ArticlePDF Available

Hyperglycaemia and apoptosis of microglial cells in human septic shock

Authors:

Abstract and Figures

The effect of hyperglycaemia on the brain cells of septic shock patients is unknown. The objective of this study was to evaluate the relationship between hyperglycaemia and apoptosis in the brains of septic shock patients. In a prospective study of 17 patients who died from septic shock, hippocampal tissue was assessed for neuronal ischaemia, neuronal and microglial apoptosis, neuronal Glucose Transporter (GLUT) 4, endothelial inducible Nitric Oxide Synthase (iNOS), microglial GLUT5 expression, microglial and astrocyte activation. Blood glucose (BG) was recorded five times a day from ICU admission to death. Hyperglycaemia was defined as a BG 200 mg/dL g/l and the area under the BG curve (AUBGC) > 2 g/l was assessed. Median BG over ICU stay was 2.2 g/l. Neuronal apoptosis was correlated with endothelial iNOS expression (rho = 0.68, P = 0.04), while microglial apoptosis was associated with AUBGC > 2 g/l (rho = 0.70; P = 0.002). Neuronal and microglial apoptosis correlated with each other (rho = 0.69, P = 0.006), but neither correlated with the duration of septic shock, nor with GLUT4 and 5 expression. Neuronal apoptosis and ischaemia tended to correlate with duration of hypotension. In patients with septic shock, neuronal apoptosis is rather associated with iNOS expression and microglial apoptosis with hyperglycaemia, possibly because GLUT5 is not downregulated. These data provide a mechanistic basis for understanding the neuroprotective effects of glycemic control.
Content may be subject to copyright.
RESEARCH Open Access
Hyperglycaemia and apoptosis of microglial cells
in human septic shock
Andrea Polito
1
, Jean-Philippe Brouland
2
, Raphael Porcher
3
, Romain Sonneville
1,6
, Shidasp Siami
1
,
Robert D Stevens
4
, Céline Guidoux
1
, Virginie Maxime
1
, Geoffroy Lorin de la Grandmaison
5
, Fabrice C Chrétien
6
,
Françoise Gray
2
, Djillali Annane
1
and Tarek Sharshar
1*
Abstract
Introduction: The effect of hyperglycaemia on the brain cells of septic shock patients is unknown. The objective of
this study was to evaluate the relationship between hyperglycaemia and apoptosis in the brains of septic shock
patients.
Methods: In a prospective study of 17 patients who died from septic shock, hippocampal tissue was assessed for
neuronal ischaemia, neuronal and microglial apoptosis, neuronal Glucose Transporter (GLUT) 4, endothelial
inducible Nitric Oxide Synthase (iNOS), microglial GLUT5 expression, microglial and astrocyte activation. Blood
glucose (BG) was recorded five times a day from ICU admission to death. Hyperglycaemia was defined as a BG 200
mg/dL g/l and the area under the BG curve (AUBGC) > 2 g/l was assessed.
Results: Median BG over ICU stay was 2.2 g/l. Neuronal apoptosis was correlated with endothelial iNOS expression
(rho = 0.68, P= 0.04), while microglial apoptosis was associated with AUBGC > 2 g/l (rho = 0.70; P= 0.002).
Neuronal and microglial apoptosis correlated with each other (rho = 0.69, P= 0.006), but neither correlated with
the duration of septic shock, nor with GLUT4 and 5 expression. Neuronal apoptosis and ischaemia tended to
correlate with duration of hypotension.
Conclusions: In patients with septic shock, neuronal apoptosis is rather associated with iNOS expression and
microglial apoptosis with hyperglycaemia, possibly because GLUT5 is not downregulated. These data provide a
mechanistic basis for understanding the neuroprotective effects of glycemic control.
Introduction
Sepsis and septic shock are associated with hyperglycae-
mia and peripheral insulin resistance [1,2]. Glycemic
control strategies are commonly instituted as adjunctive
therapeutic measures in critically ill patients, although
recent studies have not consistently shown a benefit
from intensive insulin therapy [3-6]. One argument sup-
porting blood glucose control is that intensive insulin
therapy is associated with a protective effect on the per-
ipheral and central nervous system [7]. While it has
been shown that intensive insulin therapy reduces the
incidence of critical illness neuromyopathy [7,8] and
that hyperglycaemia worsens brain injury in ischemic
stroke [9-11] and head trauma [12], the effect of hyper-
glycaemia or insulin on sepsis related brain dysfunction
is not well understood. A recent in vitro study showed
that hyperglycaemia increased microglial vulnerability to
lipopolysaccharide (LPS) mediated toxicity [13], through
formation of oxidative free radicals. Interestingly, it has
also been shown that experimental sepsis induces oxida-
tivedamagesinthebrain[14].Inapreviousneuro-
pathological study, we found that septic shock is
associated with neuronal ischaemia, microglial activation
and apoptosis as well as neuronal apoptosis, which was
statistically correlated with endothelial expression of
iNOS [15]. However, the relationships between BG and
neuropathological findings have not been thoroughly
assessed. The objective of the present study was to
address this issue and also to assess whether hypergly-
caemia is associated with neuronal or microglial
* Correspondence: tarek.sharshar@rpc.aphp.fr
1
General Intensive Care Medicine, Raymond Poincaré Hospital (AP-HP),
University of Versailles Saint Quentin en Yvelines, 104 bd R. Poincaré,
Garches 92210, France
Full list of author information is available at the end of the article
Polito et al.Critical Care 2011, 15:R131
http://ccforum.com/content/15/3/R131
© 2011 Polito et al.; licensee BioMed Central Ltd. This is an open access article distributed under the terms of the Creative Commons
Attribution License (http://creative commons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in
any medium, pro vided the original work is properly cited.
apoptosis after adjustment to other pro-apoptotic fac-
tors. We also evaluated brain expression of Glucose
Transporter (GLUT) proteins given their role in trans-
membrane glucose transport in neurons and microglial
cells during stress conditions [16]. Assessment of the
relationships between hyperglycaemia and neuropatholo-
gical abnormalities might provide insight on the
mechanisms of sepsis associated neurological and psy-
cho-cognitive long-term consequences.
Materials and methods
Patients
We investigated consecutive patients who died from
septic shock while receiving treatment in the ICU at
Raymond Poincaré University Hospital, Garches, France
[15]. Exclusion criteria were: age younger than 18 years;
pregnancy; evidence of an underlying degenerative neu-
rological disease determined clinically or on post-mor-
tem examination, or any concomitant disease other than
infection that might have accounted for shock and
death. We obtained informed consent from the patients
closest relatives. The protocol was approved by the
Comité Consultatif de Protection des Personnes se Prê-
tantàlaRechercheBiomédicaledeSaintGermainen
Laye, France.
Data collection
Demographic characteristics, pre-existing risk factors for
vascular disease, and severity of illness using simplified
acute physiology score II (SAPS II) [17] and sequential
organ failure assessment (SOFA) [17,18] score were rou-
tinely recorded. Vital signs were recorded continuously,
enabling calculation of duration of shock and cumula-
tive time passed with a mean blood pressure of less
than 60 mmHg. Standard laboratory tests and relevant
microbiological data were recorded daily. All arterial
and capillary BG levels measured between admission
and death were collected. Hyperglycaemia and hypogly-
caemia were considered when BG levels were above 2 g/
l and 0.4 g/l, respectively [5,6,19]. Then, we assessed the
highest and lowest BG, highest variation (Δmax) in one
dayinBG,meanBG,areaundertheBGcurve
(AUBGC), and AUBGC above 2 g/l (that is, hyperglycae-
mia). The AUBGC cut-off of 2 g/L was chosen because
it reflected a compromise between the duration of
hyperglycaemia and the value of blood glucose. This
cut-off also allows to account for the irregular times
intervals between sample collection. We also assessed
the percentage of follow-up time in hypoglycaemia and
in hyperglycaemia, as well as the proportion of patients
who were treated with insulin and who developed hypo-
glycaemia and hyperglycaemia. We defined prolonged
hyperglycaemia as BG values higher than 2 g/l more
than 50% of follow-up time (with linear interpolation
between two consecutive blood samplings). During the
study period (from 1997 to 2001), no specific protocol
for the management of hyperglycaemia had been
implemented.
Brain sampling
Brain samples were collected within 12 h of death.
Gross examination of the brain was done after four to
six weeks of formalin-fixation on coronal sections of the
cerebral hemispheres and horizontal sections of the
brain stem and cerebellum. Macroscopic changes were
noted, and we selected the hippocampus for microscopic
examination, after paraffin embedding. We decided to
evaluate changes in the hippocampus as it is highly vul-
nerable to metabolic insults, hypoxemia and ischaemia
[20,21]. Sections were stained with haematoxylin and
eosin and Bodian silver impregnation combined with
Luxol fast blue.
Ischaemia, gliosis and apoptosis
Histological analysis was performed by one observer
(FG) who was blinded to glycemic levels. As previously
described [15,22], neurons were described as ischaemic
when they presented with shrunken eosinophilic cyto-
plasm and pyknotic nuclei. Glial reaction (that is, glio-
sis) was identified as rod-shaped microglial cells and
astrocytes with clear nuclei. Astrocyte and microglial
activation was assessed by evaluating immunohisto-
chemical expression of glial fibrillary acidic protein
(GFAP, Dako, Glostrup, Denmark) and MHC class II
antigens (HLA-DR) (Dako), CD68 (Dako). Axonal
damage was assessed using immunohistochemistry for
Amyloid Precursor ProteinA4(beta-APP)(MAB348,
Chemicon, Lyon, France). Tissue expression of GLUT1
(a3536, Dako), GLUT3 (ab41525, Abcam, Cambridge,
UK), GLUT4 (ab65976, Abcam) and GLUT5 (ab36057,
Abcam), were also assessed as well as that of tumor
necrosis factor a(TNFa) (Genzyme, Dako) and induci-
ble NO synthase [23]. We previously found that sepsis-
associated expression of TNFaand iNOS involve glial
and endothelial cells, respectively [15]. Apoptosis was
identified using a caspase 3 monoclonal antibody
(Dako), and by in-situ end labelling (ISEL) [24] with
use of ApopTag kit (Oncor, Gaithersburg, MD, USA).
Intensity of neuronal ischaemia, gliosis, glial activation
and apoptosis were graded between 0 and 3, as
described elsewhere [15]. Expression of neuronal beta-
APP, glial TNFa, endothelial iNOS, neuronal GLUT 1,
GLUT3, GLUT4 and microglial GLUT5 expression
were also graded from 0 to 3 [15]. Because immunos-
taining of GLUT3 was not satisfactory and that of
GLUT1 immunostainings did not vary among patients,
we did not assess their statistical correlation with
blood glucose level.
Polito et al.Critical Care 2011, 15:R131
http://ccforum.com/content/15/3/R131
Page 2 of 8
Statistical analyses
Quantitative and qualitative variables were expressed as
median (interquartile range, IQR) and percentage,
respectively. Association between continuous variables
was assessed by non parametric Spearman correlation
coefficient. Adjustment was performed by multiple lin-
ear models based on ranks, in accordance to the use of
non-parametric rank correlation coefficients. Continuous
and categorical variables were compared between groups
of patients by Wilcoxon rank-sum test and Fishers
exact test, respectively. Values of P<0.05wereconsid-
ered as indicating statistical significance. All statistical
analyses were performed using R 2.6.2 statistical soft-
ware (The R Foundation for Statistical Computing,
Vienna, Austria).
Results
From 1997 to 2001, 17 patients who died from septic
shock were included. Patient characteristics are pre-
sented in Table 1. Septic shock had a median duration
of four days and was mainly secondary to pneumonia or
cellulitis. Four patients had pre-existing diabetes melli-
tus. Median BG over ICU stay was 2.17 g/l. Episodes of
hyperglycaemia were observed in all patients and hypo-
glycaemia occurred in five (29%) patients. Nine (53%)
patients developed prolonged hyperglycaemia and six
(35%) were treated with insulin (with mean BG level of
2.7 g/L (1.9 to 3.0)). Macroscopic findings were ischae-
mia (n = 12), haemorrhage (n = 9) and disseminated
abcesses (n = 3). Oedema was observed in only one
patient.
In contrast to HLA-DR, expression of microglial CD68
tended to be correlated with AUBGC > 2 g/l (rho =
0.44, P= 0.08). Intensity of neuronal and microglial
apoptosis was correlated with AUBGC > 2 g/l (rho =
0.53; P= 0.03 and rho = 0.70; P= 0.002) (Table 2, Fig-
ure 1). Intensity of neuronal beta-APP expression corre-
lated with AUBGC > 2 g/l (rho = 0.61; P= 0.03) (Figure
2). Endothelial iNOS expression was correlated with
intensity of neuronal apoptosis (rho = 0.68, P= 0.005)
but not with that of microglial apoptosis (rho = 0.34, P
= 0.17). The intensities of neuronal and microglial apop-
tosis were correlated (rho = 0.56, P=0.02).Immunos-
taining of GLUT3 was not satisfactory. GLUT1 rather
stained endothelial cells than neurons and its expression
did not vary among patients. Neuronal GLUT4 (Figure
3) and microglial GLUT5 expression (Figure 4) did not
correlate with prolonged hyperglycaemia nor with neu-
ronal or microglial apoptosis (Table 3). Expressions of
endothelial iNOS and microglial GLUT5 were inversely
correlated (rho = -0.54; P= 0.03). Neuronal and micro-
glial apoptosis were not correlated with SAPS-II at
admission, highest SOFA score, duration of septic
shock, or with serum sodium (especially hyponatremia),
lowest systolic arterial pressure, PaO
2
and SaO
2
.Inten-
sity of neuronal apoptosis and ischaemia tended to be
Table 1 Patientscharacteristics
Whole population
(n = 17)
Women (%) 6 (35)
Age (years) 68 (53 to 72)
Cerebrovascular risk factors (%) 11 (65)
Diabetes (%) 4 (24)
Medical admission 11 (65)
Site of infection
Lung only (%) 9 (53)
Abdominoperitoneal only (%) 0
Urinary tract only (%) 0
Cellulitis only (%) 5 (29)
> 1 site 5 (18)
Unknown 0
Positive culture at any site (%)
Gram-positive only (%) 4 (24)
Gram-negative only (%) 6 (35)
Fungus only (%) 0
Mixed (%) 7 (411)
Positive blood culture (%) 4 (24)
SAPS-II at admission 43 (30 to 58)
Highest OSF score during ICU stay 4 (4 to 5)
Duration of septic shock (days) 4 (2 to 10)
Cumulative time spent with MAP < 60 mm Hg (h) 11 (4 to 25)
Lowest SAP (mm Hg) 57 (33 to 66)
Lowest PaO
2
(kPa) 8.1 (6.1 to 9.0)
Lowest SaO
2
(%) 85 (73 to 90)
Highest blood sodium level (mmol/L) 139 (135 to 149)
Lowest blood sodium level (mmol/L) 132 (128 to 137)
Blood glucose level
Lowest BG (gr/l) 0.6 (0.3 to 1.1)
Highest BG (gr/l) 3.5 (3.3 to 5.4)
Δmax BG (gr/l) 3.4 (2.1 to 4.8)
Mean BG (gr/l) 2.2 (1.4 to 2.8)
Patients with hypoglycaemia (%) 5 (29)
Patients with hyperglycaemia (%) 15 (88)
Patients with prolonged hyperglycaemia (%) 9 (53)
Patients treated with insulin (%) 6 (35)
Neuronal apoptosis 1.0 (1.0 to 2.0)
Microglial apoptosis 1.0 (1.0 to 1.5)
GFAP expression 2.0 (2.0 to 3.0)
HLA-DR expression 1.0 (1.0 to 2.0)
CD68 expression 1.0 (0.5 to 1.5)
Glial TNF-aexpression 1.0 (0 to 1.0)
iNOS expression 1.0 (1.0 to 1.0)
Neuronal GLUT4 1.5 (1.0 to 2.0)
Microglial GLUT5 1.0 (0.5 to 1.0)
CD68 1.5 (1.0 to 2.0)
Beta-APP 1.0 (1.0 to 1.5)
Beta APP, beta-amyloid precursor protein; BG, blood glucose; CD68, Cluster of
Differentiation; GFAP, Glial Fibrillary Acid Protein; GLUT, glucose transporter;
HLA-DR, Major Histocompatibility Complex Class II cell surface receptor; ICU,
intensive care unit; iNOS, inducible Nitric Oxide Synthase; MAP, mean arterial
pressure; OSF, organ systemic failure; PaO
2
, partial pressure of oxygen in
arterial blood; SAP, systolic arterial pressure; SAPS-II, simplified acute
physiology score; SaO
2
, saturation of oxygen in arterial blood; TNFa, tumor
necrosis factor alpha.
Polito et al.Critical Care 2011, 15:R131
http://ccforum.com/content/15/3/R131
Page 3 of 8
correlated with cumulative time of hypotension (rho =
0.45, P= 0.06 and rho = 0.38, P= 0.11).
Discussion
In patients dying of septic shock, hyperglycaemia was
associated with microglial apoptosis while neuronal
apoptosis was preferentially associated with endothelial
iNOS expression. We also found that hyperglycaemia
tended to be correlated with CD68 expression, which is
a marker of microglial activation. The postulated rela-
tionship between hyperglycaemia and microglial cell
apoptosis was supported by its absence of statistical cor-
relation with hypotension, hypoxemia or hypernatremia,
while it is known that hippocampus is highly vulnerable
to these factors. We also found that neuronal GLUT4
and microglial GLUT5 expressions were not correlated
with blood glucose level, suggesting impaired
downregulation.
These results are consistent with several experimental
studies. Discrepancy between microglial CD68 and
HLA-DR immunostaining has been previously observed
Table 2 Association of the area under the BG curve
above 2 g/l with clinical characteristics and
neuropathological findings
Spearman r(95%CI) P
SAPS-II at admission 0.34 (-0.17 to 0.71) 0.18
Knauss -0.21 (-0.63 to 0.30) 0.43
McCabe 0.05 (-0.44 to 0.52) 0.85
Neuropathological findings
Neuronal ischaemia 0.05 (-0.43 to 0.53) 0.82
Gliosis 0.15 (-0.36 to 0.59) 0.57
GFAP expression 0.11 (-0.39 to 0.56) 0.67
HLA-DR expression 0.06 (-0.43 to 0.53) 0.81
CD68 expression 0.44 (-0.05 to 0.76) 0.08
Beta-APP expression 0.61 (0.06 to 0.88) 0.03
Neuronal apoptosis 0.53 (0.07 to 0.81) 0.028
Microglial apoptosis 0.70 (0.33 to 0.88) 0.002
Glial TNFaexpression -0.04 (-0.51 to 0.45) 0.86
Endothelial iNOS expression 0.04 (-0.45 to 0.51) 0.87
Each neuropathological finding was score from 0 to 3 (see methods). beta-
APP, beta-amyloid precursor protein; CD68, Cluster of Differentiation; GFAP,
glial fibrillary acidic protein; HLA-DR, Major Histocompatibility Complex Class II
cell surface receptor; iNOS, inducible Nitric Oxide Synthase; TNFa, tumor
necrosis factor alpha.
Figure 1 Neuronal and microglial apoptosis in cerebral
amygdale. Case 7359, Cerebral amygdala. The back arrows show
two apoptotic neurons with darkly stained nucleus. The red arrow
shows an apoptotic microglial cell with a dark nucleus. The
cytoplasm of the apoptotic cells is also stained corresponding to
disintegration of nuclear chromatin into apoptotic bodies. (ISEL
×800).
Figure 2 Axonal damage in the hippocampal white matter.
Cortico-subcortical junction in the hippocampus. Black arrows show
axonal swellings in the white matter. These represent the
accumulation of the precursor of the beta-amyloid protein due to
alteration of the axonal flow. (APP imunostaining ABC/peroxidase/
DAB x25).
Figure 3 Hippocampal expression of GLUT4. Hippocampal
interneurons in CA1 and CA4 exhibit a homogeneous cytoplasmic
staining (arrow) with GLUT4 antibody (ABC/peroxidase/DAB, x40).
Polito et al.Critical Care 2011, 15:R131
http://ccforum.com/content/15/3/R131
Page 4 of 8
[25] and was ascribed to the fact that CD68 is a better
marker of activated microglia. Nitric oxide has been
extensively documented as pro-apoptotic factor, notably
in experimental sepsis [26-28]. In experimental models
of cerebral trauma or ischaemia, hyperglycaemia has
been linked to neuronal and glial cell injury through
various mechanisms including mitochondrial dysfunc-
tion, oxidative stress, inflammation and excitotoxicity
[29]. Although the similar mechanisms have been impli-
cated in sepsis associated encephalopathy, the potential
contribution of hyperglycaemia had not been elucidated.
It was recently shown that high glucose and LPS syner-
gistically induce microglial apoptosis by enhancing for-
mation of oxidative free radicals [13]. Interestingly, the
statistical correlation between neuronal and microglial
apoptosis suggest that they are interdependent phenom-
enon. It is established that neuronal function and survi-
val is intimately linked to both astroglial and microglial
cells [30]. Therefore, one may speculate that hypergly-
caemia induces microglial death that, synergistically with
endothelial iNOS, induces neuronal apoptosis, suggest-
ing a mechanistic sequence to account for sepsis asso-
ciated brain dysfunction. This model takes into account
the inflammatory [23] and metabolic (hyperglycaemia)
pathways that are a major pathophysiological process
and disturbance of septic shock, respectively. The
correlation between hyperglycaemia and axonal beta-
APP expression is consistent with that reported in
experimental brain ischaemia [31]. It suggests also
another scenario in which hyperglycaemia would first
induce axonal injury, then secondary degeneration of
microglia [31]. Interestingly, this finding proposes a new
pathophysiological mechanism for the long-term cogni-
tive decline in septic patients [32].
Thepresentstudyisthefirsttodescribetheneuro-
pathological consequences of hyperglycaemia in patients
who had died from septic shock. However, our study
has several limitations. First, one may argue that apopto-
sis was rather a post-mortem phenomenon. Although
this possibility cannot be ruled out, we have previously
shown that cell death did not correlate with time to
brain sampling [15]. Second, since BG levels were not
assessed continuously, it is likely that discrete hypogly-
caemic or hyperglycaemic events were not detected.
However, the rate of BG assessment was not different
between patients with and without hyperglycaemia or
prolonged hyperglycaemia. Third, it has been shown
that the capillary test does not provide an accurate mea-
surement of BG, notably overestimating it [33]. How-
ever, despite this flaw, capillary meter is used both in
clinical trials and in routine for titrating insulin therapy.
It has to be noted that microglial apoptosis was also
correlated with median BG. Fourth, we have limited our
investigation to the hippocampus as it is highly sensitive
to hemodynamic, hypoxic or metabolic insults but also
involved in ICU associated delirium pathophysiology
[34,35]. The impact of neuronal and microglial apoptosis
on hippocampal function cannot be obviously inferred
from these simple neuropathological observations. It
would be of interest to determine experimentally if
hyperglycaemia is associated with alterations in hippo-
campal electrophysiological function and with cognitive
impairments mediated by hippocampal structures. It has
been reported that high glucose level is associated with
occurrence of delirium in ICU patients [36]. Conversely,
it has been shown that infusion of glucose is a memory
enhancer in septic rats, suggesting that glucose tight
control, or at least hypoglicaemia, may affect hippocam-
pal functions [37].
While we have demonstrated an association between
hyperglycaemia and cell death in the brains of septic
shock patients, these data do not allow us to make any
Figure 4 Hippocampal expression of GLUT5.Inhippocampal
interneurons (CA1 and CA4), microglial cells are strongly stained
(arrows) whereas neurons are not labelled with GLUT5 antibody
(ABC/peroxidase/DAB, x25).
Table 3 Association of neuronal GLUT4 and microglial GLUT5 expression with glycaemia and cell apoptosis
Spearman r(95% CI) Neuronal GLUT4 expression PMicroglial GLUT5 expression P
Area under the BG curve > 2 g/l -0.006 (-0.49 to 0.48) 0.98 0.03 (-0.46 to 0.50) 0.91
Neuronal apoptosis -0.29 (-0.68 to 0.22) 0.25 -0.40 (-0.74 to 0.10) 0.11
Microglial apoptosis -0.002(-0.48 to 0.48) 0.99 -0.25 (-0.65 to 0.26) 0.33
Each neuropathological finding was score from 0 to 3 (see methods). BG, blood glucose; GLUT, glucose transporter.
Polito et al.Critical Care 2011, 15:R131
http://ccforum.com/content/15/3/R131
Page 5 of 8
definitive conclusions on hyperglycaemia as a causative
mechanism for cell death. Indeed, statistical correlations
between ante-mortem variables and post-mortem find-
ings do not prove a causal relationship. Demonstration
ofsuchalinkwouldrequireamoredetailedinvestiga-
tion of how glucose levels affect microglial cellular and
molecular function and the demonstration that glycemic
control reduces microglial apoptosis. Only experimental
studies could reasonably address these issues. Indeed,
post-mortem examination does not yield insight into the
proximal processes that precede apoptosis in humans.
This may explain that hyperglycaemia tended to be cor-
related with microglial activation (reflected by CD68
expression), which is prior to apoptosis. Assessment of
neuropathological effect of BG control would require
brain sampling in patients who had died from septic
shock and who had or not been treated with insulin
therapy: a task not so easily achievable. Our neuropatho-
logical samples were obtained before the widespread
implementation of glycemic control with intensive insu-
lin therapy in many critical care units. This is illustrated
by the fact that insulin was administered in a small pro-
portion of patients and was not targeted to normogly-
caemia. These observations prevented us from assessing
the neuropathological effect of insulin. Moreover, antici-
pating a neurological benefit from insulin therapy is pre-
mature. First of all, even if microglial cells play a major
role in host defence of the brain, and are involved in
neuroinflammatory and neurodegenerative processes,
their implication in sepsis related brain dysfunction is
not demonstrated [38]. It is unknown whether micro-
glial apoptosis is an adaptive, negligible or deleterious
phenomenon. Unlike the situation in neurons, interpre-
tation of positive ISEL staining in glial and microglial
cells is not straightforward. As ISEL is not absolutely
specific for double-stranded DNA breaks and can also
detect single-stranded breaks as observed in cell multi-
plication [39], positive staining may also reflect cell pro-
liferation. On the other hand, Petito and Roberts [40]
suggested that apoptotic death of reactive astrocytes
might be a physiological mechanism whereby the brain
removesanexcessnumberofastrocytesthathavepro-
liferated after certain types of brain injury. This can also
apply for microglia [41]. Second, cerebral glucose meta-
bolism is highly complex and its disturbances in sepsis
insufficiently elucidated. Therefore, neuronal sensitivity
to hypoglycaemia and hyperglycaemia might be deeply
changed in sepsis, making the effect of insulin on neuro-
nal metabolism unpredictable. We have found that neu-
ronal GLUT4 and microglial GLUT5 expression were
neither correlated with blood glucose levels or cell apop-
tosis. This does not rule out that glucose transporters
are involved in cell death process. For instance, it has
been experimentally shown that GLUT5 is implicated in
hyperglycaemia-related microglial cell death [13].
Furthermore, one may have expected that glucose trans-
porter expression would have been inversely proportion-
ate to blood glucose level [40,42,43]. Therefore, it is
conceivable that its absence of downregulation might
have increased intracellular glucose concentration and,
thereby, its toxicity. We acknowledge that absence of
correlation between microglial apoptosis and GLUT
expression does not rule out an alteration of GLUT
functioning, which in future studies could be indirectly
evaluated by measuring intracellular glucose load and
protein glycation. Additionally, it is biologically plausible
that hypoglycaemia potentially is far more harmful for
the brain than hyperglycaemia. It will be worthwhile to
assess the neuropathological correlates of hypoglycaemia
in patients who had died from septic shock. This would
require a greater proportion of patients who had devel-
oped hypoglycaemia than that observed in the present
study. It is interesting to note that iNOS has been
shown to decrease cerebral GLUT1 expression [44].
One may argue that the slight GLUT1 immunostaining
of neurons reflects a downregulation. Although expres-
sion of GLUT3 could not have been assessed for techni-
cal reasons, it has to be noted that alteration of GLUT3
cannot account for the relationship between hypergly-
caemia and apoptosis microglial cells as it is not
expressed by these cells.
The present study suggests a similar effect on micro-
glial GLUT5 expression. Other mechanisms could be
involved, especially perivascular edema that can compro-
mise substrate and oxygen delivery. Although we have
not specifically assessed this mechanism, it is established
that the BBB is altered in experimental sepsis but also
in septic shock patients [45].
Despite these limitations, our study suggests that
hyperglycaemia may contribute to the complex web of
abnormal signalling, which causes sepsis associated
brain dysfunction. Future studies should investigate the
mechanisms of hyperglycaemia related microglial apop-
tosis, particularly the impaired downregulation of
GLUT, and assess the neuropathological as well as neu-
rological effects of BG control by insulin therapy.
Conclusions
It appears likely that hemodynamic, inflammatory and
metabolic factors contribute to brain cell dysfunction
and death during septic shock, and may account for
sepsis associated brain dysfunction, which is associated
with increased mortality [46]. More research is needed
to understand the pathogenic significance of these fac-
tors and how they may be modulated to therapeutic
ends.
Polito et al.Critical Care 2011, 15:R131
http://ccforum.com/content/15/3/R131
Page 6 of 8
Key messages
In septic shock patients microglia is strongly
activated.
Hemodynamic, inflammatory and metabolic factors
contribute to brain cell dysfunction and death during
septic shock.
Hyperglycaemia is associated with microglial apop-
tosis while neuronal apoptosis is preferentially asso-
ciated with endothelial iNOS expression.
Hyperglycaemia may contribute to the complex
web of abnormal signaling which causes sepsis asso-
ciated brain dysfunction.
Abbreviations
iNOS: inducible Nitric Oxide Synthase; AUBGC: area under the BG curve;
Beta-APP: amyloid precursor protein A4; BG: blood glucose; GLUT: glucose
transporter; IQR: interquartile range; ISEL: in-situ end labelling; LPS:
lipopolysaccharide; SAPS II: Simplified Acute Physiologic Score II; SOFA:
Sequential Organ Failure Assessment; TNF-α: tumor necrosis factor α.
Author details
1
General Intensive Care Medicine, Raymond Poincaré Hospital (AP-HP),
University of Versailles Saint Quentin en Yvelines, 104 bd R. Poincaré,
Garches 92210, France.
2
Department of Pathology, Lariboisière Hospital (AP-
HP), University Denis Diderot-Paris 7, 2 rue Ambroise Paré, Paris 75010,
France.
3
Departement of Biostatistic and Medical Informatics, Saint-Louis
Hospital (APHP), University Denis Diderot-Paris 7, 47-83, boulevard de
lHôpital, Paris 75010, France.
4
Department of Anesthesiology and Critical
Care Medicine, Johns Hopkins University School of Medicine, 600 North
Wolfe Street, Baltimore, MD 21287, USA.
5
Department of Pathology,
Raymond Poincaré Hospital (AP-HP), University of Versailles Saint Quentin en
Yvelines, 104 bd R. Poincaré, Garches 92210, France.
6
HISTO, Human
Histopathology and Animal Models; Institut Pasteur; Département Infection
et Epidémiologie, 25 rue du Dr Roux, 75015 Paris.
Authorscontributions
AP conceived the study, acquired data and wrote the manuscript. JPB
helped in interpretation of the data and in drafting the manuscript. RP
participated in the design of the study, performed the statistical analysis and
helped to draft the manuscript. RS helped to draft the manuscript. SS
helped in acquisition of data and revising the manuscript, while RDS also
helped to revise the manuscript. CG, FC, FG, DA and VM helped to draft the
manuscript. GLG helped in acquisition and interpretation of data. TS
conceived the study, participated in the design of the study and helped to
draft the manuscript. All the authors read and approved the final
manuscript.
Competing interests
The authors declare that they have no competing interests.
Received: 15 February 2011 Revised: 3 April 2011
Accepted: 25 May 2011 Published: 25 May 2011
References
1. Brunkhorst FM, Engel C, Bloos F, Meier-Hellmann A, Ragaller M, Weiler N,
Moerer O, Gruendling M, Oppert M, Grond S, Olthoff D, Jaschinski U,
John S, Rossaint R, Welte T, Schaefer M, Kern P, Kuhnt E, Kiehntopf M,
Hartog C, Natanson C, Loeffler M, Reinhart K, German Competence Network
Sepsis (SepNet): Intensive insulin therapy and pentastarch resuscitation
in severe sepsis. N Engl J Med 2008, 358:125-139.
2. Van den Berghe G: How does blood glucose control with insulin save
lives in intensive care? J Clin Invest 2004, 114:1187-1195.
3. Dellinger RP, Levy MM, Carlet JM, Bion J, Parker MM, Jaeschke R, Reinhart K,
Angus DC, Brun-Buisson C, Beale R, Calandra T, Dhainaut JF, Gerlach H,
Harvey M, Marini JJ, Marshall J, Ranieri M, Ramsay G, Sevransky J,
Thompson BT, Townsend S, Vender JS, Zimmerman JL, Vincent JL, et al:
Surviving Sepsis Campaign: international guidelines for management of
severe sepsis and septic shock: 2008. Crit Care Med 2008, 36:296-327.
4. Langouche L, Vanhorebeek I, Van den Berghe G: Therapy insight: the
effect of tight glycemic control in acute illness. Nat Clin Pract Endocrinol
Metab 2007, 3:270-278.
5. Van den Berghe G, Wilmer A, Hermans G, Meersseman W, Wouters PJ,
Milants I, Van Wijngaerden E, Bobbaers H, Bouillon R: Intensive insulin
therapy in the medical ICU. N Engl J Med 2006, 354:449-461.
6. Van den Berghe G, Wouters P, Weekers F, Verwaest C, Bruyninckx F,
Schetz M, Vlasselaers D, Ferdinande P, Lauwers P, Bouillon R: Intensive
insulin therapy in the critically ill patients. N Engl J Med 2001,
345:1359-1367.
7. Van den Berghe G, Schoonheydt K, Becx P, Bruyninckx F, Wouters PJ:
Insulin therapy protects the central and peripheral nervous system of
intensive care patients. Neurology 2005, 64:1348-1353.
8. Hermans G, Wilmer A, Meersseman W, Milants I, Wouters PJ, Bobbaers H,
Bruyninckx F, Van den Berghe G: Impact of intensive insulin therapy on
neuromuscular complications and ventilator dependency in the medical
intensive care unit. Am J Respir Crit Care Med 2007, 175:480-489.
9. Bhalla A, Tilling K, Kolominsky-Rabas P, Heuschmann P, Megherbi SE,
Czlonkowska A, Kobayashi A, Mendel T, Giroud M, Rudd A, Wolfe C:
Variation in the management of acute physiological parameters after
ischaemic stroke: a European perspective. Eur J Neurol 2003, 10:25-33.
10. Capes SE, Hunt D, Malmberg K, Pathak P, Gerstein HC: Stress
hyperglycemia and prognosis of stroke in nondiabetic and diabetic
patients: a systematic overview. Stroke 2001, 32:2426-2432.
11. Yong M, Kaste M: Dynamic of hyperglycemia as a predictor of stroke
outcome in the ECASS-II trial. Stroke 2008, 39:2749-2755.
12. Wahl WL, Taddonio M, Maggio PM, Arbabi S, Hemmila MR: Mean glucose
values predict trauma patient mortality. J Trauma 2008, 65:42-47,
discussion 47-48.
13. Wang JY, Yang JM, Wang JY, Tao PL, Yang SN: Synergistic apoptosis
induced by bacterial endotoxin lipopolysaccharide and high glucose in
rat microglia. Neurosci Lett 2001, 304:177-180.
14. Barichello T, Fortunato JJ, Vitali AM, Feier G, Reinke A, Moreira JC,
Quevedo J, Dal-Pizzol F: Oxidative variables in the rat brain after sepsis
induced by cecal ligation and perforation. Crit Care Med 2006,
34:886-889.
15. Sharshar T, Gray F, Lorin de la Grandmaison G, Hopkinson NS, Ross E,
Dorandeu A, Orlikowski D, Raphael JC, Gajdos P, Annane D: Apoptosis of
neurons in cardiovascular autonomic centres triggered by inducible
nitric oxide synthase after death from septic shock. Lancet 2003,
362:1799-1805.
16. Douard V, Ferraris RP: Regulation of the fructose transporter GLUT5 in
health and disease. Am J Physiol Endocrinol Metab 2008, 295:E227-237.
17. Le Gall JR, Lemeshow S, Saulnier F: A new Simplified Acute Physiology
Score (SAPS II) based on a European/North American multicenter study.
JAMA 1993, 270:2957-2963.
18. Vincent JL, Moreno R, Takala J, Willatts S, De Mendonca A, Bruining H,
Reinhart CK, Suter PM, Thijs LG: The SOFA (Sepsis-related Organ Failure
Assessment) score to describe organ dysfunction/failure. On behalf of
the Working Group on Sepsis-Related Problems of the European Society
of Intensive Care Medicine. Intensive Care Med 1996, 22:707-710.
19. Van den Berghe G: First do no harm ... hypoglycemia or hyperglycemia?
Crit Care Med 2006, 34:2843-2844.
20. Nikonenko AG, Radenovic L, Andjus PR, Skibo GG: Structural features of
ischemic damage in the hippocampus. Anat Rec (Hoboken) 2009,
292:1914-1921.
21. Strokin M, Chechneva O, Reymann KG, Reiser G: Neuroprotection of rat
hippocampal slices exposed to oxygen-glucose deprivation by
enrichment with docosahexaenoic acid and by inhibition of hydrolysis
of docosahexaenoic acid-containing phospholipids by calcium
independent phospholipase A2. Neuroscience 2006, 140:547-553.
22. Poirier GF, Escourolle R: Manual of Basic Neuropathology. 3 edition.
Philadelphia, PA: WB Saunders Company; 1990.
23. Morrison RS, Kinoshita Y, Johnson MD, Guo W, Garden GA: p53-dependent
cell death signaling in neurons. Neurochem Res 2003, 28:15-27.
24. Freyer D, Manz R, Ziegenhorn A, Weih M, Angstwurm K, Docke WD,
Meisel A, Schumann RR, Schonfelder G, Dirnagl U, Weber JR: Cerebral
endothelial cells release TNF-alpha after stimulation with cell walls of
Streptococcus pneumoniae and regulate inducible nitric oxide synthase
Polito et al.Critical Care 2011, 15:R131
http://ccforum.com/content/15/3/R131
Page 7 of 8
and ICAM-1 expression via autocrine loops. J Immunol 1999,
163:4308-4314.
25. Lemstra AW, Groen int Woud JC, Hoozemans JJ, van Haastert ES,
Rozemuller AJ, Eikelenboom P, van Gool WA: Microglia activation in
sepsis: a case-control study. J Neuroinflammation 2007, 4:4.
26. Hara MR, Snyder SH: Cell signaling and neuronal death. Annu Rev
Pharmacol Toxicol 2007, 47:117-141.
27. Love S: Oxidative stress in brain ischemia. Brain Pathol 1999, 9:119-131.
28. Sastry PS, Rao KS: Apoptosis and the nervous system. J Neurochem 2000,
74:1-20.
29. Garg R, Chaudhuri A, Munschauer F, Dandona P: Hyperglycemia, insulin,
and acute ischemic stroke: a mechanistic justification for a trial of
insulin infusion therapy. Stroke 2006, 37:267-273.
30. Bessis A, Bechade C, Bernard D, Roumier A: Microglial control of neuronal
death and synaptic properties. Glia 2007, 55:233-238.
31. Lin B, Ginsberg MD, Busto R: Hyperglycemic but not normoglycemic
global ischemia induces marked early intraneuronal expression of beta-
amyloid precursor protein. Brain Res 2001, 888:107-116.
32. Iwashyna TJ, Ely EW, Smith DM, Langa KM: Long-term cognitive
impairment and functional disability among survivors of severe sepsis.
JAMA 2010, 304:1787-1794.
33. Kanji S, Buffie J, Hutton B, Bunting PS, Singh A, McDonald K, Fergusson D,
McIntyre LA, Hebert PC: Reliability of point-of-care testing for glucose
measurement in critically ill adults. Crit Care Med 2005, 33:2778-2785.
34. Fricchione GL, Nejad SH, Esses JA, Cummings TJ Jr, Querques J, Cassem NH,
Murray GB: Postoperative delirium. Am J Psychiatry 2008, 165:803-812.
35. Miczek KA, Covington HE, Nikulina EM Jr, Hammer RP: Aggression and
defeat: persistent effects on cocaine self-administration and gene
expression in peptidergic and aminergic mesocorticolimbic circuits.
Neurosci Biobehav Rev 2004, 27:787-802.
36. Heymann A, Sander M, Krahne D, Deja M, Weber-Carstens S, MacGuill M,
Kastrup M, Wernecke KD, Nachtigall I, Spies CD: Hyperactive delirium and
blood glucose control in critically ill patients. J Int Med Res 2007,
35:666-677.
37. Tuon L, Comim CM, Petronilho F, Barichello T, Izquierdo I, Quevedo J, Dal-
Pizzol F: Memory-enhancing treatments reverse the impairment of
inhibitory avoidance retention in sepsis-surviving rats. Crit Care 2008, 12:
R133.
38. Siami S, Annane D, Sharshar T: The encephalopathy in sepsis. Crit Care Clin
2008, 24:67-82, viii.
39. Ansari B, Coates PJ, Greenstein BD, Hall PA: In situ end-labelling detects
DNA strand breaks in apoptosis and other physiological and
pathological states. J Pathol 1993, 170:1-8.
40. Petito CK, Roberts B: Effect of postmortem interval on in situ end-
labeling of DNA oligonucleosomes. J Neuropathol Exp Neurol 1995,
54:761-765.
41. Gray F, Chretien F, Adle-Biassette H, Dorandeu A, Ereau T, Delisle MB,
Kopp N, Ironside JW, Vital C: Neuronal apoptosis in Creutzfeldt-Jakob
disease. J Neuropathol Exp Neurol 1999, 58:321-328.
42. Duelli R, Maurer MH, Staudt R, Heiland S, Duembgen L, Kuschinsky W:
Increased cerebral glucose utilization and decreased glucose transporter
Glut1 during chronic hyperglycemia in rat brain. Brain Res 2000,
858:338-347.
43. Koranyi L, Bourey RE, James D, Mueckler M, Fiedorek FT Jr, Permutt MA:
Glucose transporter gene expression in rat brain: Pretranslational
changes associated with chronic insulin-induced hypoglycemia, fasting,
and diabetes. Mol Cell Neurosci 1991, 2:244-252.
44. Takemori K, Ito H, Suzuki T: Effects of inducible nitric oxide synthase
inhibition on cerebral edema in severe hypertension. Acta Neurochir
Suppl 2000, 76:335-338.
45. Sharshar T, Carlier R, Bernard F, Guidoux C, Brouland JP, Nardi O, Lorin de la
Grandmaison G, Aboab J, Gray F, Menon D, Annane D: Brain lesions in
septic shock: a magnetic resonance imaging study. Intensive Care Med
2007, 33:798-806.
46. Eidelman LA, Putterman D, Putterman C, Sprung CL: The spectrum of
septic encephalopathy. Definitions, etiologies, and mortalities. JAMA
1996, 275:470-473.
doi:10.1186/cc10244
Cite this article as: Polito et al.: Hyperglycaemia and apoptosis of
microglial cells in human septic shock. Critical Care 2011 15:R131.
Submit your next manuscript to BioMed Central
and take full advantage of:
Convenient online submission
Thorough peer review
No space constraints or color figure charges
Immediate publication on acceptance
Inclusion in PubMed, CAS, Scopus and Google Scholar
Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Polito et al.Critical Care 2011, 15:R131
http://ccforum.com/content/15/3/R131
Page 8 of 8
... These data ascribe the hippocampal GLUT4 protein the same functional role it plays in the classic insulin-sensitive tissues [40], placing the transporter as a key player in the hippocampal metabolic glucose utilization. Additionally, we and others have already described GLUT4 in hippocampal neurons of the human brain, where it seems to play an important role under pathological conditions, as further discussed [74][75][76]. ...
... Low levels of GLUT5 have been detected in astrocytic and microglial cells of murine brains [20,35], and it has already been observed in human hippocampus [75]. However, GLUT5 is considered a fructose transporter, highly expressed in intestinal cells where it plays an important role in the absorption of this substrate [34,35]. ...
Article
Full-text available
The association of diabetes with cognitive dysfunction has at least 60 years of history, which started with the observation that children with type 1 diabetes mellitus (T1D), who had recurrent episodes of hypoglycemia and consequently low glucose supply to the brain, showed a deficit of cognitive capacity. Later, the growing incidence of type 2 diabetes mellitus (T2D) and dementia in aged populations revealed their high association, in which a reduced neuronal glucose supply has also been considered as a key mechanism, despite hyperglycemia. Here, we discuss the role of glucose in neuronal functioning/preservation, and how peripheral blood glucose accesses the neuronal intracellular compartment, including the exquisite glucose flux across the blood–brain barrier (BBB) and the complex network of glucose transporters, in dementia-related areas such as the hippocampus. In addition, insulin resistance-induced abnormalities in the hippocampus of obese/T2D patients, such as inflammatory stress, oxidative stress, and mitochondrial stress, increased generation of advanced glycated end products and BBB dysfunction, as well as their association with dementia/Alzheimer’s disease, are addressed. Finally, we discuss how these abnormalities are accompained by the reduction in the expression and translocation of the high capacity insulin-sensitive glucose transporter GLUT4 in hippocampal neurons, which leads to neurocytoglycopenia and eventually to cognitive dysfunction. This knowledge should further encourage investigations into the beneficial effects of promising therapeutic approaches which could improve central insulin sensitivity and GLUT4 expression, to fight diabetes-related cognitive dysfunctions.
... Ayrıca ensefalopatiye neden olabilecek diğer hastalıkları dışlamada ŞEKİL 1: Bilateral periyodik lateralizan epileptik deşarjlar (pnömokok menenjiti tanılı 5 aylık erkek hasta). 46 da yararlıdır. 37,38 Transkraniyal doppler sonografi(TCD) kısmi basınç ya da pH değişikliklerinde beynin perfüzyon basıncını sağlamaya çalışma yeteneğini kullanarak serebral vazomotor aktiviteyi izlemek için kullanılmaktadır. ...
... 45 Bunlara ek olarak ilaçlar veya metabolik değişiklikler septik hastalarda nörolojik bozulmaya katkıda bulunabileceği, hiper/hipogliseminin nöronal hücrelerde apoptoza neden olabileceği; renal replasman tedavisi sırasında sıvı elektrolit dengesizliğinin diyaliz desuquluibrum sendromu ve beyin ödemine neden olabileceği, antikoagülan ilaçların yanlış kullanımının intraserebral kanamaya neden olabileceği akılda tutulmalıdır. 46,47 ŞEKİL 2: Nonkonvulzif status epileptikus (akut viral meningoensefalit tanılı 11 yaş erkek hasta). 50 Prognoz: Septik ensefalopati geri dönüşümlü bir sendrom olmasına rağmen, bazı hastalarda sepsis düzeldikten sonra uzun süreli bilişsel ve psikiyatrik rahatsızlıklar da görülmüştür. ...
Chapter
ABS TRACT Infection-related encephalopathy is the most common cause of encephalopathy in childhood. Although the clinical manifestation of encephalopathy extends from personality disorders to delirium and coma, it can lead to irreversible consequences when diagnosed late. Clinical findings are very important in its diagnosis. Electroencephalography is supportive and can help predict prognosis. Among the causes leading to infectious encephalopathy, there are septic encephalopathy as well as central nervous system infections. Here, infectious diseases involving the brain parenchyma and meninges as the cause of infection-mediated encephalopathy and sepsis-related encephalopathy are discussed.
... A series of studies suggested that microglia was consistently activated in animal models and patients of sepsis (11,37). For example, the increasing microglia with amoeboid shape was observed in the hippocampus and cortex of the patients who died of septic shock (38,39). ...
Article
Full-text available
Sepsis-associated encephalopathy (SAE), the most popular cause of coma in the intensive care unit (ICU), is the diffuse cerebral damage caused by the septic challenge. SAE is closely related to high mortality and extended cognitive impairment in patients in septic shock. At present, many studies have demonstrated that SAE might be mainly associated with blood–brain barrier damage, abnormal neurotransmitter secretion, oxidative stress, and neuroimmune dysfunction. Nevertheless, the precise mechanism which initiates SAE and contributes to the long-term cognitive impairment remains largely unknown. Recently, a growing body of evidence has indicated that there is close crosstalk between SAE and peripheral immunity. The excessive migration of peripheral immune cells to the brain, the activation of glia, and resulting dysfunction of the central immune system are the main causes of septic nerve damage. This study reviews the update on the pathogenesis of septic encephalopathy, focusing on the over-activation of immune cells in the central nervous system (CNS) and the “neurocentral–endocrine–immune” networks in the development of SAE, aiming to further understand the potential mechanism of SAE and provide new targets for diagnosis and management of septic complications.
... In an effort to deepen our understanding of the neuroinflammatory mechanisms relevant to sepsis-associated encephalopathy, Moraes et al. offer a review, published in Pharmaceuticals, focusing on the activation of microglial cells by danger-associated molecular patterns and pathogen-associated molecular patterns and their consequences in the context of brain dysfunction during sepsis. These authors relate findings of several postmortem studies showing increased cerebral expression of CD68, which can be taken as an indicator of microglial activation, in patients who had succumbed to sepsis [129][130][131]. Furthermore, Moraes et al. hypothesize that "IL-1beta derived from activated microglia is responsible for the synaptic deficits observed in sepsis" [132] (p. ...
Article
Full-text available
In spite of the brain-protecting tissues of the skull, meninges, and blood-brain barrier, some forms of injury to or infection of the CNS can give rise to cerebral cytokine production and action and result in drastic changes in brain function and behavior. Interestingly, peripheral infection-induced systemic inflammation can also be accompanied by increased cerebral cytokine production. Furthermore, it has been recently proposed that some forms of psychological stress may have similar CNS effects. Different conditions of cerebral cytokine production and action will be reviewed here against the background of neuroinflammation. Within this context, it is important to both deepen our understanding along already taken paths as well as to explore new ways in which neural functioning can be modified by cytokines. This, in turn, should enable us to put forward different modes of cerebral cytokine production and action in relation to distinct forms of neuroinflammation.
... Similarly, in a postmortem case-control study of patients with delirium, there was an increase in microglial markers CD68 and HLA-DR compared to age-matched controls, suggesting that microglia activation could be related to delirium [61]. In a prospective study of 17 patients who died from septic shock, hippocampal tissue was assessed for a neuropathological study that observed increased microglia activation and apoptosis in septic patients with hyperglycemia [62]. ...
Article
Full-text available
Frequently underestimated, encephalopathy or delirium are common neurological manifestations associated with sepsis. Brain dysfunction occurs in up to 80% of cases and is directly associated with increased mortality and long-term neurocognitive consequences. Although the central nervous system (CNS) has been classically viewed as an immune-privileged system, neuroinflammation is emerging as a central mechanism of brain dysfunction in sepsis. Microglial cells are major players in this setting. Here, we aimed to discuss the current knowledge on how the brain is affected by peripheral immune activation in sepsis and the role of microglia in these processes. This review focused on the molecular pathways of microglial activity in sepsis, its regulatory mechanisms, and their interaction with other CNS cells, especially with neuronal cells and circuits.
... Prevention of hyperglycemia is critical to neuroprotection as microglial activation, loss of astrocytes and neuronal damage in the hippocampus and frontal cortex were more prevalent in hyperglycemic than in normoglycemic critically ill patients [6]. Furthermore, microglial apoptosis was found in patients who died from a septic shock and had suffered from hyperglycemia during their intensive care unit stay [7]. ...
Article
Full-text available
BACKGROUND AND PURPOSE: Hyperglycemia can lead to an increased rate of apoptosis of microglial cells and to damaged neurons. The relation between hyperglycemia and cerebrovascular markers on MRI is unknown. Our aim was to study the association between intraoperative hyperglycemia and cerebrovascular markers. METHODS: In this further analysis of a subgroup investigation of the BIOCOG study, 65 older non-demented patients (median 72 years) were studied who underwent elective surgery of ≥ 60 minutes. Intraoperative blood glucose maximum was determined retrospectively in each patient. In these patients, preoperatively and at 3 months follow-up a MRI scan was performed and white matter hyperintensity (WMH) volume and shape, infarcts, and perfusion parameters were determined. Multivariable logistic regression analyses were performed to determine associations between preoperative cerebrovascular markers and occurrence of intraoperative hyperglycemia. Linear regression analyses were performed to assess the relation between intraoperative hyperglycemia and pre- to postoperative changes in WMH volume. Associations between intraoperative hyperglycemia and postoperative WMH volume at 3 months follow-up were also assessed by linear regression analyses. RESULTS: Eighteen patients showed intraoperative hyperglycemia (glucose maximum ≥ 150 mg/dL). A preoperative more smooth shape of periventricular and confluent WMH was related to the occurrence of intraoperative hyperglycemia [convexity: OR 33.318 (95 % CI (1.002 - 1107.950); p = 0.050]. Other preoperative cerebrovascular markers were not related to the occurrence of intraoperative hyperglycemia. Intraoperative hyperglycemia showed no relation with pre- to postoperative changes in WMH volume nor with postoperative WMH volume at 3 months follow-up. CONCLUSIONS: We found that a preoperative more smooth shape of periventricular and confluent WMH was related to the occurrence of intraoperative hyperglycemia. These findings may suggest that a similar underlying mechanism leads to a certain pattern of vascular brain abnormalities and an increased risk of hyperglycemia.
Chapter
This chapter lays out the elementary principles of fractal geometry underpinning much of the rest of this book. It assumes a minimal mathematical background, defines the key principles and terms in context, and outlines the basics of a fractal analysis method known as box counting and how it is used to perform fractal, lacunarity, and multifractal analyses. As a standalone reference, this chapter grounds the reader to be able to understand, evaluate, and apply essential methods to appreciate and heal the exquisitely detailed fractal geometry of the brain.
Article
Full-text available
To date no biomarker has been identified bringing together perfect sensitivity and specificity to discriminate between inflammation and infections. Since the 1930s new markers of tissue damage and endothelial damage have been identified but which are incapable of identifying infections in every clinical setting to enable initiation of early antibiotic treatment. In this review the most important classical biomarkers and upcoming new PCR-based approaches are addressed. These markers are highlighted with respect to special clinical settings and to control the success of antibiotic treatment. The issue of discrimination between inflammation and infection is not yet solved. Based on one single biomarker it is impossible to decide whether infection is the reason for the patient’s worsening condition but the combination of biomarkers or the integration of new biomarkers may be a meaningful supplement. The measurement of different biomarkers of infection or inflammation is part of the routine in critical care and will be essential in the future.
Article
Full-text available
Sepsis, a leading cause of morbidity and mortality, is caused by a deregulated host response to pathogens, and subsequent life-threatening organ dysfunctions. All major systems, including the cardiovascular, respiratory, renal, hepatic, hematological, and the neurological system may be affected by sepsis. Sepsis associated neurological dysfunction is triggered by multiple factors including neuro-inflammation, excitotoxicity, and ischemia. Ischemia results from reduced cerebral blood flow, caused by extreme variations of blood pressure, occlusion of cerebral vessels, or more subtle defects of the microcirculation. International guidelines comprehensively describe the initial hemodynamic management of sepsis, revolving around the normalization of systemic hemodynamics and of arterial lactate. By contrast, the management of sepsis patients suffering from brain dysfunction is poorly detailed, the only salient point being mentioned is that sedation and analgesia should be optimized. However, sepsis and the hemodynamic consequences thereof as well as vasopressors may have severe untoward neurological consequences. The current review describes the general neurological complications, as well as the consequences of vasopressor therapy on the brain and its circulation and addresses methods for cerebral monitoring during sepsis.
Article
Full-text available
Hyperglycemia and insulin resistance are common in critically ill patients, even if they have not previously had diabetes. Whether the normalization of blood glucose levels with insulin therapy improves the prognosis for such patients is not known. We performed a prospective, randomized, controlled study involving adults admitted to our surgical intensive care unit who were receiving mechanical ventilation. On admission, patients were randomly assigned to receive intensive insulin therapy (maintenance of blood glucose at a level between 80 and 110 mg per deciliter [4.4 and 6.1 mmol per liter]) or conventional treatment (infusion of insulin only if the blood glucose level exceeded 215 mg per deciliter [11.9 mmol per liter] and maintenance of glucose at a level between 180 and 200 mg per deciliter [10.0 and 11.1 mmol per liter]). At 12 months, with a total of 1548 patients enrolled, intensive insulin therapy reduced mortality during intensive care from 8.0 percent with conventional treatment to 4.6 percent (P<0.04, with adjustment for sequential analyses). The benefit of intensive insulin therapy was attributable to its effect on mortality among patients who remained in the intensive care unit for more than five days (20.2 percent with conventional treatment, as compared with 10.6 percent with intensive insulin therapy, P=0.005). The greatest reduction in mortality involved deaths due to multiple-organ failure with a proven septic focus. Intensive insulin therapy also reduced overall in-hospital mortality by 34 percent, bloodstream infections by 46 percent, acute renal failure requiring dialysis or hemofiltration by 41 percent, the median number of red-cell transfusions by 50 percent, and critical-illness polyneuropathy by 44 percent, and patients receiving intensive therapy were less likely to require prolonged mechanical ventilation and intensive care. Intensive insulin therapy to maintain blood glucose at or below 110 mg per deciliter reduces morbidity and mortality among critically ill patients in the surgical intensive care unit.
Article
Full-text available
Intensive insulin therapy reduces morbidity and mortality in patients in surgical intensive care units (ICUs), but its role in patients in medical ICUs is unknown. In a prospective, randomized, controlled study of adult patients admitted to our medical ICU, we studied patients who were considered to need intensive care for at least three days. On admission, patients were randomly assigned to strict normalization of blood glucose levels (80 to 110 mg per deciliter [4.4 to 6.1 mmol per liter]) with the use of insulin infusion or to conventional therapy (insulin administered when the blood glucose level exceeded 215 mg per deciliter [12 mmol per liter], with the infusion tapered when the level fell below 180 mg per deciliter [10 mmol per liter]). There was a history of diabetes in 16.9 percent of the patients. In the intention-to-treat analysis of 1200 patients, intensive insulin therapy reduced blood glucose levels but did not significantly reduce in-hospital mortality (40.0 percent in the conventional-treatment group vs. 37.3 percent in the intensive-treatment group, P=0.33). However, morbidity was significantly reduced by the prevention of newly acquired kidney injury, accelerated weaning from mechanical ventilation, and accelerated discharge from the ICU and the hospital. Although length of stay in the ICU could not be predicted on admission, among 433 patients who stayed in the ICU for less than three days, mortality was greater among those receiving intensive insulin therapy. In contrast, among 767 patients who stayed in the ICU for three or more days, in-hospital mortality in the 386 who received intensive insulin therapy was reduced from 52.5 to 43.0 percent (P=0.009) and morbidity was also reduced. Intensive insulin therapy significantly reduced morbidity but not mortality among all patients in the medical ICU. Although the risk of subsequent death and disease was reduced in patients treated for three or more days, these patients could not be identified before therapy. Further studies are needed to confirm these preliminary data. (ClinicalTrials.gov number, NCT00115479.)
Article
An abstract is unavailable. This article is available as HTML full text and PDF.
Article
The present study investigates whether bacterial lipopolysaccharide (LPS) in the presence of increased levels of glucose induced synergistic cytotoxicity in primary cultured microglia. Significant cytotoxicity was only observed while the concentrations of LPS were increased to 10 μg/ml. D-glucose concentration-dependently (25–125 mM) generated cytotoxicity. Synergistic apoptosis of microglia was seen by LPS in the presence of increased levels of D-glucose. This synergistic cytotoxicity was attenuated by the use of superoxide dimutase and catalase, suggesting the involvement of oxidative free radicals. Collectively, the present results suggest that increased ambient levels of glucose rendered microglia vulnerable to LPS insults, and led to a synergistic apoptosis. The findings here may be important in certain patho-physiological implications in which hyperglycemia exacerbated the ambient functions contributed by microglia, and may provide new insight into a novel therapeutic intervention.
Article
Whereas acute hyperglycemia has been shown to result in an unchanged local cerebral glucose utilization (LCGU) the changes of LCGU during chronic hyperglycemia are a matter of dispute. The present study had three aims: (1) To compare the effects of acute and chronic hyperglycemia on LCGU and to investigate in vivo the lactate level as a potential indicator of glycolytic flux. (2) To investigate local changes in brain Glut1 and/or Glut3 glucose transporter densities during chronic hyperglycemia. (3) To analyze the relationship between LCGU and local Glut densities during chronic hyperglycemia. To induce chronic hyperglycemia in rats steptozotocin was given i.p. and experiments were performed 3 weeks later. LCGU was measured by the method and intraparenchymal lactate concentration by MR-spectroscopy. Local densities of the glucose transport proteins were determined by immunoautoradiographic methods. During chronic hyperglycemia weighted average of LCGU increased by 13.9% whereas it remained unchanged during acute hyperglycemia. The cerebral lactate/choline ratio was increased by 143% during chronic hyperglycemia. The average density of glucose transporters Glut1 decreased by 7.5%. Local densities of Glut1 were decreased in 12 of 28 brain structures. Glut3 remained unchanged. Positive correlations were found between LCGU and local Glut densities during control conditions and during chronic hyperglycemia. It was concluded that (1) Chronic, but not acute hyperglycemia is followed by an increased LCGU. (2) The capacity to transport glucose is decreased during chronic hyperglycemia. (3) Increased LCGU and decreased densities of Glut1 are matched on a local level.
Article
Apoptosis is now recognized as a normal feature in the development of the nervous system and may also play a role in neurodegenerative diseases and aging. This phenomenon has been investigated intensively during the last 6-7 years, and the progress made in this field is reviewed here. Besides a few in vivo studies, a variety of neuronal preparations from various parts of the brain, the majority of which were primary cultures, and some cell lines have been investigated. Several apoptosis-inducing agents have been identified, and these include lack of neurotrophic support, neurotransmitters, neurotoxicants, modulators of protein phosphorylation and calcium homeostasis, DNA-damaging agents, oxidative stress, nitric oxide, and ceramides. The precise signaling cascade is not well established, and there are lacunae in many suggested pathways. However, it appears certain that the Bcl family of proteins is involved in the apoptotic pathway, and these proteins in turn affect the processing of interleukin-1beta converting enzyme (ICE)/caspases. The available evidence suggests that there may be several apoptotic pathways that may depend on the cell type and the inducing agent, and most of the pathways may converge at the ICE/caspases step.