPs), which in turn give rise

to granulocyte-macrophage progenitors (GMPs) and then cMoPs (FIG. 1). During times of

stress, such as infection, the myeloid branch of haematopoiesis can be expanded to increase

monocyte production through the action of a number of well-characterized cytokines5. This

process is also important in chronic inflammatory disorders, including metabolic disorders

and cardiovascular disease, where monocytosis is present (detailed below). Monocyte

subset production in the bone marrow and the factors influencing blood trafficking and

extravasation into tissue are represented in Figure 1.

[H2] Blood monocytes

Circulation in the blood is a defining characteristic of a monocyte3. Although previously

considered intermediary cells awaiting stimuli towards terminal differentiation into a

macrophage, dendritic cell, or other tissue descendant4,28,29, blood monocytes are now

known to be a heterogeneous population of cells with differing steady-state phenotypes and

as effector cells in response to microbial stimuli3. Furthermore, monocyte gene expression

profiles and functions are conserved across vertebrate species30.

Monocytes can be characterized by the expression levels of surface proteins, RNA

expression profiles and morphology3,31-33. Morphologically, monocytes are spherical cells

with prominent surface ruffles and blebs, a large reniform nucleus with a small nucleolus34.

Differences in size and granularity exist between subsets (detailed below), with non-classical

4

Page 5: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

cells much smaller and less granular than classical monocytes3. Heterogeneity in monocytes

exists in many species35, including humans, rats, mice, and pigs32,35-42. Two major monocyte

subsets have been identified in mice — ‘classical’ and ‘nonclassical’ — and three subsets

have been identified in humans — classical, intermediate, and nonclassical — with classical

monocytes being the most abundant (~80%) (TABLE 1). In mice, monocytes are commonly

identified in flow cytometry analyses by their characteristic forward and side scatter profiles

and expression of CD11543,44. Monocyte subsets can then be differentiated on the basis of

expression levels of a number of surface markers, including lymphocyte antigen 6C (LY6C),

CD117, CX3C-chemokine ligand 1 (CX3CR1), C-C chemokine receptor type 2 (CCR2), and

CD431,38,43. Classical monocytes are defined as LY6ChighCD117–CX3CR1intCCR2+CD62L+CD43low

(where “int” refers to intermediate). Nonclassical monocytes are

LY6ClowCD117intCX3CR1highCCR2–CD62L–CD43high 38,45-47.

Murine monocyte subsets have equivalents in human. Gene expression arrays show that

classical LY6Chigh mouse monocytes resemble human monocytes that express CD14highCD16low

surface markers32. Conversely, nonclassical LY6Clow monocytes are most analogous to human

CD14lowCD16high monocytes32. A third, ‘intermediate’ subset has been reported in humans,

defined by high levels of both CD14 and CD16. This intermediate subset aligns functionally

with murine classical cells3,48, but transcriptionally with nonclassical CD14lowCD16high

monocytes49. The subset of intermediate monocytes expands in a variety of infectious and

inflammatory conditions (for example, cardiovascular disease, rheumatoid arthritis, Crohn’s

disease, and chronic kidney disease)35,50.

Monocyte subsets differ in their response to pathogens. Classical monocytes respond

strongly to bacterial stimuli via Toll-like receptor (TLR) 4, nonclassical monocytes respond to

viral stimuli via the intracellular receptors TLR7 and TLR8, and intermediate monocytes

respond to both bacterial and viral stimuli producing high levels of inflammatory cytokines

such as interleukin (IL)-1. Therefore, intermediate monocytes might represent a subset in

transition between classical and nonclassical cells, displaying characteristics of both

subsets51. Indeed, evidence from murine models indicates that classical monocytes can

transition towards a nonclassical phenotype in bone marrow and blood7 and in the setting of

murine myocardial infarction and healing52, although this transition is yet to be confirmed in

other species and under different environmental conditions. Monocyte ontogeny remains

5

Page 6: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

enigmatic, particularly in the context of subset origins. For instance, what the half-life of

monocytes is and where monocytes eventually go are still unclear. The current hypothesis is

that both subsets develop from bone marrow progenitors. However, as mentioned

previously, data in mice suggest that in the steady state, LY6Chigh monocytes can give rise to

LY6Clow monocytes7,53, both in the bone marrow and in the blood, although evidence to

conclude confidently a linear relationship between both monocyte subsets is lacking.

Conversion of LY6Chigh to LY6Clow is further supported by stark differences in mouse

monocyte subset survival. Turnover of LY6Chigh monocytes is thought to be 1 day, whereas

LY6Clow monocytes can survive from 2 days to as long as 2 weeks under certain conditions7.

The transcription factor nuclear receptor subfamily 4 group A member 1 (NR4A1; also

known as NUR77) has an essential role in the development and survival of LY6Clow

monocytes54 (reviewed in REFS. 52,55-57). LY6Clow monocytes have significantly higher amounts

of NR4A1 than LY6Chigh cells at both the transcriptional52,54 and protein levels54. Nr4a1–/– mice

lack LY6Clow monocytes in the bone marrow, blood, and spleen, and do not have any

patrolling monocytes as assessed by intravital microscopy55. Moreover, MDP precursors that

would usually give rise to blood LY6Clow monocytes do not do so in NR4A1-deficient mice54.

These data suggest that either NR4A1 is involved in monocyte ‘conversion’ or that mature

LY6Chigh monocytes are in fact not the precursors of LY6Clow . One might speculate that three

monocyte subsets exist in mice: bona fide LY6Chigh, LY6Clow matured from LY6Chigh cells, and

LY6Clow patrolling monocytes at the endothelial interface derived from the differentiation of

precursors driven by NR4A1 (FIG. 1). Experiments are underway to examine this hypothesis.

Interestingly, homozygous deletion of Nr4a1 has been shown to accelerate atherosclerosis

in the high-fat diet (HFD)-fed Ldlr-/- mouse model of atherosclerosis in some studies58,59, but

not others60. Finally, definitions of monocyte subsets may be further complicated by recent

reports that LY6Chigh monocytes may be defined by CXCR4 expression in the bone marrow

giving rise to further functional subsets 61, and that a subpopulation of LY6Chigh monocytes

give rise to inflammatory dendritic cells62. Moreover, an ‘atyical’ monocyte with neutrophil

characteristics may be involved with fibrosis 63. More work is now needed to characterise

the ‘plasticity’ of monocytes.

[H2] Monocyte functions

6

Page 7: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Although the precise functions of monocyte subsets are yet to be defined, clear evidence

supports subset-specific functions. In mice, classical LY6Chigh monocytes are rapidly recruited

to sites of inflammation, extravasate and differentiate into monocyte-derived

macrophages64. By contrast, mouse nonclassical LY6Clow monocytes, similar to their human

counterparts3, crawl and patrol the luminal side of the vascular endothelium37,55, as shown

by intravital microscopy. Evidence showing the differentiation of LY6Clow monocytes from

LY6Chigh monocytes, the longer half-life of the LY6Clow subset, and its patrolling role have led

some investigators to suggest that these cells are terminally differentiated ‘blood

macrophages’ rather than blood monocytes1. However, we and other researchers have

shown monocytes can migrate out of the blood during dyslipidaemia (detailed below) or

autoimmune inflammation65-67, implying they are not terminally differentiated cells and can

continue to respond to tissue cues. Similarly, the question remains whether the highly

mobile, responsive LY6Chigh monocytes that extravasate into the tissue not always

differentiate into macrophages or dendritic cells, and can uptake antigens as part of a

tissue-surveillance function47.

Nonclassical LY6Clow monocytes, by patrolling the endothelium also have surveillance

functions. Through expression of surface molecules such as CD11a (also known as integrin

αL)55, CX3CR137, intercellular adhesion molecule (ICAM) 1 and ICAM236,68, and protein CYR61

(also known as CCN1)69, LY6Clow monocytes remain in close proximity to the endothelium

and can respond to endothelial damage, perhaps monitoring endothelial integrity and

responding to deposited immune complexes by recruiting inflammatory neutrophils55,70.

Moreover, these surveying monocytes might remove luminal vascular β-amyloid deposits,

as shown in a murine model of Alzheimer disease71. Nonclassical monocytes have also been

shown to scavenge metastasizing tumour cells in a lung tumour mouse model72. More work

is needed to identify the unique functional properties of nonclassical LY6clow monocytes.

Whether these nonclassical monocytes have a direct role in cardiovascular disease is

unclear, although these monocytes have been shown to patrol along the coronary vascular

tree73.

[H2] Monocytes in atherosclerosis

7

Page 8: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Atherosclerosis is a dynamic and chronic inflammatory disease, and thus seems to elicit very

different responses from blood monocytes and tissue-resident macrophages compared with

traditional inflammatory scenarios. Preclinical models of atherosclerosis involve

homozygous deletion of genes encoding proteins associated with lipoprotein clearance,

namely Ldlr and Apoe, in order to generate chronic hyperlipidaemia and the formation of

atherosclerotic plaques. These models have helped to shed light on monocyte production,

migration into the atherosclerotic plaque, and subsequent development of lipid-laden

macrophage foam cells (FIG. 2). In these preclinical models, the hypercholesterolaemic

environment 74-77 or the disruption of cholesterol efflux mediated by ABC transporters78,79

promotes monocytosis, an observation consistent with the monocytosis found in people

with cardiovascular disease80-82. This increase in blood monocytes is a result of an expansion

of the bone marrow progenitors HSPCs, CMPs, and GMPs. Importantly, use of competitive

bone marrow transplantation experiments allowed the tracking of the HSPC progeny into

the atherosclerotic lesion, revealing a preferential accumulation in atherosclerotic lesions of

Apoe–/– monocytes and macrophages compared with wild type cells 74. Interestingly,

monocytosis in the bone marrow and blood compartments might be dependent on

chemokine signalling, specifically mediated by C-C motif chemokine 2 (CCL2), CCR2, CX3CR1,

and C-C chemokine receptor type 5 (CCR5)76,83. Moreover, these chemokines have a pro-

atherogenic role, as shown by the reduction in atherosclerotic plaque size following

combined inhibition of CCL2, CX3CR1, and CCR5 despite higher total cholesterol levels

present in Apoe-knockout mice 83. These studies on atherogenesis subscribe to the dogmatic

model of circulating monocyte extravasation and differentiation into macrophages, followed

by subsequent accumulation in macrophages of modified lipids via scavenger-receptor

uptake to generate foam cells. Bone marrow transplantation models in atherosclerosis-

prone mice indicate that an ’inflammatory’ (LY6Chigh) monocyte subset is readily recruited

into the arterial intimal layer84,85. Atherosclerosis progression depends on the presence and

viability of this monocyte subset, and recruitment from this monocyte pool continues during

lesion growth84-86. Importantly, this inflammatory or classical subset is the predominant

subset to extravasate during atherogenesis. However, reports show nonclassical LY6Clow

monocyte migration to discrete atherosclerotic regions87. This migration might be important

during specific types of dyslipidaemia associated with atherosclerosis, such as

8

Page 9: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

hypertriglyceridaemia, which has been associated with increased tissue recruitment of

nonclassical monocytes66.

The idea that plaque macrophages arise strictly from continued recruitment of blood

monocytes has been challenged. In 2013, a study demonstrated through a series of elegant

experiments that maintenance and expansion of the lesional pool of macrophages in

established lesions is mainly sustained by local cell proliferation rather than by continuous

recruitment of blood monocytes 2 (FIG. 2a). Interestingly, the study also showed that the

macrophage area in the growing lesions remained unchanged2. These paradoxical results

are difficult to reconcile, but an explanation might be that most of the newly recruited

monocytes undergo rapid cell death, whereas a small pool of macrophages proliferate but

then die after a few divisions 44. However, in contrast to the short-term experiments, long-

term follow-up revealed that almost all lesional macrophages were replaced by newly

recruited cells2 . Collectively, the findings from this study highlight the importance of

monocyte recruitment, macrophage proliferation, and macrophage death during

atherosclerosis progression88.

A population of tissue-resident macrophages, termed adventitial macrophages, has been

identified in the arterial adventitia89. In adulthood, these adventitial macrophages derive

from a pool of adventitial macrophage progenitor cells89. These adventitial macrophages

also derive embryonically from CX3CR1+ erythro-myeloid progenitors (from the yolk sac) and

from fetal liver monocytes 90. Interestingly, the adventitial macrophage population has been

shown to expand in the setting of hyperlipidaemia 89. However, these adventitial

macrophages seem to be a minority within the macrophage pool in the atheroma89, but

might have an important indirect role through the release of cytokines or by inducing

neovascularization.

The above studies focussed on plaque macrophage populations during atherogenesis.

However, recurrent insults, such as those frequently following an acute myocardial

infarction (MI), initiate an additional wave of altered haematopoiesis91. Interestingly, an

increased macrophage burden was observed within atherosclerotic lesions after an acute

MI91. The increase in macrophage burden was independent of local cell proliferation and

was a result of enhanced monocyte recruitment from the spleen91. The acute MI caused

sympathetic overdrive, which mobilises HSPCs from the bone marrow into the spleen,

9

Page 10: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

where HSPCs undergo extramedullary haematopoiesis and give rise to monocytes involved

in atherosclerosis91. Furthermore, in addition to the role of blood monocyte-derived

macrophages in atherosclerotic lesion progression, monocytes and macrophages with mixed

ontogenies are important in driving an inflammatory response and subsequent repair of the

myocardium after an acute MI92.

[H1] Dyslipidaemia

Dyslipidaemia encompasses blood lipid levels beyond the normal range either when

elevated or low, however is most commonly applied when lipid levels associated with

cardiovascular disease are elevated, such as LDL or when low levels of HDL cholesterol are

found93. Elevations in blood cholesterol levels, in particular are linked with an increased risk

of cardiovascular disease94. The blood lipid pool is made up of lipoproteins that transport

cholesterol, cholesterol esters and fatty acids predominantly as triglycerides to muscle,

adipose tissue, and to the liver for processing and clearance (BOX 1). Free, nonesterified

fatty acids are also thought to be present in the blood, most probably bound to albumin95.

Experimental animal models and human histopathological samples confirm the presence of

cholesterol in atherosclerotic plaques, predominantly in foam cells (thought to be lipid-

laden macrophages)96. Fatty acids have also been shown to accumulate in the

atherosclerotic plaque97. Interestingly, this accumulation is dynamic and the composition of

the fatty acid pool changes over time, which might provide selective stimuli to foam cells

and might represent a dietary influence on plaque composition97.

[H2] Classification

Dyslipidaemias can be classified as primary if the dyslipidaemia has a genetic cause (familial

or inherited dyslipidaemia) or as secondary if the dyslipidaemia results from other medical

conditions (BOX 1). The Frederickson classification98 of familial dyslipidaemia has been

adopted by the WHO99, and remains one of the most frequently used in clinical decision

making, however does not account for HDL nor for gene variants associated with abnormally

elevated lipid levels. For this reason, guidelines produced in by European (ESC/EAS)100 and

American (ACC/AHA)101 associations do not focus on such classifications though they do

10

Page 11: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

often still retain the names proposed by Frederickson, instead make recommendations for

testing and treatment if lipoprotein parameters and other clinical features are suggestive of

an inherited, familial dyslipidaemia.

Although familial hypercholesterolaemia (FH) is a monogenic disorder and most strongly

correlates with cardiovascular disease, most inherited lipid disorders are polygenic102.

Individuals with FH can have the homozygous or heterozygous form of the disease, which

correlates with both severity of the LDL abnormality and the risk of cardiovascular

disease103,104.

[H2] Dyslipidaemia in cardiovascular disease

[H3] Hypercholesterolaemia. The association between high LDL cholesterol levels in blood

and the development and progression of cardiovascular disease has been known for >100

years105,106. Discovery of the LDL receptor and subsequent development and success of LDL-

lowering drugs, such as statins, have shown a convincing link between LDL-level reduction

and reductions in cardiovascular morbidity and mortality107-111. Beyond the extensively used

statin therapy, which lowers blood cholesterol levels through inhibition of HMG-CoA

reductase, a key and rate-limiting step in its synthesis in the liver. Newer, alternative LDL-

lowering agents, such as ezetimibe (which acts by inhibition of Niemann-Pick C1-like

1 (NPC1L1) protein resulting in reduced absorption of cholesterol in the small intestine, have

also successfully demonstrated in clinical trials improvements in cardiovascular outcome

following heart attack112. These findings suggest that the reduction in the risk of

cardiovascular disease is not unique to statins and probably extends to alternative LDL-

lowering strategies. For example, a potential alternative strategy is to target oxidized forms

of LDL (oxLDL), which have been shown to induce a proinflammatory response in monocytes

in vitro113-115, and whose circulating levels are elevated in cardiovascular disease116-119.

[H3] HDL as a therapeutic strategy. Beyond LDL-levels, interest in HDL cholesterol levels as

a potential therapeutic target has been explored, mainly owing to the role of HDL in reverse

cholesterol transport (RCT), antioxidative and anti-inflammatory properties 120. HDL

promotes cellular cholesterol efflux by interaction with the ATP-binding cassette

transporters ABCA1 and ABCG1121-124. The ratio of total cholesterol to HDL level is used as a

11

Page 12: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

predictive tool in assessing the risk of cardiovascular disease125. The consistent inverse

relationship between HDL levels and the risk of cardiovascular disease126 prompted

optimism into research aimed at elevating blood HDL levels. However, clinical trials on

therapies aimed to raise HDL levels for reducing the risk of cardiovascular disease have been

largely disappointing 127-132. These negative findings are supported by a Mendelian

randomization trial showing that individuals with a single nucleotide polymorphism (SNPs)

in LIPG that results in elevated HDL levels in blood (approximately a 12% increase) was not

associated with a reduction in the risk of MI 133. Furthermore, other SNPs associated with

lifetime elevations of HDL also did not associate with a reduction in the risk of MI 133.

However, evidence now shows that static measurements of HDL alone are not necessarily

an accurate assessment of the functional role of HDL particles. For instance, HDL particle

size134 and efflux capacity135 might be better predictors for assessing the role of HDL in

altering the risk of cardiovascular disease.

[H3] Hypertriglyceridaemia. Despite some early interest in the role of dietary triglycerides

in the risk of cardiovascular disease136-141, epidemiological data do not support a substantial

link between the levels of triglycerides in blood and the risk of cardiovascular disease 142.

Moreover, pharmacological methods for reducing triglyceride blood levels with the aim of

decreasing the risk of cardiovascular disease in addition to the risk reductions achieved with

statins have been unsuccessful so far143. Nevertheless, lowering triglyceride levels as a

therapeutic strategy is experiencing renewed interest on the basis of findings from

Mendelian randomization analyses144-146, genome-wide association studies147,148 and studies

on SNPs149 suggesting that the residual risk of cardiovascular disease persists despite

achievement of LDL target levels is related to elevated triglyceride levels, rather than low

HDL levels.

[H2] Effect of dyslipidaemia on monocytes

A number of studies have examined the changes in the phenotype of monocytes in response

to lipids, predominantly in the context of atherosclerosis models65,66,150-157. Although

mechanisms associated with atherogenesis remain the focus of much work, the effect of

12

Page 13: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

dyslipidaemia (that is, variance in particular lipid species) on monocyte phenotype and

function might be more complex. Possible effects of dyslipidaemia on immune function

include human data suggesting an apparent immunoparesis in surgical patients with very

high or low total cholesterol levels are at an increased risk of infection 158. Multiple

experimental data show poor responses of both innate and adaptive immunity to fungal and

bacterial infection in transgenic mice with dyslipidaemia159-161. Indeed, leukocyte interaction

with lipids in hyperlipidaemic environments has been documented for >50 years, when

lipid-filled rat monocytes in response to a HFD were denoted as ‘lipophages’ 162, and

monocyte invasion of the vascular endothelium in HFD-reared pigs was imaged by

transmission and scanning electron microscopy in 1979163 and 1981164, respectively. Some

studies in zebra fish have also shown monocytes can uptake lipids in circulation in a TLR4-

dependent process165.

[H3] Effects of cholesterol. The presence of cholesterol within the atherosclerotic plaque

and the identification of monocyte extravasation into the subendothelial space has focussed

the trajectory of much monocyte research in this pathophysiological context. Elevation in

blood cholesterol, predominantly owing to raised LDL levels, is the most commonly studied

dyslipidaemic condition. As monocytes and lipoprotein particles both circulate in the blood,

monocytes are likely to have physical encounters with cholesterol well before monocytes

migrate into the atherosclerotic lesion and differentiate into tissue macrophages. In most of

the literature in this field, the term ‘monocyte’ is often used interchangeably with

(monocyte-derived) ‘macrophages’, and experimental models are often in vitro scenarios

and/or cell lines that are used to recapitulate potential pathophysiological conditions.

Additionally, these experiments frequently involve transgenic animal models where

cholesterol levels are dramatically elevated to obtain results within an acceptable

timeframe. Therefore, data from experimental studies should be interpreted with some

caution when translating into the human clinical context.

Monocytes undergo morphological, phenotypical, and functional changes in

hypercholesterolaemic environments. Work in patients with familial hypercholesterolaemia

demonstrated intracytoplasmic vacuolation in monocytes and suggested the presence of

monocytosis in these patients166. CD14highCD16low (classical) monocytes from patients with

13

Page 14: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

familial hypercholesterolaemia preferentially take up oxLDL167.  Exposure of these

monocytes to oxLDL induces an increase in the expression of CD68 (also known as

macrosialin), a macrophage marker and CD11c (also known as integrin αX), an activation

marker, and an improved ability for phagocytosis167. Human monocytes have been shown to

uptake LDL in vitro, but do so more readily with oxLDL via scavenger receptors, including

SRA1, SRA2, SRB1 , CD36, and lectin-like oxidized LDL receptor 1 (LOX1) 168,169 (FIG. 2).

Transgenic animal models of elevated total cholesterol show evidence of neutral lipid

droplets in monocytes150 and, although the most probable explanation for this finding is lipid

uptake, the mechanisms remain unclear, and de novo lipogenesis (described in monocyte-

derived macrophage-like cells170) remains a possibility.

Atherosclerosis has long been considered an inflammatory condition171 and the

identification of cholesterol crystals not just in mature atherosclerotic plaques, but also in

early atherosclerotic plaques172,173, adds weight to the notion that cholesterol might drive

inflammatory atherogenesis (FIG. 2). Cholesterol crystals can induce the release of IL-1

through activation of the NOD-, LRR- and pyrin domain-containing 3 (NLRP3) inflammasome

in pre-primed mononuclear cells in vitro172. Activation of the NLRP3 inflammasome and

release of active IL-1 requires two signals. The first (priming) signal is required for the

upregulation of NLRP3 and pro-IL-1β, and can be triggered for example by TLR activation.

The second signal induces the activation of the inflammasome and caspase 1 to cleave pro-

IL-1. Minimally modified LDL (mmLDL), which provides a chemotactic stimulus to

monocytes174,175 , has also been shown to provide a priming signal for mononuclear cells in

vitro172. Interestingly, the dietary saturated fatty acid palmitic acid induced TLR2-mediated

IL-1 release in a human monocytic cell line in vitro, a process that was abrogated by use of

the polyunsaturated fatty acid docosahexaenoic acid176. Similarly, TLR4 activation induced by

oleic acid and palmitic acid has been reported in macrophage cell lines177. TLR4178,179 and its

downstream signalling adaptors TIR domain-containing adaptor molecule 1 (TICAM-1, also

known as TRIF) and TIR domain-containing adaptor molecule 2 (TICAM-2, also known as

TRAM)180 have been implicated in atherosclerosis. TLR4 stimulation will, on the most part,

result in activation of nuclear factor (NF)-κB, and NF-κB activation can either provide

proinflammatory atherosclerotic stimuli or be atheroprotective depending on the initial

route of activation181,182. A report published in 2016 suggests an alternative pathway of

14

Page 15: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

NLRP3 inflammasome activation in human monocytes that does not require a second signal

when stimulated through lipopolysaccharide (LPS)–TLR4 signalling183. However,

atherosclerosis might occur independently of NLRP3 signalling184, supporting the notion that

inflammasome activation is not ubiquitous in the plaque. OxLDL is thought to induce

activation of TLR2, TLR4, and TLR6 mediated by CD36 signalling185, which in turn can activate

macrophages186. OxLDL can independently promote the differentiation of human monocytes

into macrophage-like cells in vitro187-189. Moreover, oxLDL might induce monocyte

reprogramming or ‘innate immune memory’ that might alter secondary ligand responses115.

Some data suggest that cholesterol not always represents an inflammatory stimulus.

Intermediates of the cholesterol biosynthetic pathway, such as desmosterol, provide an

anti-inflammatory stimulus via LXR signalling in elicited peritoneal macrophages from Ldlr–/–

mice190 (FIG. 2b). Whether these cholesterol intermediates have anti-inflammatory potential

in monocytes is unknown. Of note, other fatty acids, such as palmitoleic acid, that are

present in neutral lipid-droplets of foamy monocytes have also been shown to have anti-

inflammatory functions191. The cholesterol efflux transporter ABCA1 might also have an anti-

inflammatory role by modulating TLR signalling. In murine macrophages, ABCA1 reduces TLR

trafficking to lipid rafts by reducing lipid raft cholesterol content192. This finding also

supports an anti-inflammatory role of HDL cholesterol. Furthermore, cell cholesterol

removal by cholesterol efflux pathways and administration of reconstituted HDL have been

shown to have direct anti-inflammatory effects on leukocytes120,193,194, and in the production

of classical monocytes by acting on HSPCs79. A study published in 2016 showed the specific

immunomodulatory effects of apolipoprotein A1 (the main HDL apolipoprotein) on

monocyte behaviour195. Acute exposure to apolipoprotein A1 inhibited macrophage

chemotaxis in vitro and monocyte recruitment in vivo195, consistent with previous findings120.

The different aspects of cholesterol, inflammation, and immune interaction have been

reviewed previously196.

[H3] Effects of triglyceride-rich lipoproteins. For the purposes of this section, TGRL is

defined as lipoprotein particles predominantly composed of chylomicrons, chylomicron

remnants, and very low-density lipoprotein (VLDL). Most of the research in blood lipid

abnormalities has focused on the effects of LDL and monocyte/macrophage responses.

15

Page 16: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

However, some studies have addressed the effects of TGRL on blood monocytes.

Fluctuations of chylomicron levels and, to a lesser degree, VLDL levels occur after ingesting

lipid-rich or carbohydrate-rich meals197,198 . Importantly, people, especially those in Western

countries, spend much of their day in the postprandial state owing to increased snacking

associated with obesity 199. Allied to this concept, the role of nonfasting triglyceride-levels as

a risk predictor for cardiovascular disease has been shown to be of value, and is endorsed by

the European Atherosclerosis Society (EAS) for clinical assessment200. Furthermore, beyond

their potential role in atherosclerosis, elevated lipid levels are found in individuals with

HIV201 and in conditions characterized by chronic inflammation, such as systemic lupus

erythematosus 202, implying potential alternative interactions between dysregulated

immune cells in these conditions and the presence of elevated blood lipid levels.

Ldlr–/– mice are frequently used to study the effects of dyslipidaemia on innate immunity.

These mice unsurprisingly have elevated LDL levels; however, these mice also have

markedly elevated levels of the chylomicron and VLDL subfractions (which are rich in both

apolipoprotein B-100 and apolipoprotein B-48), suggesting elevations in TGRL203. VLDL is

composed predominantly of triglycerides (70%), but has a small proportion of cholesterol

content (10%)204, some of which can be influenced by dietary intake197. A study published

in 2016 demonstrated the appearance of neutral lipid droplets in monocytes (both subsets)

from Ldlr–/– mice maintained on a HFD compared with monocytes from mice fed a control

chow diet, with evidence of impaired monocyte migration and retention in extravascular

tissue in response to inflammation65. This phenotype of tissue-retained monocytes after lipid

loading might be related to the plaque macrophage retention seen during HFD-induced

atherosclerosis models205,206. Indeed, lipid loading is also seen in monocytes from

hypercholesterolaemic Apoe–/– mice fed a HFD87,157. These lipid-loaded monocytes show an

activated phenotype with increased expression of CD11c in lipid rafts in the cellular

membrane157. These findings are supported by studies in human monocytes — in particular

the classical CD14highCD16low subset — showing an increase in membrane cholesterol content

in response to LDL, but more so following incubation with VLDL in vitro65, echoing the

concept that dyslipidaemic conditions can alter monocyte biology. Whether lipid loading of

monocytes during HFD leads to an exclusive proinflammatory phenotype is a matter of

debate. Nevertheless, the evidence indicates that lipids do alter monocyte behaviour.

16

Page 17: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Indeed, lipoproteins (particularly TGRL) are thought to inhibit monocyte response to

endotoxins by scavenging LPS207.

Similar to mouse monocytes in vivo, human monocytes of both subsets accumulate neutral

lipid in vitro in response to incubation with VLDL (and peculiarly LDL)65,151,152,154,156. The

accumulation of neutral lipid is associated with an impairment in chemotaxis and

chemokinesis in in vitro assays, potentially explained by alterations in cytoskeletal

rearrangement modulated by the small GTPase RAS homologue gene family member A

(RHOA)65 (FIG. 2b) . However in mouse models, in vivo monocyte lipid accumulation did not

result in altered patrolling behaviour65, which suggests that defects in migration after lipid

loading could be restricted to the extravascular tissue compartment. In vivo work in mouse

models where chronic TGRL elevation is induced pharmacologically support a more complex

role for nonclassical LY6Clow monocytes. Administration of poloxamer-407 elevates

circulating triglyceride levels in wild type mice by inhibiting lipoprotein lipase (LPL) — which

catalyses the conversion of VLDL to intermediate-density lipoprotein (IDL) — and cholesterol

7α-hydoxylase (CYP7A1) — involved in the transformation of cholesterol into bile acids —

without causing apparent hypercholesterolaemia66,208. This model of hypertriglyceridaemia

results in a noticeable reduction in peripheral counts of nonclassical LY6Clow monocytes

owing to the increase in their crawling behaviour and endothelial retention (without

alteration of bone marrow cMoP, total monocyte, or subset numbers)66. This increase in

crawling and retention led to mass monocyte extravasation into all organs examined (heart,

liver, and kidney) and accumulation of F4/80+CD68+ macrophages66. The exact mechanisms

are unknown, but did not produce an overt inflammatory response, although the model did

involve the overexpression in tissue macrophages of the chemokine CCL4 and a G protein-

coupled receptor-dependent mechanism66. Fascinatingly, in this model of poloxamer-407-

induced hypertriglyceridaemia monocytes did not have intracellular lipid accumulation,

which might indicate a role for LPL in lipid uptake or lipophage formation. Indeed, lipid

droplet incorporation into monocytes might be mediated by LPL. LPL generates free,

nonesterified fatty acid from TGRL that might promote an inflammatory phenotype, as

shown in some studies152,209.

Taken together, these findings indicate that cholesterol and TGRL can influence monocyte

subset effector functions (FIG. 2b). Further work is required to understand the potential

17

Page 18: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

effect of dyslipidaemic environments on immune functions and cardiovascular disease.

Speculating that monocytes can deliver a lipid load during atherogenesis, rather than the

restricted theory of only tissue macrophages responding to retaining modified cholesterols,

would be intriguing. The contribution of monocytes as effective lipid carriers, and indeed

the role of monocytes in systemic lipid metabolism, warrants further study.

[H2] Effect of dietary lipids on monocytes

Contributions of dietary lipids to cardiovascular disease remain enigmatic, and any potential

role of dietary lipids in modulating monocyte and macrophage biology has not been studied

in detail. Dietary studies are inherently difficult to perform, as the majority require

substitution of one diet component while maintaining similar calorific content. Analyses in

dietary studies usually involve an element of observation, and are reliant on questionnaires

and participant reporting; therefore, these studies are subject to numerous sources of bias.

Despite these limitations, long-term and short-term dietary studies have helped to inform

us of potential deleterious effects of dietary lipids. The effects of so-called ‘healthy eating’

on disease continues to be of interest, particularly its role in cardiovascular disease210. Diets

rich in saturated fatty acids have been implicated in cardiovascular disease211-219, whereas

monounsaturated fatty acids (MUFA)217,220,221 and polyunsaturated fatty acids (PUFA) 222-224

are suggested as cardioprotective. Whether dietary saturated fat is indeed associated with

cardiovascular disease remains controversial, particularly in light of updated guidelines

continuing to support a diet low in saturated fat225, even after several retrospective meta-

analyses have found no link between the incidence of cardiovascular disease and saturated

fat intake224,226,227. However, these studies support the notion that diets rich in trans-fat are

deleterious to health and that MUFA-rich diets and omega-3 PUFA-rich diets are

beneficial224,226,227. Although these data help to establish epidemiological trends, these

studies do not help to clarify the mechanisms by which dietary lipid might influence the risk

of cardiovascular disease. Short-term and acute feeding studies have been used to elucidate

these mechanisms. Diets rich in saturated fat can activate proinflammatory pathways in

peripheral blood mononuclear cells228 and in monocytes152,209,229, whereas MUFA-rich diets

seem to induce anti-inflammatory signalling pathways228. Possible mechanisms through

which saturated fat might drive an inflammatory response are varied; for instance,

18

Page 19: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

saturated fat might promote trafficking of gut endotoxin into the systemic circulation229-232,

and although early in vitro data177,233-236 and limited evidence in vivo237 suggested that the

mechanism might be mediated through direct stimulation of TLR by saturated fat , other

studies indicate that this mechanism is unlikely and show that the inflammatory response is

a result of endotoxinaemia238. Immune cells are altered within hours of meal consumption. A

meal rich in saturated fat can activate monocytes, as assessed by increased expression of

CD11c, accumulation of cytosolic intracellular lipid analysed ex vivo, and increased

adherence to vascular cell adhesion molecule 1 (VCAM1) substrate in a shear stress

model152. In flow cytometry analyses, human leukocytes239 and monocytes152 isolated from

blood in the postprandial phase show an increase in side scatter profile, implying an

increase in granularity resulting from an increase in lipid internalization. This increase in side

scatter is also found in monocytes from Apoe–/– mouse raised on a chronic HFD87,157.

Therefore, similar to dyslipidaemic environments, the postprandial response of consuming a

diet high in saturated fat might lead to formation of lipophages or ‘foamy’

monocytes152,157,240. A study published in 2016 showed that the postprandial response

mediates monocyte activation in individuals with metabolic syndrome156. The detailed

mechanisms of when and how monocyte subsets respond to dietary lipid loading remain to

be determined.

[H1] Conclusions

In this Review, we have briefly discussed the role of fetal macrophages and embryonic

monocytes in maintaining the tissue-resident macrophage pool (FIG. 1). The role of these

fetal tissue-resident macrophages in cardiovascular disease and response to dyslipidaemia

remains to be fully explored. We have elaborated on the heterogeneity of bone marrow

derived mouse and human monocytes and some essential differences in subset effector

functions during steady state and dyslipidaemia (FIG. 1, 2). The modulation of effector

functions of blood monocytes during dyslipidaemia might increase the risk of

atherosclerosis and accelerate the progression of the disease. In summary, dyslipidaemia —

with differing effects of hypercholesterolaemia and hypertriglyceridaemia — can alter bone

marrow production of monocyte precursors and mature monocyte subsets (particularly

classical monocytes); promotes the lipid endocytosis and accumulation of neutral lipid

19

Page 20: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

droplets in monocyte subsets; induces an increased endothelial accumulation and

extravasation of nonclassical monocytes; increases tissue retention of monocytes and

accumulation of tissue-resident macrophages; and promotes a proinflammatory phenotype

and modulates chemotaxis in monocytes (FIG. 2b). Together, these responses could

aggravate tissue damage during atherogenesis and, we hypothesize, this increased tissue

damage might increase the risk of plaque rupture. This hypothesis requires more

investigation, but does raise the possibility that lipid-lowering therapies can modulate

monocyte behaviour towards a response that might help to reduce cardiovascular events.

References

1. Ginhoux, F. & Jung, S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat. Rev. Immunol. 14, 392–404 (2014).

2. Robbins, C. S. et al. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat. Med. 19, 1166–1172 (2013).

3. Cros, J. et al. Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors. Immunity 33, 375–386 (2010).

4. van Furth, R. & Cohn, Z. A. The origin and kinetics of mononuclear phagocytes. J Exp Med 128, 415–435 (1968).

5. Shi, C. & Pamer, E. G. Monocyte recruitment during infection and inflammation. Nat. Rev. Immunol. 11, 762–774 (2011).

6. Swirski, F. K. et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science 325, 612–616 (2009).

7. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38, 79–91 (2013).

8. Takahashi, K., Yamamura, F. & Naito, M. Differentiation, maturation, and proliferation of macrophages in the mouse yolk sac: a light-microscopic, enzyme-cytochemical, immunohistochemical, and ultrastructural study. J. Leukoc. Biol. 45, 87–96 (1989).

9. Takahashi, K. & Naito, M. Development, differentiation, and proliferation of macrophages in the rat yolk sac. Tissue Cell 25, 351–362 (1993).

10. Palis, J. et al. Spatial and temporal emergence of high proliferative potential hematopoietic precursors during murine embryogenesis. Proc. Natl. Acad. Sci. U.S.A. 98, 4528–4533 (2001).

11. McGrath, K. E., Koniski, A. D., Malik, J. & Palis, J. Circulation is established in a stepwise pattern in the mammalian embryo. Blood 101, 1669–1676 (2003).

12. Orkin, S. H. & Zon, L. I. Hematopoiesis: an evolving paradigm for stem cell biology. Cell 132, 631–644 (2008).

13. Guilliams, M. et al. Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J Exp Med 210, 1977–1992 (2013).

14. Ginhoux, F. et al. Fate mapping analysis reveals that adult microglia derive from

20

Page 21: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

primitive macrophages. Science 330, 841–845 (2010).15. Schulz, C. et al. A lineage of myeloid cells independent of Myb and hematopoietic

stem cells. Science 336, 86–90 (2012).16. Epelman, S. et al. Embryonic and adult-derived resident cardiac macrophages are

maintained through distinct mechanisms at steady state and during inflammation. Immunity 40, 91–104 (2014).

17. Hoeffel, G. et al. C-Myb(+) erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity 42, 665–678 (2015).

18. Hoeffel, G. et al. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J Exp Med 209, 1167–1181 (2012).

19. Jaensson, E. et al. Small intestinal CD103+ dendritic cells display unique functional properties that are conserved between mice and humans. J Exp Med 205, 2139–2149 (2008).

20. Bain, C. C. et al. Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice. Nature Immunology 15, 929–937 (2014).

21. Cecchini, M. G. et al. Role of colony stimulating factor-1 in the establishment and regulation of tissue macrophages during postnatal development of the mouse. Development 120, 1357–1372 (1994).

22. Dai, X.-M. et al. Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects. Blood 99, 111–120 (2002).

23. Wiktor-Jedrzejczak, W. & Gordon, S. Cytokine regulation of the macrophage (M phi) system studied using the colony stimulating factor-1-deficient op/op mouse. Physiological Reviews 76, 927–947 (1996).

24. Robbins, C. S. et al. Extramedullary Hematopoiesis Generates Ly-6Chigh Monocytes That Infiltrate Atherosclerotic LesionsClinical Perspective. Circulation 125, 364–374 (2012).

25. Fogg, D. K. et al. A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science 311, 83–87 (2006).

26. Sathe, P. et al. Lymphoid Tissue and Plasmacytoid Dendritic Cells and Macrophages Do Not Share a Common Macrophage-Dendritic Cell-Restricted Progenitor. 41, 104–115 (2014).

27. Hettinger, J. et al. Origin of monocytes and macrophages in a committed progenitor. Nature Immunology 14, 821–830 (2013).

28. Imhof, B. A. & Aurrand-Lions, M. Adhesion mechanisms regulating the migration of monocytes. Nat. Rev. Immunol. 4, 432–444 (2004).

29. Volkman, A. The origin and fate of the monocyte. Ser Haematol 3, 62–92 (1970).30. Vu Manh, T.-P. et al. Defining Mononuclear Phagocyte Subset Homology Across

Several Distant Warm-Blooded Vertebrates Through Comparative Transcriptomics. Front Immunol 6, 299 (2015).

31. Etzrodt, M. et al. Regulation of monocyte functional heterogeneity by miR-146a and Relb. Cell Reports 1, 317–324 (2012).

32. Ingersoll, M. A. et al. Comparison of gene expression profiles between human and mouse monocyte subsets. Blood 115, e10–e19 (2010).

21

Page 22: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

33. Mildner, A. et al. Mononuclear phagocyte miRNome analysis identifies miR-142 as critical regulator of murine dendritic cell homeostasis. Blood 121, 1016–1027 (2013).

34. Douglas, S. D. & Douglas, A. G. in Williams Manual of Hematology (eds. Kaushansky, K., Beutler, E., Seligsohn, U., Lichtman, M. A. & Prchal, J. T.) 1045–1073 (McGraw-Hill Professional, 2011).

35. Ziegler-Heitbrock, L. The CD14+ CD16+ blood monocytes: their role in infection and inflammation. J. Leukoc. Biol. 81, 584–592 (2007).

36. Ancuta, P. et al. Transcriptional profiling reveals developmental relationship and distinct biological functions of CD16+ and CD16- monocyte subsets. BMC Genomics 2009 10:1 10, 1 (2009).

37. Auffray, C. et al. Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science 317, 666–670 (2007).

38. Geissmann, F., Jung, S. & Littman, D. R. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 19, 71–82 (2003).

39. Sunderkötter, C. et al. Subpopulations of mouse blood monocytes differ in maturation stage and inflammatory response. The Journal of Immunology 172, 4410–4417 (2004).

40. Zhao, C. et al. Identification of novel functional differences in monocyte subsets using proteomic and transcriptomic methods. J. Proteome Res. 8, 4028–4038 (2009).

41. Yona, S. & Jung, S. Monocytes: subsets, origins, fates and functions. Current Opinion in Hematology 17, 53–59 (2010).

42. Ziegler-Heitbrock, L. Monocyte subsets in man and other species. Cell. Immunol. 289, 135–139 (2014).

43. Woollard, K. J. & Geissmann, F. Monocytes in atherosclerosis: subsets and functions. Nature Reviews Cardiology 1–10 (2010). doi:10.1038/nrcardio.2009.228

44. Randolph, G. J. Mechanisms that regulate macrophage burden in atherosclerosis. Circulation Research 114, 1757–1771 (2014).

45. Jung, S. et al. Analysis of fractalkine receptor CX(3)CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol. Cell. Biol. 20, 4106–4114 (2000).

46. Palframan, R. T. et al. Inflammatory Chemokine Transport and Presentation in HEV A Remote Control Mechanism for Monocyte Recruitment to Lymph Nodes in Inflamed Tissues. J Exp Med 194, 1361–1374 (2001).

47. Jakubzick, C. et al. Minimal differentiation of classical monocytes as they survey steady-state tissues and transport antigen to lymph nodes. Immunity 39, 599–610 (2013).

48. Moore, K. J., Sheedy, F. J. & Fisher, E. A. Macrophages in atherosclerosis: a dynamic balance. Nat. Rev. Immunol. 13, 709–721 (2013).

49. Wong, K. L. et al. Gene expression profiling reveals the defining features of the classical, intermediate, and nonclassical human monocyte subsets. Blood 118, e16–31 (2011).

50. Yang, J., Zhang, L., Yu, C., Yang, X.-F. & Wang, H. Monocyte and macrophage differentiation: circulation inflammatory monocyte as biomarker for inflammatory diseases. Biomarker Research 2014 2:1 2, 1 (2014).

51. Mukherjee, R. et al. Non-Classical monocytes display inflammatory features:

22

Page 23: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Validation in Sepsis and Systemic Lupus Erythematous. Nature Publishing Group 5, 13886 (2015).

52. Hilgendorf, I. et al. Ly-6Chigh monocytes depend on Nr4a1 to balance both inflammatory and reparative phases in the infarcted myocardium. Circulation Research 114, 1611–1622 (2014).

53. Varol, C. et al. Monocytes give rise to mucosal, but not splenic, conventional dendritic cells. J Exp Med 204, 171–180 (2007).

54. Hanna, R. N. et al. The transcription factor NR4A1 (Nur77) controls bone marrow differentiation and the survival of Ly6C− monocytes. Nature Immunology 12, 778–785 (2011).

55. Carlin, L. M. et al. Nr4a1-Dependent Ly6Clow Monocytes Monitor Endothelial Cells and Orchestrate Their Disposal. Cell 153, 362–375 (2013).

56. Shaked, I. et al. Transcription factor Nr4a1 couples sympathetic and inflammatory cues in CNS-recruited macrophages to limit neuroinflammation. Nature Immunology 16, 1228–1234 (2015).

57. Hanna, R. et al. NR4A1 (Nur77) dependent monocytes patrol the lung vasculature and inhibit tumor cell invasion (P5068). The Journal of Immunology 190, 180.17–180.17 (2013).

58. Hanna, R. N. et al. NR4A1 (Nur77) deletion polarizes macrophages toward an inflammatory phenotype and increases atherosclerosis. Circulation Research 110, 416–427 (2012).

59. Hamers, A. A. J. et al. Bone marrow-specific deficiency of nuclear receptor Nur77 enhances atherosclerosis. Circulation Research 110, 428–438 (2012).

60. Chao, L. C. et al. Bone marrow NR4A expression is not a dominant factor in the development of atherosclerosis or macrophage polarization in mice. J. Lipid Res. 54, 806–815 (2013).

61. Chong, S. Z. et al. CXCR4 identifies transitional bone marrow premonocytes that replenish the mature monocyte pool for peripheral responses. J Exp Med 213, 2293–2314 (2016).

62. Menezes, S. et al. The Heterogeneity of Ly6Chi Monocytes Controls Their Differentiation into iNOS+ Macrophages or Monocyte-Derived Dendritic Cells. Immunity 45, 1205–1218 (2016).

63. Satoh, T. et al. Identification of an atypical monocyte and committed progenitor involved in fibrosis. Nature 541, 96–101 (2017).

64. Ziegler-Heitbrock, L. & Hofer, T. P. J. Toward a refined definition of monocyte subsets. Front Immunol 4, 23 (2013).

65. Jackson, W. D., Weinrich, T. W. & Woollard, K. J. Very-low and low-density lipoproteins induce neutral lipid accumulation and impair migration in monocyte subsets. Nature Publishing Group 1–12 (2016). doi:10.1038/srep20038

66. Saja, M. F. et al. Triglyceride-Rich Lipoproteins Modulate the Distribution and Extravasation of Ly6C/Gr1low Monocytes. Cell Reports 12, 1802–1815 (2015).

67. Misharin, A. V. et al. Nonclassical Ly6C− Monocytes Drive the Development of Inflammatory Arthritis in Mice. Cell Reports 9, 591–604 (2014).

68. Sampson, M. J., Davies, I. R., Brown, J. C., Ivory, K. & Hughes, D. A. Monocyte and neutrophil adhesion molecule expression during acute hyperglycemia and after antioxidant treatment in type 2 diabetes and control patients. Arteriosclerosis, Thrombosis, and Vascular Biology 22, 1187–1193 (2002).

23

Page 24: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

69. Imhof, B. A. et al. CCN1/CYR61-mediated meticulous patrolling by Ly6Clow monocytes fuels vascular inflammation. Proceedings of the National Academy of Sciences 113, E4847–56 (2016).

70. Finsterbusch, M. et al. Patrolling monocytes promote intravascular neutrophil activation and glomerular injury in the acutely inflamed glomerulus. Proceedings of the National Academy of Sciences 113, E5172–81 (2016).

71. Michaud, J.-P., Bellavance, M.-A., Préfontaine, P. & Rivest, S. Real-time in vivo imaging reveals the ability of monocytes to clear vascular amyloid beta. Cell Reports 5, 646–653 (2013).

72. Hanna, R. N. et al. Patrolling monocytes control tumor metastasis to the lung. Science 350, 985–990 (2015).

73. Li, W. et al. Intravital 2-photon imaging of leukocyte trafficking in beating heart. J. Clin. Invest. 122, 2499–2508 (2012).

74. Murphy, A. J. et al. ApoE regulates hematopoietic stem cell proliferation, monocytosis, and monocyte accumulation in atherosclerotic lesions in mice. J. Clin. Invest. 121, 4138–4149 (2011).

75. Swirski, F. K. et al. Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J. Clin. Invest. 117, 195–205 (2007).

76. Tacke, F. et al. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J. Clin. Invest. 117, 185–194 (2007).

77. Tolani, S. et al. Hypercholesterolemia and reduced HDL-C promote hematopoietic stem cell proliferation and monocytosis: studies in mice and FH children. Atherosclerosis 229, 79–85 (2013).

78. Yvan-Charvet, L., Wang, N. & Tall, A. R. Role of HDL, ABCA1, and ABCG1 transporters in cholesterol efflux and immune responses. Arteriosclerosis, Thrombosis, and Vascular Biology 30, 139–143 (2010).

79. Yvan-Charvet, L. et al. ATP-binding cassette transporters and HDL suppress hematopoietic stem cell proliferation. Science 328, 1689–1693 (2010).

80. Chapman, C. M. L., Beilby, J. P., McQuillan, B. M., Thompson, P. L. & Hung, J. Monocyte count, but not C-reactive protein or interleukin-6, is an independent risk marker for subclinical carotid atherosclerosis. Stroke 35, 1619–1624 (2004).

81. Johnsen, S. H. et al. Monocyte count is a predictor of novel plaque formation: a 7-year follow-up study of 2610 persons without carotid plaque at baseline the Tromsø Study. Stroke 36, 715–719 (2005).

82. Ganda, A. et al. Mild renal dysfunction and metabolites tied to low HDL cholesterol are associated with monocytosis and atherosclerosis. Circulation 127, 988–996 (2013).

83. Combadière, C. et al. Combined inhibition of CCL2, CX3CR1, and CCR5 abrogates Ly6C(hi) and Ly6C(lo) monocytosis and almost abolishes atherosclerosis in hypercholesterolemic mice. Circulation 117, 1649–1657 (2008).

84. Swirski, F. K. et al. Monocyte accumulation in mouse atherogenesis is progressive and proportional to extent of disease. Proc. Natl. Acad. Sci. U.S.A. 103, 10340–10345 (2006).

85. Lessner, S. M., Prado, H. L., Waller, E. K. & Galis, Z. S. Atherosclerotic lesions grow through recruitment and proliferation of circulating monocytes in a murine model. Am. J. Pathol. 160, 2145–2155 (2002).

24

Page 25: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

86. Landsman, L. et al. CX3CR1 is required for monocyte homeostasis and atherogenesis by promoting cell survival. Blood 113, 963–972 (2009).

87. Potteaux, S. et al. Suppressed monocyte recruitment drives macrophage removal from atherosclerotic plaques of Apoe-/- mice during disease regression. J. Clin. Invest. 121, 2025–2036 (2011).

88. Murphy, A. J. & Tall, A. R. Proliferating macrophages populate established atherosclerotic lesions. Circulation Research 114, 236–238 (2014).

89. Psaltis, P. J. et al. Characterization of a resident population of adventitial macrophage progenitor cells in postnatal vasculature. Circulation Research 115, 364–375 (2014).

90. Ensan, S. et al. Self-renewing resident arterial macrophages arise from embryonic CX3CR1(+) precursors and circulating monocytes immediately after birth. Nature Immunology 17, 159–168 (2016).

91. Dutta, P. et al. Myocardial infarction accelerates atherosclerosis. Nature 487, 325–329 (2012).

92. Heidt, T. et al. Differential contribution of monocytes to heart macrophages in steady-state and after myocardial infarction. Circulation Research 115, 284–295 (2014).

93. Fodor, G. Primary prevention of CVD: treating dyslipidaemia. BMJ Clin Evid 2010, (2010).

94. Prospective Studies Collaboration et al. Blood cholesterol and vascular mortality by age, sex, and blood pressure: a meta-analysis of individual data from 61 prospective studies with 55,000 vascular deaths. Lancet 370, 1829–1839 (2007).

95. Karpe, F., Dickmann, J. R. & Frayn, K. N. Fatty acids, obesity, and insulin resistance: time for a reevaluation. Diabetes 60, 2441–2449 (2011).

96. Hansson, G. K. & Libby, P. The immune response in atherosclerosis: a double-edged sword. Nat. Rev. Immunol. 6, 508–519 (2006).

97. Freigang, S. et al. Fatty acid-induced mitochondrial uncoupling elicits inflammasome-independent IL-1α and sterile vascular inflammation in atherosclerosis. Nature Immunology 14, 1045–1053 (2013).

98. Fredrickson, D. S. & Lees, R. S. A System for Phenotyping Hyperlipoproteinemia. Circulation 31, 321–327 (1965).

99. Beaumont, J. L. et al. Classification of hyperlipidaemias and hyperlipoproteinaemias. Bull. World Health Organ. 43, 891–915 (1970).

100. Catapano, A. L. et al. 2016 ESC/EAS Guidelines for the Management of Dyslipidaemias. European Heart Journal 37, 2999–3058 (2016).

101. Stone, N. 2013 ACC/AHA Guideline on the Treatment of Blood Cholesterol to Reduce Atherosclerotic Cardiovascular Risk in Adults. 1–85 (2013). doi:10.1161/01.cir.0000437738.63853.7a/-/DC1

102. Global Lipids Genetics Consortium. Discovery and refinement of loci associated with lipid levels. Nat. Genet. 45, 1274–1283 (2013).

103. Brown, M. S. & Goldstein, J. L. Familial hypercholesterolemia: defective binding of lipoproteins to cultured fibroblasts associated with impaired regulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity. Proc. Natl. Acad. Sci. U.S.A. 71, 788–792 (1974).

104. Fredrickson, D. S. & Levy, R. I. Familial hyperlipoproteinemia. In the metabolic basis of inherited disease. (McGraw-Hill, 1978).

25

Page 26: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

105. Vogel, J., Day, G. E., Cantab, L. M. & Baillière, H. The Pathological Anatomy of the Human Body. (H. Bailliere, Publisher, 1847).

106. Anitschkow, N. Die pathologischen Veränderungen innerer Organe bei experimenteller Cholesterinesterverfettung 1). DMW - Deutsche Medizinische Wochenschrift 39, 741–743 (2009).

107. Levy, R. I. et al. Cholestyramine in type II hyperlipoproteinemia. A double-blind trial. Ann. Intern. Med. 79, 51–58 (1973).

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

109. Shepherd, J. et al. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. West of Scotland Coronary Prevention Study Group. N. Engl. J. Med. 333, 1301–1307 (1995).

110. Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 360, 7–22 (2002).

111. Cannon, C. P. et al. Intensive versus moderate lipid lowering with statins after acute coronary syndromes. N. Engl. J. Med. 350, 1495–1504 (2004).

112. Cannon, C. P. et al. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N. Engl. J. Med. 372, 2387–2397 (2015).

113. Jovinge, S., Ares, M. P., Kallin, B. & Nilsson, J. Human monocytes/macrophages release TNF-alpha in response to Ox-LDL. Arteriosclerosis, Thrombosis, and Vascular Biology 16, 1573–1579 (1996).

114. Terkeltaub, R. et al. Oxidized LDL induces monocytic cell expression of interleukin-8, a chemokine with T-lymphocyte chemotactic activity. Arterioscler. Thromb. 14, 47–53 (1994).

115. Bekkering, S. et al. Oxidized low-density lipoprotein induces long-term proinflammatory cytokine production and foam cell formation via epigenetic reprogramming of monocytes. Arteriosclerosis, Thrombosis, and Vascular Biology 34, 1731–1738 (2014).

116. Berliner, J. A. & Heinecke, J. W. The role of oxidized lipoproteins in atherogenesis. Free Radical Biology and Medicine 20, 707–727 (1996).

117. Berliner, J. A. et al. Oxidized lipids in atherogenesis: formation, destruction and action. Thromb. Haemost. 78, 195–199 (1997).

118. Holvoet, P. et al. Circulating oxidized LDL is a useful marker for identifying patients with coronary artery disease. Arteriosclerosis, Thrombosis, and Vascular Biology 21, 844–848 (2001).

119. Koenig, W. et al. Oxidized LDL and the risk of coronary heart disease: results from the MONICA/KORA Augsburg Study. Clin. Chem. 57, 1196–1200 (2011).

120. Murphy, A. J. et al. High-density lipoprotein reduces the human monocyte inflammatory response. Arteriosclerosis, Thrombosis, and Vascular Biology 28, 2071–2077 (2008).

121. Barter, P. J. et al. Antiinflammatory properties of HDL. Circulation Research 95, 764–772 (2004).

122. Wang, N., Ranalletta, M., Matsuura, F., Peng, F. & Tall, A. R. LXR-Induced Redistribution of ABCG1 to Plasma Membrane in Macrophages Enhances Cholesterol Mass Efflux to HDL. Arteriosclerosis, Thrombosis, and Vascular Biology

26

Page 27: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

26, 1310–1316 (2006).123. Du, X.-M. et al. HDL particle size is a critical determinant of ABCA1-mediated

macrophage cellular cholesterol export. Circulation Research 116, 1133–1142 (2015).

124. Navab, M., Reddy, S. T., Van Lenten, B. J. & Fogelman, A. M. HDL and cardiovascular disease: atherogenic and atheroprotective mechanisms. Nature Reviews Cardiology 8, 222–232 (2011).

125. D'Agostino, R. B. et al. General cardiovascular risk profile for use in primary care: the Framingham Heart Study. Circulation 117, 743–753 (2008).

126. Wilson, P. W., Abbott, R. D. & Castelli, W. P. High density lipoprotein cholesterol and mortality. The Framingham Heart Study. Arteriosclerosis 8, 737–741 (1988).

127. AIM-HIGH Investigators et al. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N. Engl. J. Med. 365, 2255–2267 (2011).

128. HPS2-THRIVE Collaborative Group et al. Effects of extended-release niacin with laropiprant in high-risk patients. N. Engl. J. Med. 371, 203–212 (2014).

129. Barter, P. J. et al. Effects of torcetrapib in patients at high risk for coronary events. N. Engl. J. Med. 357, 2109–2122 (2007).

130. Schwartz, G. G. et al. Effects of dalcetrapib in patients with a recent acute coronary syndrome. N. Engl. J. Med. 367, 2089–2099 (2012).

131. Nicholls, S. J. et al. Efficacy and Safety of a Novel Oral Inducer of Apolipoprotein A-I Synthesis in Statin-Treated Patients With Stable Coronary Artery Disease. J. Am. Coll. Cardiol. 57, 1111–1119 (2011).

132. Nicholls, S. J. et al. ApoA-I Induction as a Potential Cardioprotective Strategy: Rationale for the SUSTAIN and ASSURE Studies. Cardiovasc Drugs Ther 26, 181–187 (2012).

133. Voight, B. F. et al. Plasma HDL cholesterol and risk of myocardial infarction: a mendelian randomisation study. Lancet 380, 572–580 (2012).

134. Mackey, R. H. et al. High-density lipoprotein cholesterol and particle concentrations, carotid atherosclerosis, and coronary events: MESA (multi-ethnic study of atherosclerosis). J. Am. Coll. Cardiol. 60, 508–516 (2012).

135. Khera, A. V. et al. Cholesterol efflux capacity, high-density lipoprotein function, and atherosclerosis. N. Engl. J. Med. 364, 127–135 (2011).

136. Zilversmit, D. B. Atherogenesis: a postprandial phenomenon. Circulation 60, 473–485 (1979).

137. Proctor, S. D. & Mamo, J. C. Retention of fluorescent-labelled chylomicron remnants within the intima of the arterial wall--evidence that plaque cholesterol may be derived from post-prandial lipoproteins. Eur. J. Clin. Invest. 28, 497–503 (1998).

138. Ginsberg, H. N. et al. Association of postprandial triglyceride and retinyl palmitate responses with newly diagnosed exercise-induced myocardial ischemia in middle-aged men and women. Arteriosclerosis, Thrombosis, and Vascular Biology 15, 1829–1838 (1995).

139. Sharrett, A. R., Chambless, L. E., Heiss, G., Paton, C. C. & Patsch, W. Association of postprandial triglyceride and retinyl palmitate responses with asymptomatic carotid artery atherosclerosis in middle-aged men and women. The Atherosclerosis Risk in Communities (ARIC) Study. Arteriosclerosis, Thrombosis, and Vascular Biology 15, 2122–2129 (1995).

140. Weintraub, M. S. et al. Clearance of chylomicron remnants in normolipidaemic

27

Page 28: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

patients with coronary artery disease: case control study over three years. BMJ 312, 935–939 (1996).

141. Karpe, F., Steiner, G., Uffelman, K., Olivecrona, T. & Hamsten, A. Postprandial lipoproteins and progression of coronary atherosclerosis. Atherosclerosis 106, 83–97 (1994).

142. Sarwar, N. et al. Triglycerides and the risk of coronary heart disease: 10,158 incident cases among 262,525 participants in 29 Western prospective studies. Circulation 115, 450–458 (2007).

143. Shapiro, M. D. & Fazio, S. From Lipids to Inflammation New Approaches to Reducing Atherosclerotic Risk. Circulation Research 118, 732–749 (2016).

144. Thomsen, M., Varbo, A., Tybjærg-Hansen, A. & Nordestgaard, B. G. Low nonfasting triglycerides and reduced all-cause mortality: a mendelian randomization study. Clin. Chem. 60, 737–746 (2014).

145. Jørgensen, A. B. et al. Genetically elevated non-fasting triglycerides and calculated remnant cholesterol as causal risk factors for myocardial infarction. European Heart Journal 34, 1826–1833 (2013).

146. Varbo, A., Benn, M., Tybjærg-Hansen, A. & Nordestgaard, B. G. Elevated remnant cholesterol causes both low-grade inflammation and ischemic heart disease, whereas elevated low-density lipoprotein cholesterol causes ischemic heart disease without inflammation. Circulation 128, 1298–1309 (2013).

147. Johansen, C. T. et al. Excess of rare variants in genes identified by genome-wide association study of hypertriglyceridemia. Nat. Genet. 42, 684–687 (2010).

148. Do, R. et al. Common variants associated with plasma triglycerides and risk for coronary artery disease. Nat. Genet. 45, 1345–1352 (2013).

149. Wittrup, H. H., Tybjaerg-Hansen, A. & Nordestgaard, B. G. Lipoprotein lipase mutations, plasma lipids and lipoproteins, and risk of ischemic heart disease. A meta-analysis. Circulation 99, 2901–2907 (1999).

150. Wu, H. et al. Functional role of CD11c+ monocytes in atherogenesis associated with hypercholesterolemia. Circulation 119, 2708–2717 (2009).

151. Hartigh, den, L. J., Connolly-Rohrbach, J. E., Fore, S., Huser, T. R. & Rutledge, J. C. Fatty acids from very low-density lipoprotein lipolysis products induce lipid droplet accumulation in human monocytes. J. Immunol. 184, 3927–3936 (2010).

152. Gower, R. M. et al. CD11c/CD18 expression is upregulated on blood monocytes during hypertriglyceridemia and enhances adhesion to vascular cell adhesion molecule-1. Arteriosclerosis, Thrombosis, and Vascular Biology 31, 160–166 (2011).

153. Guijas, C. et al. Simultaneous activation of p38 and JNK by arachidonic acid stimulates the cytosolic phospholipase A2-dependent synthesis of lipid droplets in human monocytes. J. Lipid Res. 53, 2343–2354 (2012).

154. Foster, G. A., Gower, R. M. & Stanhope, K. L. On-chip phenotypic analysis of inflammatory monocytes in atherogenesis and myocardial infarction. in (2013). doi:10.1073/pnas.1300651110/-/DCSupplemental

155. Xu, L. et al. Foamy monocytes form early and contribute to nascent atherosclerosis in mice with hypercholesterolemia. Arteriosclerosis, Thrombosis, and Vascular Biology 35, 1787–1797 (2015).

156. Khan, I. M. et al. Postprandial monocyte activation in individuals with metabolic syndrome. J. Clin. Endocrinol. Metab. jc20162732 (2016). doi:10.1210/jc.2016-2732

157. Foster, G. A. et al. CD11c/CD18 Signals Very Late Antigen-4 Activation To Initiate

28

Page 29: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Foamy Monocyte Recruitment during the Onset of Hypercholesterolemia. J. Immunol. 1501077 (2015). doi:10.4049/jimmunol.1501077

158. Delgado-Rodríguez, M., Medina-Cuadros, M., Martínez-Gallego, G. & Sillero-Arenas, M. Total cholesterol, HDL-cholesterol, and risk of nosocomial infection: a prospective study in surgical patients. Infect Control Hosp Epidemiol 18, 9–18 (1997).

159. Netea, M. G. et al. Hyperlipoproteinemia enhances susceptibility to acute disseminated Candida albicans infection in low-density-lipoprotein-receptor-deficient mice. Infect. Immun. 65, 2663–2667 (1997).

160. de Bont, N. et al. Apolipoprotein E knock-out mice are highly susceptible to endotoxemia and Klebsiella pneumoniae infection. J. Lipid Res. 40, 680–685 (1999).

161. Roselaar, S. E. & Daugherty, A. Apolipoprotein E-deficient mice have impaired innate immune responses to Listeria monocytogenes in vivo. J. Lipid Res. 39, 1740–1743 (1998).

162. Suzuki, M. & O'Neal, R. M. Accumulation of lipids in the leukocytes of rats fed atherogenic diets. J. Lipid Res. 5, 624–627 (1964).

163. Gerrity, R. G., Naito, H. K., Richardson, M. & Schwartz, C. J. Dietary induced atherogenesis in swine. Morphology of the intima in prelesion stages. Am. J. Pathol. 95, 775–792 (1979).

164. Gerrity, R. G. The role of the monocyte in atherogenesis: I. Transition of blood-borne monocytes into foam cells in fatty lesions. Am. J. Pathol. 103, 181–190 (1981).

165. Choi, S.-H. et al. Lipoprotein accumulation in macrophages via toll-like receptor-4-dependent fluid phase uptake. Circulation Research 104, 1355–1363 (2009).

166. Dresel, H. A. et al. Observations on leukocytes from patients with severe familial hypercholesterolemia. Arteriosclerosis 6, 259–264 (1986).

167. Mosig, S. et al. Different functions of monocyte subsets in familial hypercholesterolemia: potential function of CD14+ CD16+ monocytes in detoxification of oxidized LDL. FASEB J. 23, 866–874 (2009).

168. Steinberg, D. The LDL modification hypothesis of atherogenesis: an update. J. Lipid Res. 50 Suppl, S376–81 (2009).

169. Fuhrman, B. et al. Atorvastatin therapy in hypercholesterolemic patients suppresses cellular uptake of oxidized-LDL by differentiating monocytes. Atherosclerosis 164, 179–185 (2002).

170. Li, L. et al. The importance of GLUT3 for de novo lipogenesis in hypoxia-induced lipid loading of human macrophages. PLoS ONE 7, e42360 (2012).

171. Ross, R. Atherosclerosis--an inflammatory disease. N. Engl. J. Med. 340, 115–126 (1999).

172. Duewell, P. et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 464, 1357–1361 (2010).

173. Rajamäki, K. et al. Cholesterol crystals activate the NLRP3 inflammasome in human macrophages: a novel link between cholesterol metabolism and inflammation. PLoS ONE 5, e11765 (2010).

174. Quinn, M. T., Parthasarathy, S., Fong, L. G. & Steinberg, D. Oxidatively modified low density lipoproteins: a potential role in recruitment and retention of monocyte/macrophages during atherogenesis. Proc. Natl. Acad. Sci. U.S.A. 84, 2995–2998 (1987).

29

Page 30: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

175. Rajavashisth, T. B. et al. Induction of endothelial cell expression of granulocyte and macrophage colony-stimulating factors by modified low-density lipoproteins. Nature 344, 254–257 (1990).

176. Snodgrass, R. G., Huang, S., Choi, I.-W., Rutledge, J. C. & Hwang, D. H. Inflammasome-mediated secretion of IL-1β in human monocytes through TLR2 activation; modulation by dietary fatty acids. J. Immunol. 191, 4337–4347 (2013).

177. Vandanmagsar, B. et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat. Med. 17, 179–188 (2011).

178. Shalhoub, J., Falck-Hansen, M. A., Davies, A. H. & Monaco, C. Innate immunity and monocyte-macrophage activation in atherosclerosis. J Inflamm (Lond) 8, 9 (2011).

179. Michelsen, K. S. et al. Lack of Toll-like receptor 4 or myeloid differentiation factor 88 reduces atherosclerosis and alters plaque phenotype in mice deficient in apolipoprotein E. Proc. Natl. Acad. Sci. U.S.A. 101, 10679–10684 (2004).

180. Lundberg, A. M. et al. Toll-like receptor 3 and 4 signalling through the TRIF and TRAM adaptors in haematopoietic cells promotes atherosclerosis. Cardiovasc. Res. 99, 364–373 (2013).

181. Kanters, E. et al. Inhibition of NF-kappaB activation in macrophages increases atherosclerosis in LDL receptor-deficient mice. J. Clin. Invest. 112, 1176–1185 (2003).

182. Kanters, E. et al. Hematopoietic NF-kappaB1 deficiency results in small atherosclerotic lesions with an inflammatory phenotype. Blood 103, 934–940 (2004).

183. Gaidt, M. M. et al. Human Monocytes Engage an Alternative Inflammasome Pathway. Immunity (2016). doi:10.1016/j.immuni.2016.01.012

184. Menu, P. et al. Atherosclerosis in ApoE-deficient mice progresses independently of the NLRP3 inflammasome. Cell Death Dis 2, e137 (2011).

185. Stewart, C. R. et al. CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nature Immunology 11, 155–161 (2010).

186. Pasare, C. & Medzhitov, R. Toll-like receptors: linking innate and adaptive immunity. Adv. Exp. Med. Biol. 560, 11–18 (2005).

187. Barlic, J., Zhang, Y., Foley, J. F. & Murphy, P. M. Oxidized lipid-driven chemokine receptor switch, CCR2 to CX3CR1, mediates adhesion of human macrophages to coronary artery smooth muscle cells through a peroxisome proliferator-activated receptor gamma-dependent pathway. Circulation 114, 807–819 (2006).

188. Frostegård, J. et al. Oxidized low density lipoprotein induces differentiation and adhesion of human monocytes and the monocytic cell line U937. Proc. Natl. Acad. Sci. U.S.A. 87, 904–908 (1990).

189. Hayden, J. M. et al. Induction of monocyte differentiation and foam cell formation in vitro by 7-ketocholesterol. J. Lipid Res. 43, 26–35 (2002).

190. Spann, N. J. et al. Regulated accumulation of desmosterol integrates macrophage lipid metabolism and inflammatory responses. Cell 151, 138–152 (2012).

191. Guijas, C., Meana, C., Astudillo, A. M., Balboa, M. A. & Balsinde, J. Foamy Monocytes Are Enriched in cis-7- Hexadecenoic Fatty Acid (16:1n-9), a Possible Biomarker for Early Detection of Cardiovascular Disease. Cell Chemical Biology 23, 689–699 (2016).

192. Zhu, X. et al. Macrophage ABCA1 reduces MyD88-dependent Toll-like receptor trafficking to lipid rafts by reduction of lipid raft cholesterol. J. Lipid Res. 51, 3196–3206 (2010).

30

Page 31: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

193. Murphy, A. J. et al. Neutrophil Activation Is Attenuated by High-Density Lipoprotein and Apolipoprotein A-I in In Vitro and In Vivo Models of Inflammation. Arteriosclerosis, Thrombosis, and Vascular Biology 31, 1333–1341 (2011).

194. Yvan-Charvet, L. et al. Increased Inflammatory Gene Expression in ABC Transporter–Deficient Macrophages. Circulation 118, 1837–1847 (2008).

195. Iqbal, A. J. et al. Acute exposure to apolipoprotein A1 inhibits macrophage chemotaxis in vitro and monocyte recruitment in vivo. eLife Sciences 5, e15190 (2016).

196. Tall, A. R. & Yvan-Charvet, L. Cholesterol, inflammation and innate immunity. Nat. Rev. Immunol. 15, 104–116 (2015).

197. Björkegren, J., Karpe, F., Milne, R. W. & Hamsten, A. Differences in apolipoprotein and lipid composition between human chylomicron remnants and very low density lipoproteins isolated from fasting and postprandial plasma. J. Lipid Res. 39, 1412–1420 (1998).

198. Brynes, A. E. et al. A randomised four-intervention crossover study investigating the effect of carbohydrates on daytime profiles of insulin, glucose, non-esterified fatty acids and triacylglycerols in middle-aged men. BJN 89, 207–218 (2003).

199. Nordestgaard, B. G., Benn, M., Schnohr, P. & Tybjærg-Hansen, A. Nonfasting triglycerides and risk of myocardial infarction, ischemic heart disease, and death in men and women. JAMA 298, 299–308 (2007).

200. Nordestgaard, B. G. et al. Fasting is not routinely required for determination of a lipid profile: clinical and laboratory implications including flagging at desirable concentration cut-points-a joint consensus statement from the European Atherosclerosis Society and European Federation of Clinical Chemistry and Laboratory Medicine. European Heart Journal 37, 1944–1958 (2016).

201. Oh, J. & Hegele, R. A. HIV-associated dyslipidaemia: pathogenesis and treatment. Lancet Infect Dis 7, 787–796 (2007).

202. Bruce, I. N. ‘Not only...but also’: factors that contribute to accelerated atherosclerosis and premature coronary heart disease in systemic lupus erythematosus. Rheumatology 44, 1492–1502 (2005).

203. Ishibashi, S., Herz, J., Maeda, N., Goldstein, J. L. & Brown, M. S. The two-receptor model of lipoprotein clearance: tests of the hypothesis in ‘knockout’ mice lacking the low density lipoprotein receptor, apolipoprotein E, or both proteins. Proc. Natl. Acad. Sci. U.S.A. 91, 4431–4435 (1994).

204. Cox, R. A. & García-Palmieri, M. R. in Clinical Methods The History, Physical, and Laboratory Examinations (Butterworths, 1990).

205. Llodra, J. et al. Emigration of monocyte-derived cells from atherosclerotic lesions characterizes regressive, but not progressive, plaques. Proc. Natl. Acad. Sci. U.S.A. 101, 11779–11784 (2004).

206. Trogan, E. et al. Gene expression changes in foam cells and the role of chemokine receptor CCR7 during atherosclerosis regression in ApoE-deficient mice. Proc. Natl. Acad. Sci. U.S.A. 103, 3781–3786 (2006).

207. Barcia, A. M. & Harris, H. W. Triglyceride-rich lipoproteins as agents of innate immunity. Clin. Infect. Dis. 41 Suppl 7, S498–503 (2005).

208. Johnston, T. P. The P-407–Induced Murine Model of Dose-Controlled Hyperlipidemia and Atherosclerosis: A Review of Findings To Date. Journal of Cardiovascular Pharmacology 43, 595 (2004).

31

Page 32: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

209. Ono-Moore, K. D. et al. Postprandial Inflammatory Responses and Free Fatty Acids in Plasma of Adults Who Consumed a Moderately High-Fat Breakfast with and without Blueberry Powder in a Randomized Placebo-Controlled Trial. J. Nutr. 146, 1411–1419 (2016).

210. Iqbal, R. et al. Dietary patterns and the risk of acute myocardial infarction in 52 countries: results of the INTERHEART study. Circulation 118, 1929–1937 (2008).

211. Norris, J. N. Controlled trial of soya bean oil in myocardial infarction: report of a research committee to the Medical Research Council. (Lancet, 1968).

212. No Authors Listed. Los Angeles Veterans Administration diet study. Nutr. Rev. 27, 311–316 (1969).

213. Turpeinen, O. et al. Dietary prevention of coronary heart disease: the Finnish Mental Hospital Study. Int J Epidemiol 8, 99–118 (1979).

214. Watts, G. F. et al. Effects on coronary artery disease of lipid-lowering diet, or diet plus cholestyramine, in the St Thomas' Atherosclerosis Regression Study (STARS). The Lancet 339, 563–569 (1992).

215. Mozaffarian, D., Micha, R. & Wallace, S. Effects on coronary heart disease of increasing polyunsaturated fat in place of saturated fat: a systematic review and meta-analysis of randomized controlled trials. PLoS Med. 7, e1000252 (2010).

216. Leren, P. The effect of plasma cholesterol lowering diet in male survivors of myocardial infarction. A controlled clinical trial. Acta Med. Scand. Suppl. 466, 1–92 (1966).

217. Brunner, E. J. et al. Dietary patterns and 15-y risks of major coronary events, diabetes, and mortality. The American Journal of Clinical Nutrition 87, 1414–1421 (2008).

218. Schwingshackl, L. & Hoffmann, G. Comparison of effects of long-term low-fat vs high-fat diets on blood lipid levels in overweight or obese patients: a systematic review and meta-analysis. J Acad Nutr Diet 113, 1640–1661 (2013).

219. Leren, P. The Oslo Diet-Heart Study: Eleven-Year Report. Circulation 42, 935–942 (1970).

220. Khymenets, O. et al. Mononuclear cell transcriptome response after sustained virgin olive oil consumption in humans: an exploratory nutrigenomics study. OMICS 13, 7–19 (2009).

221. Estruch, R. et al. Primary prevention of cardiovascular disease with a Mediterranean diet. N. Engl. J. Med. 368, 1279–1290 (2013).

222. Bouwens, M. et al. Fish-oil supplementation induces antiinflammatory gene expression profiles in human blood mononuclear cells. The American Journal of Clinical Nutrition 90, 415–424 (2009).

223. Bouwens, M., Grootte Bromhaar, M., Jansen, J., Müller, M. & Afman, L. A. Postprandial dietary lipid-specific effects on human peripheral blood mononuclear cell gene expression profiles. The American Journal of Clinical Nutrition 91, 208–217 (2010).

224. Chowdhury, R. et al. Association of Dietary, Circulating, and Supplement Fatty Acids With Coronary Risk: A Systematic Review and Meta-analysis. Ann. Intern. Med. 160, 398–406 (2014).

225. Eckel, R. H. et al. 2013 AHA/ACC guideline on lifestyle management to reduce cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Journal of the American College of

32

Page 33: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Cardiology 63, 2960–2984 (2014).226. Harcombe, Z. et al. Evidence from randomised controlled trials did not support the

introduction of dietary fat guidelines in 1977 and 1983: a systematic review and meta-analysis. Open Heart 2, e000196 (2015).

227. de Souza, R. J. et al. Intake of saturated and trans unsaturated fatty acids and risk of all cause mortality, cardiovascular disease, and type 2 diabetes: systematic review and meta-analysis of observational studies. BMJ 351, h3978–16 (2015).

228. Bellido, C. & López-Miranda, J. Butter and walnuts, but not olive oil, elicit postprandial activation of nuclear transcription factor κB in peripheral blood mononuclear cells from healthy men. The American journal … (2004).

229. Erridge, C., Attina, T., Spickett, C. M. & Webb, D. J. A high-fat meal induces low-grade endotoxemia: evidence of a novel mechanism of postprandial inflammation. The American Journal of Clinical Nutrition 86, 1286–1292 (2007).

230. Amar, J. et al. Energy intake is associated with endotoxemia in apparently healthy men. The American Journal of Clinical Nutrition 87, 1219–1223 (2008).

231. Ghoshal, S., Witta, J., Zhong, J., de Villiers, W. & Eckhardt, E. Chylomicrons promote intestinal absorption of lipopolysaccharides. J. Lipid Res. 50, 90–97 (2008).

232. Ghanim, H. et al. Increase in plasma endotoxin concentrations and the expression of Toll-like receptors and suppressor of cytokine signaling-3 in mononuclear cells after a high-fat, high-carbohydrate meal: implications for insulin resistance. Diabetes Care 32, 2281–2287 (2009).

233. Cullberg, K. B., Larsen, J. O., Pedersen, S. B. & Richelsen, B. Effects of LPS and dietary free fatty acids on MCP-1 in 3T3-L1 adipocytes and macrophages in vitro. 4, e113–5 (2014).

234. Huang, S. et al. Saturated fatty acids activate TLR-mediated proinflammatory signaling pathways. J. Lipid Res. 53, 2002–2013 (2012).

235. Eguchi, K. et al. Saturated fatty acid and TLR signaling link β cell dysfunction and islet inflammation. Cell Metab. 15, 518–533 (2012).

236. Chait, A. & Kim, F. Saturated fatty acids and inflammation: who pays the toll? Arteriosclerosis, Thrombosis, and Vascular Biology 30, 692–693 (2010).

237. Youssef-Elabd, E. M. et al. Acute and chronic saturated fatty acid treatment as a key instigator of the TLR-mediated inflammatory response in human adipose tissue, in vitro. The Journal of Nutritional Biochemistry 23, 39–50 (2012).

238. Erridge, C. & Samani, N. J. Saturated Fatty Acids Do Not Directly Stimulate Toll-Like Receptor Signaling. Arteriosclerosis, Thrombosis, and Vascular Biology 29, 1944–1949 (2009).

239. Klop, B. et al. Leukocyte cell population data (volume conductivity scatter) in postprandial leukocyte activation. Int. Jnl. Lab. Hem. 35, 644–651 (2013).

240. Varela, L. M. et al. ScienceDirect. The Journal of Nutritional Biochemistry 24, 2031–2039 (2013).

241. Bays, H. E. Adiposopathy is ‘sick fat’ a cardiovascular disease? J. Am. Coll. Cardiol. 57, 2461–2473 (2011).

242. Bays, H. E. et al. Obesity, adiposity, and dyslipidemia: a consensus statement from the National Lipid Association. J Clin Lipidol 7, 304–383 (2013).

243. Bays, H. E. Adiposopathy, diabetes mellitus, and primary prevention of atherosclerotic coronary artery disease: treating ‘sick fat’ through improving fat function with antidiabetes therapies. Am. J. Cardiol. 110, 4B–12B (2012).

33

Page 34: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

244. Pearce, E. N. Update in Lipid Alterations in Subclinical Hypothyroidism. J. Clin. Endocrinol. Metab. 97, 326–333 (2012).

245. Omran, J., Al-Dadah, A. & Dellsperger, K. C. Dyslipidemia in patients with chronic and end-stage kidney disease. Cardiorenal Med 3, 165–177 (2013).

246. Kolovou, G. D., Anagnostopoulou, K. K., Kostakou, P. M., Bilianou, H. & Mikhailidis, D. P. Primary and secondary hypertriglyceridaemia. Curr Drug Targets 10, 336–343 (2009).

247. Kasiske, B. L. Effects of Antihypertensive Therapy on Serum Lipids. Ann. Intern. Med. 122, 133–141 (1995).

248. Darling, G. M., Johns, J. A., McCloud, P. I. & Davis, S. R. Estrogen and progestin compared with simvastatin for hypercholesterolemia in postmenopausal women. N. Engl. J. Med. 337, 595–601 (1997).

249. Lobo, R. A., Skinner, J. B., Lippman, J. S. & Cirillo, S. J. Plasma lipids and desogestrel and ethinyl estradiol: a meta-analysis. Fertility and Sterility 65, 1100–1109 (1996).

250. Henderson, D. C. Clozapine, Diabetes Mellitus, Weight Gain, and Lipid Abnormalities: A Five-Year Naturalistic Study. American Journal of Psychiatry 157, 975–981 (2000).

251. Nahrendorf, M. et al. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med 204, 3037–3047 (2007).

252. Serbina, N. V. & Pamer, E. G. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nature Immunology 7, 311–317 (2006).

253. Debien, E. et al. S1PR5 is pivotal for the homeostasis of patrolling monocytes. Eur. J. Immunol. 43, 1667–1675 (2013).

254. Tacke, F. Functional role of intrahepatic monocyte subsets for the progression of liver inflammation and liver fibrosis in vivo. Fibrogenesis Tissue Repair 5, S27 (2012).

Acknowledgments

K.J.W laboratory is funded by BHF CRTF to S.R. (FS/14/50/30856), MRC (MR/M003159/1),

Kidney Research UK (RP_019_20160303) and Imperial Biomedical Research Centre. A.J.M is

supported by a Career Development Fellowship from the NHMRC, a Future Leader

Fellowship from the NHF, and Project Grants from the NHMRC and Diabetes Australia. We

would like to thank members of the Woollard lab for helpful discussion and lab work

underpinning data for this Review.

Author contributions

S.R. and K.J.W. researched data for the article and wrote the manuscript. A.J.M. and K.J.W.

provided substantial contribution to the discussion of content, and reviewed and edited the

manuscript before submission.

34

Page 35: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Competing interests statement

The authors declare no competing interests.

Key points

The mononuclear phagocyte system comprises heterogeneous cells derived from

fetal and bone marrow precursors

Seeding of fetal tissue macrophages is distinct from monocyte-derived macrophages,

highlighting the independent effector functions of monocyte subsets

Dyslipidaemia, characterized by hypercholesterolaemia and/or

hypertriglyceridaemia, increases the risk of cardiovascular disease

Dyslipidaemia increases the production of monocytes through myelopoiesis

Blood monocyte subsets under dyslipidaemic conditions change their behaviour and

activation, and increase their extravasation in response to triglyceride-rich

lipoproteins and cholesterol

Box 1 | Definitions of hyperlipidaemia

[b1] Hyperlipidaemia

Hyperlipidaemia is used to describe elevated levels of one or more of total

cholesterol, low-density lipoprotein cholesterol and triglycerides (TG).

[b1] Hypertriglyceridaemia

Hypertriglyceridaemia is an aberrantly elevated level of TG in the blood (normal <1.7,

high 1.7-9.9, severe >10 mmol/L). TG represent the bulk form in which fatty acids (fat)

are transported in the blood. Triglycerides are packaged into lipoproteins,

predominantly in chylomicrons and very-low density lipoproteins (VLDL). Chylomicrons

transport the bulk of dietary fat, whereas VLDL transport fatty acids coming from the

diet (exogenous) and produced in the body (endogenous).

35

Page 36: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Elevated levels of nonfasting TG are correlated with increased risk of cardiovascular

disease144,145,147,148.

[b1] Hypercholesterolaemia

Hypertriglyceridaemia can be found in isolation or in addition to elevated levels of

cholesterol (hypercholesterolemia). Of note, elevated cholesterol levels refer to levels of

cholesterol-carrying lipoproteins, LDL and HDL.

High LDL levels strongly correlate with increased risk of cardiovascular disease103,104,107-111.

Conversely, high HDL levels might be correlated with reduced a reduced risk of

cardiovascular disease126.

[b1] Classification

Hyperlipidaemia can be caused by primary (genetic) factors or by secondary

(nongenetic) factors such as lifestyle habits or medical conditions

[b2] Genetic (primary) hyperlipidaemia.

Familial combined hyperlipidaemia. Polygenetic cause. Elevation of LDL, TG or

both.

Familial hypercholesterolemia (heterozygous or homozygous). Mutation in LDL

receptor leads to elevated LDL cholesterol.

Familial dysbetalipoproteinaemia. Autosomal recessive. Majority homozygous

mutation in ApoE. Elevated total cholesterol and TG. Associated with metabolic

syndrome.

Hypertriglyceridemia. Elevated TG. Common and rare genetic variants. Moderate

severity (2-10 mmol/L) caused by polygenetic effect influencing VLDL production

and removal. Monogenic causes: LPL, ApoC2, ApoA5, LMF1, GPIHBP1, GPD1. A

gain of function mutation in ApoC3 leads to high ApoC3/TG levels.

Other genetic disorders of lipoprotein metabolism, e.g. Tangiers disease (very

low HDL).

[b2] Secondary hyperlipidaemia. Several factors can cause hyperlipidaemia, such as

lifestyle habits (high fat diet, physical inactivity, alcohol consumption)241,242, medical

conditions including diabetes mellitus243, hypothyroidism244, and some forms of

36

Page 37: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

kidney disease245,246, and the use of certain drugs (e.g. anti-hypertensive agents247,

hormone replacement therapy248, contraceptives249 and some antipsychotics250).

37

Page 38: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Figure 1 | Monocyte and macrophage ontogeny. Monocytes derive from haematopoietic

precursors in bone marrow or spleen. Monocytes give rise to classical subsets — defined in

mice as lymphocyte antigen 6C (LY6C)high and in humans as CD14highCD16low — and

nonclassical subsets — defined in mice as LY6Clow and in humans as CD14lowCD16+.

Production and emigration of monocyte subsets depends on defined signals and chemokine

receptors, such as C-C motif chemokine 2 (CCL2)36,83, C-C chemokine receptor type 2

(CCR2)16,36,38,76,251,252, and C-X-C chemokine receptor type 4 (CXCR4)61 for LY6Chigh monocytes,

and the microRNA miR-14631, nuclear receptor subfamily 4 group A member 1 (NR4A1)52,

transcription factor RelB31, and sphingosine 1-phosphate receptor 5 (S1PR5)253 for LY6Clow

monocytes. Some monocytes remain attached to the endothelium and patrol the blood

vessel lumen; other LY6Clow monocytes can derive from LY6Chigh monocytes. Inflammatory

cues induce monocyte extravasation (particularly of LY6Chigh monocytes) and polarization

into macrophages. These monocyte-derived macrophages mix with tissue-resident

macrophages derived from yolk sac and fetal liver precursors. The macrophage pool

receives tissue specific signals to define phenotypes and effector functions and, along with

other cues such as damage-associated molecular patterns (DAMPs) and pathogen-

associated molecular patterns (PAMPs), define the spectrum of tissue macrophages found in

vivo. Dotted lines represent hypothesized or unknown pathways. AGTR1A, type 1A

angiotensin II receptor; Ang II, angiotensin II; cMoP, common monocyte progenitor; CMP,

common myeloid progenitor; CX3CL1, CX3C-chemokine ligand 1; CX3CR1, CX3C-chemokine

receptor 1; GMP, granulocyte-macrophage progenitor; HSC, haematopoietic stem cell,

ICAM, intercellular adhesion molecule; Int, intermediate; JAMs, junctional adhesion

molecules; LFA1, lymphocyte function-associated antigen 1; MDP, macrophage and

dendritic cell precursor; PECAM1, platelet endothelial cell adhesion molecule; PSGL1, P-

selectin glycoprotein ligand 1; VLA4, very late antigen 4; VCAM1, vascular cell adhesion

molecule 1.

Figure 2 | Effect of dyslipidaemia on monocyte recruitment, migration, and cell behaviour.

a | Hypercholesterolaemia promotes production of classical monocytes, defined in mice as

lymphocyte antigen 6C (LY6C)high, by inducing the proliferation of bone marrow precursors,

38

Page 39: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

which in turn leads to blood monocytosis, endothelial dysfunction, and increased

recruitment mediated by chemokine receptor signals such as C-C chemokine receptor type 5

(CCR5)83,254, and ultimately leads to increased tissue monocyte-derived macrophages.

Hypertriglyceridaemia increases accumulation of patrolling nonclassical monocytes (LY6C low)

and extravasation, possibly mediated by C-C motif chemokine 4 (CCL4)66. Dyslipidaemia

might lead to increased proliferation of tissue-resident macrophages. Monocytes can

endocytose circulating lipids, leading to formation of ‘foamy’ monocytes, which might then

deliver lipids from the blood to peripheral tissues, contributing to increased lipid retention

in tissues (via heparin sulphate proteoglycans; HSPGs) and to increased lipid endocytosis by

tissue macrophages, promoting foam cell formation. Red lines indicate pathways

upregulated in dyslipidaemia. b | Lipids can enter the cell via regulated uptake, mediated

for example by the LDL receptor (LDLR) and the LDLR-related protein 1 (LRP1), or via

unregulated uptake, mediated among others by scavenger receptors such as CD36 and

SRA1, and by Toll-like receptors (TLRs). Neutral lipids and cholesterol can then accumulate

inside the cell, leading to proinflammatory or anti-inflammatory responses depending on

the lipid species and lipid modification. Lipid entry will also lead to activation of intracellular

cholesterol storage and efflux pathways (reverse cholesterol transport). Increased lipid

loading modulates the cytoskeleton by blocking signalling components such as the GTPase

RAS homologue gene family member A (RHOA), leading to defects in migration and possible

tissue retention of macrophages65. ABCA1, ATP-binding cassette transporter subfamily A

member 1, ABCG1, ATP-binding cassette transporter subfamily G member 1; CXCL16, C-X-C

motif chemokine 16; LOX1, lectin-type oxidized LDL receptor 1; LPL, lipoprotein lipase;

mmLDL, minimally modified LDL; oxLDL, oxidized LDL; NEFA, non-esterified fatty acid; TGRL,

triglyceride-rich lipoprotein; VLDL, very low-density lipoprotein.

39

Page 40: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

Table 1 | Monocyte subsets in blood

Monocyte subset

Common surface proteins used as lab markers to differentiate subsets

Mouse

Classical LY6Chigh CX3CR1int CD62L+ CCR2+ CD43low

Nonclassical LY6Clow CX3CR1high CD62L- CCR2- CD43high

Human

Classical CD14high CD16low CD62L+ CD43low HLA-DRlow

Nonclassical CD14low CD16high CD62L- CD43high HLA-DRhigh

Intermediate CD14high CD16high CD62Lint CD43int HLA-DRint

Glossary

Mononuclear phagocyte system: part of the immune system that consists of phagocytic cells, namely monocytes and macrophages but also tissue specific phagocytes such as Kupffer cells, microglia, osteoclasts and Langerhans cells Adoptive transfer: a technique for introducing labelled cells of interest into a host animal for the purposes of tracking and/or imaging, or to ascertain effects of the donor cells on a host

Parabiosis model: a technique used in order to study the effects of one biological system on another through the surgical conjoining of circulatory and physiological systems. Mice and rats are commonly use in these models.

Aorta–gonad–mesonephros: region of the embryonic mesoderm that includes the dorsal aorta and is implicated in definitive haematopoietic stem cell myelopoeiesis.

Common monocyte progenitor: a monocyte progenitor cell in the bone marrow distinct from macrophage and dendritic cell precursor (MDP) cells

Plasmacytoid dendritic cell: although their appearance resembles that of plasma cells, their function resembles that of conventional myeloid dendritic cells (DCs)

Lipoprotein: a carrier molecule that contains and transports lipid (namely cholesterol and triglyceride) within the circulation, and consists of apolipoprotein molecules and a single layer phospholipid membrane. Examples include LDL, VLDL and HDL as well as chylomicrons.

40

Page 41: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

ABC transporters: ATP-binding cassette transporters: actively transport molecules in or out of cells. An example is the ABCA1 transporter that plays a role in cholesterol export to HDL.

Oxidized LDL: an LDL molecule where the lipoprotein envelope and/or cholesterol content has been oxidised by free radicals. Oxidised forms of LDL are thought to play an important role in atherosclerosis and can stimulate macrophages accumulate lipid and accelerate conversion to macrophage foam cells.

Reverse cholesterol transport: the transport of cholesterol from tissues or cells out and back to the liver for clearance. Cholesterol can be transported via ABCA1 to HDL that acts as an acceptor molecule. This process is thought to be cardioprotective.

Neutral lipid droplets: lipids without a polar charge that can be found within cells, such as adipocytes and commonly refers to cholesterol esters and triglycerides.

NLRP3 inflammasome: an important oligomer molecule that forms part the cellular inflammatory response, particularly in innate immune cells resulting in the release of cytokines such as interleukin-1 beta.

Minimally modified LDL: a unique form of oxidised LDL enriched with aldehyde containing phosphatidylcholines

Reconstituted HDL: a synthetic form of HDL used in order to improve cholesterol transport out of the cell (efflux) with potential therapeutic value against cardiovascular disease.

Chylomicrons: also known as "ultra-low density lipoprotein", these large lipoprotein particles are secreted by the gut endothelial cells and predominantly transport dietary lipid (triglycerides and cholesterol) in the circulation. Chylomicron levels rise following dietary intake of fat or carbohydrate and fall following fasting.

Very low-density lipoprotein: these lipoproteins predominantly transport triglyceride produced by the body (muscle, liver) but can also be altered by dietary intake.

Author biographies

Dr Shamim Rahman is a Cardiology Specialist Registrar in London, specialising in Coronary Intervention. He is currently reading towards a PhD studying the role of monocyte subset responses to dietary lipid in the context of atherosclerosis, infection and immunity.

Associate Professor Andrew Murphy is the head of the Haematopoiesis and Leukocyte Biology Laboratory at Baker Heart and Diabetes Institute in Melbourne, Australia. His main

41

Page 42: spiral.imperial.ac.uk:8443€¦  · Web viewEffects of dyslipidaemia on monocyte production and function in cardiovascular disease . Shamim Rahman1, Andrew J. Murphy2,3 and Kevin

interest is how the haematopoietic system is altered in the setting of cardiovascular disease and comorbidities. His group also studies fundamental biological process of haematopoiesis.

Dr Kevin Woollard is a group leader within the Division of Immunology and Inflammation at Imperial College London. His lab studies the ontogeny and effector functions of monocyte subpopulations and their role in vascular inflammation. Their main focus is the biology of monocytes and macrophages in autoimmune vasculopathies and atherosclerosis.

42