Introduction
Sepsis is defined as a life-threatening organ dysfunction caused by dysregulated host responses to an infection (
1- Singer M.
- Deutschman C.S.
- Seymour C.W.
- Shankar-Hari M.
- Annane D.
- Bauer M.
- Bellomo R.
- Bernard G.R.
- Chiche J.D.
- Coopersmith C.M.
- Hotchkiss R.S.
- Levy M.M.
- Marshall J.C.
- Martin G.S.
- Opal S.M.
- et al.
The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3).
). At admission, up to 71% of septic patients develop potentially irreversible acute cerebral dysfunction (
2Progress in clinical neurosciences: sepsis-associated encephalopathy: evolving concepts.
,
3Long-term cerebral consequences of sepsis.
), a condition caused by systemic inflammation without brain infection and clinically characterized by slowing of mental processes, impaired attention, disorientation, delirium, or coma (
4- Semmler A.
- Hermann S.
- Mormann F.
- Weberpals M.
- Paxian S.A.
- Okulla T.
- Schäfers M.
- Kummer M.P.
- Klockgether T.
- Heneka M.T.
Sepsis causes neuroinflammation and concomitant decrease of cerebral metabolism.
). Recent studies have shown that long-lasting consequences following sepsis recovery often include brain disorders (
5- Winters B.D.
- Eberlein M.
- Leung J.
- Needham D.M.
- Pronovost P.J.
- Sevransky J.E.
Long-term mortality and quality of life in sepsis: a systematic review.
). Even after full recovery, animals subjected to sepsis induced by cecal ligation and perforation (CLP)
2The abbreviations used are: CLP
cecal ligation and perforation
RAGE
receptor for advanced glycation end product
sRAGE
soluble form of RAGE
RAGE
abRAGE antibody
AGE
advanced glycation end product
Aβ
amyloid-β
AβPP
amyloid precursor protein
BBB
blood–brain barrier
GFAP
glial fibrillary acidic protein
mTOR
mechanistic target of rapamycin
nNOS
neuronal nitric-oxide synthase
AD
Alzheimer's disease
ANOVA
analysis of variance
CML
Nɛ-(carboxymethyl)lysine
b.w.
body weight
TBI
traumatic brain injury.
demonstrated significant difficulties in performing behavioral tasks, indicating cognitive deficits (
6- Barichello T.
- Martins M.R.
- Reinke A.
- Feier G.
- Ritter C.
- Quevedo J.
- Dal-Pizzol F.
Cognitive impairment in sepsis survivors from cecal ligation and perforation.
). In recovered patients, persistent deficits in functional abilities and general quality of life are observed (
7- Yende S.
- Austin S.
- Rhodes A.
- Finfer S.
- Opal S.
- Thompson T.
- Bozza F.A.
- LaRosa S.P.
- Ranieri V.M.
- Angus D.C.
Long-term quality of life among survivors of severe sepsis: analyses of two international trials.
), which may be associated with long-lasting impairment in cognitive capacities associated with memory and executive function (
8- Iwashyna T.J.
- Ely E.W.
- Smith D.M.
- Langa K.M.
Long-term cognitive impairment and functional disability among survivors of severe sepsis.
).
This long-term impairment in brain function was suggested to result from neurodegenerative or ischemic mechanisms triggered by systemic inflammation (
9Cognitive decline after sepsis.
). Peripherally produced cytokines can enter the central nervous system (CNS), as sepsis is able to induce transient disruption of the blood-brain barrier (BBB) (
10The blood-brain barrier in systemic inflammation.
,
11- Dal-Pizzol F.
- Rojas H.A.
- dos Santos E.M.
- Vuolo F.
- Constantino L.
- Feier G.
- Pasquali M.
- Comim C.M.
- Petronilho F.
- Gelain D.P.
- Quevedo J.
- Moreira J.C.
- Ritter C.
Matrix metalloproteinase-2 and metalloproteinase-9 activities are associated with blood-brain barrier dysfunction in an animal model of severe sepsis.
). This in turn leads to microglia/astrocyte activation and local production of pro-inflammatory mediators and reactive species (
12- Michels M.
- Steckert A.V.
- Quevedo J.
- Barichello T.
- Dal-Pizzol F.
Mechanisms of long-term cognitive dysfunction of sepsis: from blood-borne leukocytes to glial cells.
,
13- Michels M.
- Vieira A.S.
- Vuolo F.
- Zapelini H.G.
- Mendonça B.
- Mina F.
- Dominguini D.
- Steckert A.
- Schuck P.F.
- Quevedo J.
- Petronilho F.
- Dal-Pizzol F.
The role of microglia activation in the development of sepsis-induced long-term cognitive impairment.
). However, details of the molecular cascades linking systemic inflammation to neuroinflammation and brain dysfunction still need to be better understood. Comprehension of these mechanisms in detail may reveal valuable information that can be used as a basis to develop new strategies to treat sepsis co-morbidities. In addition, unveiling new details on the molecular events linking systemic inflammation to brain dysfunction may also uncover new insights to the understanding of the onset of neurodegeneration itself (
14- Cunningham C.
- Hennessy E.
Co-morbidity and systemic inflammation as drivers of cognitive decline: new experimental models adopting a broader paradigm in dementia research.
).
Neurodegenerative processes may evolve over the course of many years, and the diagnosis is generally performed only in advanced or late stages, when brain function is impaired due to significant neuronal loss. Neurodegeneration is characterized by progressive neuronal death associated with the accumulation of misfolded, aberrant forms of cellular proteins or peptides with neurotoxic activity. In Alzheimer's disease (AD), neuronal death occurs concomitant to progressive formation of neurofibrillary tangles and amyloid (senile) plaques. Neurofibrillary tangles are formed due to aberrant hyperphosphorylation of the microtubule-stabilizing protein Tau, whereas amyloid plaques originate by hydrophobic aggregates of misfolded amyloid-β peptide (Aβ). Hyperphosphorylation of Tau occurs in at least 22 other brain conditions, such as amyotrophic lateral sclerosis, Down's syndrome, and prion diseases (
15- Spires-Jones T.L.
- Stoothoff W.H.
- de Calignon A.
- Jones P.B.
- Hyman B.T.
Tau pathophysiology in neurodegeneration: a tangled issue.
), and because disruption of Tau homeostasis is associated with cognitive deficits (
16- Gendron T.F.
- Petrucelli L.
The role of Tau in neurodegeneration.
), modulation of Tau phosphorylation by pathogenic processes may also be associated with the onset of neurodegenerative processes. Similarly, cleavage of the amyloid precursor protein (AβPP) by different secretases may generate small peptides, including a variety of Aβ peptides (
17- Multhaup G.
- Huber O.
- Buée L.
- Galas M.C.
Amyloid precursor protein (APP) metabolites APP intracellular fragment (AICD), Aβ42, and tau in nuclear roles.
). Necrotic and apoptotic neuronal death is induced by extracellular Aβ both through activation of death receptors in neurons and induction of microglial pro-inflammatory activation and astrocytosis (
18- Nalivaeva N.N.
- Turner A.J.
The amyloid precursor protein: a biochemical enigma in brain development, function and disease.
). Pro-inflammatory activation, in turn, further induces Aβ aggregation due to amino acid oxidation and peptide destabilization (
19- Zuo L.
- Hemmelgarn B.T.
- Chuang C.C.
- Best T.M.
The role of oxidative stress-induced epigenetic alterations in amyloid-β production in Alzheimer's disease.
).
The receptor for advanced glycation end products (RAGE) is a multiligand pattern-recognition receptor, belonging to the immunoglobulin superfamily of proteins (
20- Creagh-Brown B.C.
- Quinlan G.J.
- Evans T.W.
- Burke-Gaffney A.
The RAGE axis in systemic inflammation, acute lung injury and myocardial dysfunction: an important therapeutic target?.
). Although it was first described to be activated by advanced glycation end products (AGEs), RAGE can interact with a number of different classes of agonists, including S100 family proteins (
e.g. S100B), high-mobility group box protein 1 (HMGB1), 70-kDa heat-shock protein (HSP70), and
Nɛ-(carboxymethyl)lysine (CML) (
21- Batkulwar K.B.
- Bansode S.B.
- Patil G.V.
- Godbole R.K.
- Kazi R.S.
- Chinnathambi S.
- Shanmugam D.
- Kulkarni M.J.
Investigation of phosphoproteome in RAGE signaling.
), among others. RAGE is expressed only by lungs and endothelial cells in a constitutive fashion and is generally repressed in other tissues; however, an increase in the concentration of circulating RAGE ligands induces the expression of this receptor in most cell types (
20- Creagh-Brown B.C.
- Quinlan G.J.
- Evans T.W.
- Burke-Gaffney A.
The RAGE axis in systemic inflammation, acute lung injury and myocardial dysfunction: an important therapeutic target?.
). RAGE activation triggers different signaling cascades for immune responses, including the ERK1/2-dependent activation of NF-κB and consequent transcriptional activation of pro-inflammatory genes (
22- Tóbon-Velasco J.C.
- Cuevas E.
- Torres-Ramos M.A.
Receptor for AGEs (RAGE) as mediator of NF-κB pathway activation in neuroinflammation and oxidative stress.
), whereas RAGE inhibition was demonstrated to have a protective role against sepsis and LPS-induced endotoxemia (
23- Lutterloh E.C.
- Opal S.M.
- Pittman D.D.
- Keith Jr, J.C.
- Tan X.Y.
- Clancy B.M.
- Palmer H.
- Milarski K.
- Sun Y.
- Palardy J.E.
- Parejo N.A.
- Kessimian N.
Inhibition of the RAGE products increases survival in experimental models of severe sepsis and systemic infection.
,
24- van Zoelen M.A.
- Schmidt A.M.
- Florquin S.
- Meijers J.C.
- de Beer R.
- de Vos A.F.
- Nawroth P.P.
- Bierhaus A.
- van der Poll T.
Receptor for advanced glycation end products facilitates host defense during Escherichia coli-induced abdominal sepsis in mice.
25- van Zoelen M.A.
- van der Poll T.
Targeting RAGE in sepsis.
). In the CNS, RAGE expression has been linked to neuroinflammation, Aβ influx through the BBB, and neurodegeneration associated with AD, among other conditions (
26- Srikanth V.
- Maczurek A.
- Phan T.
- Steele M.
- Westcott B.
- Juskiw D.
- Münch G.
Advanced glycation end products and their receptor RAGE in Alzheimer's disease.
,
27- Li X.H.
- Lv B.L.
- Xie J.Z.
- Liu J.
- Zhou X.W.
- Wang J.Z.
AGEs induce Alzheimer-like tau pathology and memory deficit via RAGE-mediated GSK-3 activation.
). It was previously demonstrated that HMGB1, a RAGE ligand, mediates cognitive dysfunction in sepsis survivors (
28- Chavan S.S.
- Huerta P.T.
- Robbiati S.
- Valdes-Ferrer S.I.
- Ochani M.
- Dancho M.
- Frankfurt M.
- Volpe B.T.
- Tracey K.J.
- Diamond B.
HMGB1 mediates cognitive impairment in sepsis survivors.
) and that RAGE is increased in the brain of rats 30 days after sepsis induction by CLP (
29- Dal-Pizzol F.
- Pasquali M.
- Quevedo J.
- Gelain D.P.
- Moreira J.C.
Is there a role for high mobility group box 1 and the receptor for advanced glycation end products in the genesis of long-term cognitive impairment in sepsis survivors?.
,
30- Schwalm M.T.
- Pasquali M.
- Miguel S.P.
- Dos Santos J.P.
- Vuolo F.
- Comim C.M.
- Petronilho F.
- Quevedo J.
- Gelain D.P.
- Moreira J.C.
- Ritter C.
- Dal-Pizzol F.
Acute brain inflammation and oxidative damage are related to long-term cognitive deficits and markers of neurodegeneration in sepsis-survivor rats.
). Following the observation that Aβ binds and activates RAGE (
31- Deane R.
- Singh I.
- Sagare A.P.
- Bell R.D.
- Ross N.T.
- LaRue B.
- Love R.
- Perry S.
- Paquette N.
- Deane R.J.
- Thiyagarajan M.
- Zarcone T.
- Fritz G.
- Friedman A.E.
- Miller B.L.
- Zlokovic B.V.
A multimodal RAGE-specific inhibitor reduces amyloid β-mediated brain disorder in a mouse model of Alzheimer disease.
), RAGE was identified as an important link in the intertwined signaling of inflammatory, amyloidogenic, and pro-apoptotic cascades during the progression of AD, as it maintains a chronic pro-inflammatory state via Aβ-dependent stimulation of microglia (
32Microglial Aβ receptors in Alzheimer's disease.
). Nonetheless, a possible role of RAGE in other CNS disorders, including those associated with pro-inflammatory states, has been relatively neglected.
This study was performed to evaluate whether RAGE is associated with the changes in CNS homeostasis that arise after the recovery of the acute pro-inflammatory phase of sepsis. Therefore, the levels of pro-inflammatory markers, Aβ, phosphorylated Tau, and RAGE-associated molecules (including RAGE, several RAGE ligands, and intracellular downstream targets) were monitored in serum and brain from 24 h to 30 days after CLP surgery. The serum was evaluated for peripheral inflammatory markers and RAGE ligands, and the hippocampus and prefrontal cortex were evaluated because these structures are directly associated with cognitive dysfunctions observed in this rat model (
6- Barichello T.
- Martins M.R.
- Reinke A.
- Feier G.
- Ritter C.
- Quevedo J.
- Dal-Pizzol F.
Cognitive impairment in sepsis survivors from cecal ligation and perforation.
,
33- Barichello T.
- Martins M.R.
- Reinke A.
- Constantino L.S.
- Machado R.A.
- Valvassori S.S.
- Moreira J.C.
- Quevedo J.
- Dal-Pizzol F.
Behavioral deficits in sepsis-surviving rats induced by cecal ligation and perforation.
). RAGE signaling was blocked by immune neutralization in the hippocampus between 15 and 19 days after CLP, and the cognitive function (inhibitory avoidance and object recognition tasks), neuroinflammatory (RAGE, Iba-1, and GFAP), and neurodegenerative markers (Aβ and phosphorylated Tau) were evaluated 30–31 days after CLP. The results presented here indicate the following: (i) RAGE signaling increases as acute pro-inflammatory markers decrease over the 30 days following CLP, in serum, and CNS; and (ii) blocking of RAGE in the hippocampus inhibits neuroinflammatory and neurodegenerative markers in this brain region, as well as cognitive deficits that are observed 30 days after CLP. Overall these data suggest that RAGE signaling in the CNS exerts an important role in the progressive impairment of brain function that arises after the recovery from the acute phase of sepsis, and it may be involved in the long-term development of brain dysfunction and neurodegeneration triggered by episodes of acute systemic inflammation, including polymicrobial sepsis.
Discussion
In this study, animals surviving sepsis presented brain alterations commonly associated with the onset of neurodegenerative processes, and RAGE was demonstrated to play a key role in the progression of these changes. Sepsis enhances transcription of pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6 (
4- Semmler A.
- Hermann S.
- Mormann F.
- Weberpals M.
- Paxian S.A.
- Okulla T.
- Schäfers M.
- Kummer M.P.
- Klockgether T.
- Heneka M.T.
Sepsis causes neuroinflammation and concomitant decrease of cerebral metabolism.
), which affect the arrangement of tight junctions in BBB endothelial cells (
34- Gutierrez E.G.
- Banks W.A.
- Kastin A.J.
Murine tumor necrosis factor α is transported from blood to brain in the mouse.
,
35- Yarlagadda A.
- Alfson E.
- Clayton A.H.
The blood brain barrier and the role of cytokines in neuropsychiatry.
36- Pan W.
- Stone K.P.
- Hsuchou H.
- Manda V.K.
- Zhang Y.
- Kastin A.J.
Cytokine signaling modulates blood-brain barrier function.
). This increases BBB permeability, contributing to neuroinflammation and cell death (
11- Dal-Pizzol F.
- Rojas H.A.
- dos Santos E.M.
- Vuolo F.
- Constantino L.
- Feier G.
- Pasquali M.
- Comim C.M.
- Petronilho F.
- Gelain D.P.
- Quevedo J.
- Moreira J.C.
- Ritter C.
Matrix metalloproteinase-2 and metalloproteinase-9 activities are associated with blood-brain barrier dysfunction in an animal model of severe sepsis.
). The present model of polymicrobial sepsis induced by CLP requires the use of antibiotics to maintain survival around 40%. However, the antibiotic therapy, as observed here and in previous works, does not prevent the progression of sepsis, although it significantly enhances survival (
37- Dal-Pizzol F.
- Di Leone L.P.
- Ritter C.
- Martins M.R.
- Reinke A.
- Pens Gelain D.
- Zanotto-Filho A.
- de Souza L.F.
- Andrades M.
- Barbeiro D.F.
- Bernard E.A.
- Cammarota M.
- Bevilaqua L.R.
- Soriano F.G.
- Cláudio J.
- et al.
Gastrin-releasing peptide receptor antagonist effects on an animal model of sepsis.
,
38- Ritter C.
- Andrades M.E.
- Reinke A.
- Menna-Barreto S.
- Moreira J.C.
- Dal-Pizzol F.
Treatment with N-acetylcysteine plus deferoxamine protects rats against oxidative stress and improves survival in sepsis.
). The variation in pro-inflammatory cytokines at the systemic level presented a typical pattern here. Pro-inflammatory cytokines may accumulate in brain due to saturable influx transport, retrograde axonal transport systems, or simple diffusion in areas where BBB is impaired. A similar pattern was observed here, as brain pro-inflammatory markers increased, systemic cytokines decreased following acute inflammation recovery. All cytokines were elevated in the brain 24 h after CLP and decreased with time, except TNF-α in the prefrontal cortex (which peaked at 15 days and then decreased). In contrast, alterations in TLR4, GFAP, and nNOS, which are markers associated with inflammatory activation in specific cells, were not observed earlier than 15 days after CLP. Indeed, TLR4 and GFAP levels were altered only 30 days after CLP. These data unveil a bimodal profile of inflammation in the CNS following sepsis, with a more prominent role of pro-inflammatory cytokines at earlier stages, probably resulting from transient BBB disruption during the acute phase of sepsis, followed by resident activation of local cells as BBB is restored and animals recover from acute inflammation. As mentioned above, BBB is disrupted during the acute phase of sepsis, but it restores its integrity and selective permeability as animals recover from acute inflammation (
11- Dal-Pizzol F.
- Rojas H.A.
- dos Santos E.M.
- Vuolo F.
- Constantino L.
- Feier G.
- Pasquali M.
- Comim C.M.
- Petronilho F.
- Gelain D.P.
- Quevedo J.
- Moreira J.C.
- Ritter C.
Matrix metalloproteinase-2 and metalloproteinase-9 activities are associated with blood-brain barrier dysfunction in an animal model of severe sepsis.
,
39- du Moulin G.C.
- Paterson D.
- Hedley-Whyte J.
- Broitman S.A.
E. coli peritonitis and bacteremia cause increased blood-brain barrier permeability.
).
Previous work has demonstrated that CLP induces microglial activation, and this is associated with long-term cognitive dysfunction caused by sepsis (
13- Michels M.
- Vieira A.S.
- Vuolo F.
- Zapelini H.G.
- Mendonça B.
- Mina F.
- Dominguini D.
- Steckert A.
- Schuck P.F.
- Quevedo J.
- Petronilho F.
- Dal-Pizzol F.
The role of microglia activation in the development of sepsis-induced long-term cognitive impairment.
). A similar profile was observed here for astrocytes, as GFAP levels increased late after CLP. Altogether, these observations suggest that the glia exert an important role in the chronic neuroinflammation and long-term impairments in brain function observed after sepsis recovery. In this context, the late increase in TLR4 levels (30 days after CLP) may be associated with late astrocyte/microglial activation instead of polymicrobial-dependent up-regulation. TLR4 triggers production of cytokines, nitric oxide (NO), and reactive oxygen species in microglia and astrocytes (
40- Lee J.Y.
- Lee J.D.
- Phipps S.
- Noakes P.G.
- Woodruff T.M.
Absence of toll-like receptor 4 (TLR4) extends survival in the hSOD1G93A mouse model of amyotrophic lateral sclerosis.
). Abnormal Tau phosphorylation is associated with several neurodegenerative conditions, including AD and other tauopathies where formation of highly dense hydrophobic aggregates of misfolded proteins causes neuronal death (
41- Wang J.Z.
- Xia Y.Y.
- Grundke-Iqbal I.
- Iqbal K.
Abnormal hyperphosphorylation of Tau: sites, regulation, and molecular mechanism of neurofibrillary degeneration.
). Tau presents at least 85 potential sites of phosphorylation, which are regulated by several different protein kinases and phosphatases. The specific phosphorylation of some of these sites has been associated with particular physiological processes, whereas the overstimulation of some sites has been suggested to be more critical for the induction of abnormal aggregation (
42- Goedert M.
- Jakes R.
- Crowther R.A.
- Six J.
- Lübke U.
- Vandermeeren M.
- Cras P.
- Trojanowski J.Q.
- Lee V.M.
The abnormal phosphorylation of tau protein at Ser-202 in Alzheimer disease recapitulates phosphorylation during development.
,
43- Goedert M.
- Jakes R.
- Crowther R.A.
- Hasegawa M.
- Smith M.J.
- Spillantini M.G.
Intraneuronal filamentous tau protein and α-synuclein deposits in neurodegenerative diseases.
44- Šimič G.
- Babič Leko M.
- Wray S.
- Harrington C.
- Delalle I.
- Jovanov-Milošević N.
- Bažadona D.
- Buée L.
- de Silva R.
- Di Giovanni G.
- Wischik C.
- Hof P.R.
Tau protein hyperphosphorylation and aggregation in Alzheimer's disease and other tauopathies, and possible neuroprotective strategies.
). Abnormal stimulation of Tau Ser-202 phosphorylation correlates with AD in adult humans and is associated with neurofibrillary tangle formation (
43- Goedert M.
- Jakes R.
- Crowther R.A.
- Hasegawa M.
- Smith M.J.
- Spillantini M.G.
Intraneuronal filamentous tau protein and α-synuclein deposits in neurodegenerative diseases.
). The involvement of aberrant Tau phosphorylation in brain function impairment following sepsis is consistent with previous observations, including progressive decline in cognitive functions and oxidative damage to CNS (
45- Sharshar T.
- Gray F.
- Lorin de la Grandmaison G.
- Hopkinson N.S.
- Ross E.
- Dorandeu A.
- Orlikowski D.
- Raphael J.C.
- Gajdos P.
- Annane D.
Apoptosis of neurons in cardiovascular autonomic centres triggered by inducible nitric-oxide synthase after death from septic shock.
,
46- Barichello T.
- Fortunato J.J.
- Vitali A.M.
- Feier G.
- Reinke A.
- Moreira J.C.
- Quevedo J.
- Dal-Pizzol F.
Oxidative variables in the rat brain after sepsis induced by cecal ligation and perforation.
). RAGE expression in the adult CNS is more often associated with microglia and astrocytes, although it may be induced in neurons as well. The mechanistic relationship between RAGE and Tau phosphorylation in the brain may involve RAGE up-regulation as a consequence of microglia and astrocyte activation, release of pro-inflammatory mediators to the extracellular milieu, and consequent activation of neurotoxic pathways in neurons, including GSK-3β- and CDK5-regulated cascades that participate in Tau phosphorylation (
47- Zilka N.
- Kazmerova Z.
- Jadhav S.
- Neradil P.
- Madari A.
- Obetkova D.
- Bugos O.
- Novak M.
Who fans the flames of Alzheimer's disease brains? Misfolded tau on the crossroad of neurodegenerative and inflammatory pathways.
). This hypothesis is supported by our finding that in the hippocampus RAGE up-regulation precedes Tau phosphorylation at 15 days.
The immunostaining profile of phosphorylated Tau in the hippocampus is indicative of intracellular sublocalization, whereas no evident formation of tangles was detected. Extracellular accumulation of neurofibrillary tangles is characteristically observed in post-mortem analyses of AD brains, being normally associated with late stages of neurodegeneration. However, Tau abnormal phosphorylation, aggregation, and proteolysis constitute earlier steps in the formation of neurofibrillary tangles, being crucial events in the pathogenesis of AD and other sporadic tauopathies (
48- Šimić G.
- Babić Leko M.
- Wray S.
- Harrington C.
- Delalle I.
- Jovanov-Milošević N.
- Bažadona D.
- Buée L.
- de Silva R.
- Di Giovanni G.
- Wischik C.
- Hof P.R.
Tau protein hyperphosphorylation and aggregation in Alzheimer's disease and other tauopathies, and possible neuroprotective strategies.
,
49Pathological tau proteins in argyrophilic grain disease.
). It is postulated that the process of Tau abnormal phosphorylation and aggregation in tauopathies follows a progression through three stages: pre-tangle formation, when soluble oligomers and small aggregates are observed as intracellular inclusions; intracellular neurofibrillary tangle stage, when it is possible to observe phosphorylated Tau in the form of intracellular filaments; and extraneuronal neurofibrillary tangles stage, when neurons that originated the processes are no longer viable (
48- Šimić G.
- Babić Leko M.
- Wray S.
- Harrington C.
- Delalle I.
- Jovanov-Milošević N.
- Bažadona D.
- Buée L.
- de Silva R.
- Di Giovanni G.
- Wischik C.
- Hof P.R.
Tau protein hyperphosphorylation and aggregation in Alzheimer's disease and other tauopathies, and possible neuroprotective strategies.
). Our observations are suggestive that the first two stages are taking place in hippocampus. In prefrontal cortex, however, Tau phosphorylation had already returned to basal levels 30 days after CLP, although NeuN staining suggests a decreased number of neurons.
The progressive accumulation of Aβ is also consistent with this scenario. AβPP metabolism is normally accelerated in AD and related diseases and may lead to Aβ generation (
50Amyloid precursor protein processing and Alzheimer's disease.
). However, the amyloid plaques observed in pathological analysis of AD brains are believed to take decades to form (
51- Edwards 3rd, G.
- Moreno-Gonzalez I.
- Soto C.
Amyloid-β and tau pathology following repetitive mild traumatic brain injury.
). Accumulation of Aβ in amyloid plaques is affected by transcriptional regulation of AβPP, modifications in expression and/or activity of secretases involved in AβPP cleavage, and oxidative damage resulting from glial activation (
19- Zuo L.
- Hemmelgarn B.T.
- Chuang C.C.
- Best T.M.
The role of oxidative stress-induced epigenetic alterations in amyloid-β production in Alzheimer's disease.
). Interestingly, secretases responsible for AβPP cleavage also act in RAGE shedding (
52- Cho H.J.
- Son S.M.
- Jin S.M.
- Hong H.S.
- Shin D.H.
- Kim S.J.
- Huh K.
- Mook-Jung I.
RAGE regulates BACE1 and Aβ generation via NFAT1 activation in Alzheimer's disease animal model.
,
53- Takuma K.
- Fang F.
- Zhang W.
- Yan S.
- Fukuzaki E.
- Du H.
- Sosunov A.
- McKhann G.
- Funatsu Y.
- Nakamichi N.
- Nagai T.
- Mizoguchi H.
- Ibi D.
- Hori O.
- Ogawa S.
- et al.
RAGE-mediated signaling contributes to intraneuronal transport of amyloid-β and neuronal dysfunction.
), indicating that RAGE and Aβ share important regulatory steps, and their homeostasis may be disrupted by common mechanisms. RAGE is also a major regulator of systemic Aβ translocation into CNS through BBB (
54- Candela P.
- Gosselet F.
- Saint-Pol J.
- Sevin E.
- Boucau M.C.
- Boulanger E.
- Cecchelli R.
- Fenart L.
Apical-to-basolateral transport of amyloid-β peptides through blood-brain barrier cells is mediated by the receptor for advanced glycation end-products and is restricted by P-glycoprotein.
), and Aβ-RAGE interaction enhances Tau phosphorylation (
27- Li X.H.
- Lv B.L.
- Xie J.Z.
- Liu J.
- Zhou X.W.
- Wang J.Z.
AGEs induce Alzheimer-like tau pathology and memory deficit via RAGE-mediated GSK-3 activation.
). Brain accumulation of Aβ following CLP recovery has been previously associated with long-term cognitive impairment (
30- Schwalm M.T.
- Pasquali M.
- Miguel S.P.
- Dos Santos J.P.
- Vuolo F.
- Comim C.M.
- Petronilho F.
- Quevedo J.
- Gelain D.P.
- Moreira J.C.
- Ritter C.
- Dal-Pizzol F.
Acute brain inflammation and oxidative damage are related to long-term cognitive deficits and markers of neurodegeneration in sepsis-survivor rats.
). Here, a progressive increase in Aβ immunodetection was observed with time after CLP and RAGE immune neutralization in hippocampus was able to inhibit this effect in both structures at 30 days, suggesting a link between RAGE and Aβ induction. This is in agreement with previous observations showing that RAGE inhibition prevented Aβ production, inflammation, oxidative stress, and cognitive deficits in an AD mouse model and in rats receiving intrahippocampal AGEs (
31- Deane R.
- Singh I.
- Sagare A.P.
- Bell R.D.
- Ross N.T.
- LaRue B.
- Love R.
- Perry S.
- Paquette N.
- Deane R.J.
- Thiyagarajan M.
- Zarcone T.
- Fritz G.
- Friedman A.E.
- Miller B.L.
- Zlokovic B.V.
A multimodal RAGE-specific inhibitor reduces amyloid β-mediated brain disorder in a mouse model of Alzheimer disease.
,
55- Hong Y.
- Shen C.
- Yin Q.
- Sun M.
- Ma Y.
- Liu X.
Effects of RAGE-specific inhibitor FPS-ZM1 on amyloid-β metabolism and AGEs-induced inflammation and oxidative stress in rat hippocampus.
).
Immunofluorescence images display a staining pattern not indicative of plaque-like structures but an intraneuronal location varying between cell bodies and neurites and, apparently, axons in some cells. Although deposition of Aβ in AD senile plaques is postulated to occur in the course of years, or perhaps decades, the formation, aggregation, and deposition of Aβ in traumatic brain injury (TBI) can be highly accelerated, taking place in a span of hours from an acute episode of trauma (
56- Johnson V.E.
- Stewart W.
- Smith D.H.
Traumatic brain injury and amyloid-β pathology: a link to Alzheimer's disease?.
). The pattern of Aβ staining in hippocampus and prefrontal cortex observed here does not indicate an advanced stage of neurodegeneration commonly associated with the presence of amyloid plaques but intraneuronal accumulation of Aβ or, more likely, its full-length precursor protein, considering the morphological pattern of staining. Early intraneuronal accumulation of Aβ in neurons that are particularly vulnerable in AD were observed in AD brains, as well as Down's syndrome and numerous transgenic mouse models of AD (
57- Gouras G.K.
- Willén K.
- Faideau M.
The inside-out amyloid hypothesis and synapse pathology in Alzheimer's disease.
). This “pre-plaque” accumulation takes place in intraneuronal endosomal vesicles, reportedly located in cell soma and, at higher extent, distal neurites and synapses (
58- Takahashi R.H.
- Almeida C.G.
- Kearney P.F.
- Yu F.
- Lin M.T.
- Milner T.A.
- Gouras G.K.
Oligomerization of Alzheimer's β-amyloid within processes and synapses of cultured neurons and brain.
,
59- Capetillo-Zarate E.
- Gracia L.
- Yu F.
- Banfelder J.R.
- Lin M.T.
- Tampellini D.
- Gouras G.K.
High-resolution 3D reconstruction reveals intra-synaptic amyloid fibrils.
). Recently, the hypothesis that Aβ extracellular plaque deposits are the remnants of these degenerating neurites and synapses, as well as neuron cell soma (neurites and synapses also exist at neuron cell bodies), has been increasingly supported by detailed studies on subcellular localization of Aβ formation, oligomerization, and subsequent deposition in the progression of neurodegeneration (
57- Gouras G.K.
- Willén K.
- Faideau M.
The inside-out amyloid hypothesis and synapse pathology in Alzheimer's disease.
). Our results are in accordance with this hypothesis, as Aβ staining in our model seems to follow a pattern of intracellular localization, not associated with plaques and not as extensive as would be expected following an acute TBI episode. Notwithstanding, in non-transgenic Wistar rats, intraneuronal Aβ accumulation in response to neuroinflammation is not usually as high as observed here, especially considering the intensity of the effect and the age of the animals used in this study (3-month-old young adults). In this context, a possible cross-reactivity with full-length AβPP in our immunofluorescence images should not be ruled out. This is a plausible hypothesis to explain why CLP induces this extent of Aβ labeling in young animals at such short times after surgery. Importantly, this possibility does not invalidate the current interpretation of our observations, considering that Aβ has originated from AβPP cleavage and thus a marked increase in AβPP could constitute an earlier step of this process. Although detailed molecular steps of these events remain to be elucidated, the occurrence of augmented Aβ and Tau phosphorylation in the hippocampus and prefrontal cortex, together with previous evidence mentioned above, are highly suggestive of neurotoxic processes that potentially represent early molecular steps of neurodegenerative cascades in the course of activation. Besides, the observation that RAGE immunoneutralization inhibited Aβ immunodetection and Tau phosphorylation represents valuable information concerning the sequence of events by which sepsis and systemic inflammation trigger long-term neurodegenerative processes.
Circulating RAGE ligands were increased in serum, and RAGE was up-regulated in brain structures. These data could indicate that the influx of peripheral RAGE ligands to the CNS is occurring, as sepsis-impaired BBB allows unspecific transport of pro-inflammatory mediators to the brain (
10The blood-brain barrier in systemic inflammation.
,
11- Dal-Pizzol F.
- Rojas H.A.
- dos Santos E.M.
- Vuolo F.
- Constantino L.
- Feier G.
- Pasquali M.
- Comim C.M.
- Petronilho F.
- Gelain D.P.
- Quevedo J.
- Moreira J.C.
- Ritter C.
Matrix metalloproteinase-2 and metalloproteinase-9 activities are associated with blood-brain barrier dysfunction in an animal model of severe sepsis.
,
60- Opp M.R.
- George A.
- Ringgold K.M.
- Hansen K.M.
- Bullock K.M.
- Banks W.A.
Sleep fragmentation and sepsis differentially impact blood-brain barrier integrity and transport of tumor necrosis factor-α in aging.
). Although the determination of RAGE ligands in total tissue does not reflect the extracellular content of these molecules in the CNS, they presented little or no variations, corroborating the hypothesis that RAGE in CNS is activated and up-regulated by peripherally-produced ligands and cytokines. As a consequence, intracellular effects triggered downstream RAGE activation that may take place in a similar time frame to RAGE protein up-regulation. Importantly, a correlation between Akt/mTOR and RAGE signaling is observed. Akt phosphorylation in brain is increased concomitantly with the increase in RAGE markers in serum and brain, and this relationship was further reinforced by the observation that RAGE
ab inhibited Akt phosphorylation. ERK1/2 and Akt are important components in neurodegenerative and inflammatory pathways and cross-talk to counteract, enhance, or suppress signaling by each other (
61Crosstalk between ERK, AKT, and cell survival.
). In neurodegenerative diseases (especially AD), neuronal ERK1/2 activation is generally associated with GSK3-β cascade regulating Tau phosphorylation, but in glia these kinases are associated with inflammation (
26- Srikanth V.
- Maczurek A.
- Phan T.
- Steele M.
- Westcott B.
- Juskiw D.
- Münch G.
Advanced glycation end products and their receptor RAGE in Alzheimer's disease.
,
62- Kamat P.K.
- Rai S.
- Swarnkar S.
- Shukla R.
- Nath C.
Molecular and cellular mechanism of okadaic acid (OKA)-induced neurotoxicity: a novel tool for Alzheimer's disease therapeutic application.
). A ubiquitous consequence of RAGE binding is NF-κB activation through ERK1/2, which in turn activates inflammatory cytokines and RAGE transcription. Alternatively, Akt activation is associated with survival responses, including inhibition of pro-inflammatory activation and autophagic clearance of protein aggregates (
63- Shi S.
- Liang D.
- Bao M.
- Xie Y.
- Xu W.
- Wang L.
- Wang Z.
- Qiao Z.
Gx-50 inhibits neuroinflammation via α7 nAChR activation of the JAK2/STAT3 and PI3K/AKT pathways.
,
64- Heras-Sandoval D.
- Pérez-Rojas J.M.
- Hernández-Damián J.
- Pedraza-Chaverri J.
The role of PI3K/AKT/mTOR pathway in the modulation of autophagy and the clearance of protein aggregates in neurodegeneration.
). Nonetheless, both exhibit differential states of basal activity and response to stimuli in different brain areas. Recently, it was observed that ERK1/2- and Akt-mediated pathways share little common regulation in different brain structures of AβPP/PS1 transgenic mice, despite simultaneous dysfunctional processes (
65- Guillot F.
- Kemppainen S.
- Lavasseur G.
- Miettinen P.O.
- Laroche S.
- Tanila H.
- Davis S.
Brain-specific basal and novelty-induced alternations in PI3K-Akt and MAPK/ERK signaling in a middle-aged AβPP/PS1 mouse model of Alzheimer's disease.
). ERK1/2 phosphorylation was increased in earlier periods and returned to basal levels later; in contrast, Akt phosphorylation exhibited a pattern of progressive increase. These data point to a shift from ERK1/2-mediated pathways, in earlier periods, to Akt signaling in later periods. Also, these results are in accordance with the decrease in acute pro-inflammatory markers observed during CLP recovery. Thus, a scenario where pro-inflammatory cytokines increase in the CNS causing ERK1/2 phosphorylation during the acute phase of sepsis followed by a second phase where RAGE up-regulation evokes Akt-dependent responses as pro-inflammatory cytokines decrease is consistent with our data. In neurodegenerative conditions, mTOR is generally associated with control of autophagy and apoptosis in response to protein misfolding and aggregation. Akt and ERK1/2 affect mTOR by modulating the activation of the mTOR-inhibitory TSC1–TSC2 complex (
66Targeting molecules to medicine with mTOR, autophagy, and neurodegenerative disorders.
). Akt also controls mTOR by phosphorylation (
67- Asati V.
- Mahapatra D.K.
- Bharti S.K.
PI3K/Akt/mTOR and Ras/Raf/MEK/ERK signaling pathways inhibitors as anticancer agents: Structural and pharmacological perspectives.
,
68Phosphorylation of mammalian target of rapamycin (mTOR) at Ser-2448 is mediated by p70S6 kinase.
). Here, mTOR phosphorylation decreased concomitant to stimulation of Akt. It is possible that the late effect of CLP on mTOR phosphorylation in brain is related to inhibitory feedback mechanisms of RAGE involving Akt late phosphorylation. The observation that RAGE
ab reversed the effects of CLP on mTOR and Akt phosphorylation states after 30 days is consistent with this interpretation. Besides, it is also important to note that HSP70 levels in both structures were decreased 15 days after CLP. HSP70 is a molecular chaperone important for the control of autophagy as its function is directly related to protein misfolding and turnover. Decreased levels of HSP70 could be related to a deficit in autophagic processing, and this, in turn, could contribute to enhance the accumulation of damaged or misfolded proteins.
The time course of events observed here indicates a trend pattern. The decrease in acute systemic inflammation characteristic of sepsis occurs with progressive increase in RAGE-associated markers in serum and brain over time, suggesting a shift from acute inflammation toward chronic neuroinflammation. In different chronic diseases related to organ function degeneration, RAGE expression is elevated and associated with the support of a chronic inflammatory state (
20- Creagh-Brown B.C.
- Quinlan G.J.
- Evans T.W.
- Burke-Gaffney A.
The RAGE axis in systemic inflammation, acute lung injury and myocardial dysfunction: an important therapeutic target?.
,
69- Lukic I.K.
- Humpert P.M.
- Nawroth P.P.
- Bierhaus A.
The RAGE pathway: activation and perpetuation in the pathogenesis of diabetic neuropathy.
). The results presented here suggest that RAGE signaling increases as the acute pro-inflammatory signaling decreases as animals recover from sepsis. The spatial and temporal differences in the expression of markers associated with acute inflammation, RAGE, and neurodegenerative signaling may be related to the systemic origin of inflammation, which may affect CNS structures at different extents depending on their localization, contact area with blood vessels, and relative constitution of different cell types. However, LPS-induced systemic inflammation was reported to cause loss of neurons in both hippocampus and prefrontal cortex, with substantial loss of cholinergic innervations, resembling a neurodegenerative process; and different types of cognitive deficits were observed in this model (
70- Semmler A.
- Frisch C.
- Debeir T.
- Ramanathan M.
- Okulla T.
- Klockgether T.
- Heneka M.T.
Long-term cognitive impairment, neuronal loss and reduced cortical cholinergic innervation after recovery from sepsis in a rodent model.
). Previous works demonstrated several different cognitive deficits in sepsis survivors, and these deficits were associated with the prefrontal cortex and hippocampus inflammation (
6- Barichello T.
- Martins M.R.
- Reinke A.
- Feier G.
- Ritter C.
- Quevedo J.
- Dal-Pizzol F.
Cognitive impairment in sepsis survivors from cecal ligation and perforation.
,
13- Michels M.
- Vieira A.S.
- Vuolo F.
- Zapelini H.G.
- Mendonça B.
- Mina F.
- Dominguini D.
- Steckert A.
- Schuck P.F.
- Quevedo J.
- Petronilho F.
- Dal-Pizzol F.
The role of microglia activation in the development of sepsis-induced long-term cognitive impairment.
,
71- Barichello T.
- Martins M.R.
- Reinke A.
- Feier G.
- Ritter C.
- Quevedo J.
- Dal-Pizzol F.
Long-term cognitive impairment in sepsis survivors.
). Here, it was demonstrated that blocking RAGE signaling in the hippocampus inhibited long-term cognitive dysfunction. All these data taken together suggest that systemic inflammation induces long-term hippocampal and prefrontal alterations that are somehow similar to a neurodegenerative process. These molecular and structural alterations are linked to a phenotype of cognitive dysfunction that is dependent on hippocampus and prefrontal connections.
Antagonism of RAGE signaling using RAGE antibodies or sequestering RAGE ligands with sRAGE have been previously undertaken in models of inflammation and sepsis. The use of monoclonal, polyclonal, or Fab fragments has been extensively applied as a pharmacological inhibitor was available only very recently. RAGE inhibition by these approaches has a significant impact on sepsis, mainly through attenuation of pro-inflammatory signaling (
24- van Zoelen M.A.
- Schmidt A.M.
- Florquin S.
- Meijers J.C.
- de Beer R.
- de Vos A.F.
- Nawroth P.P.
- Bierhaus A.
- van der Poll T.
Receptor for advanced glycation end products facilitates host defense during Escherichia coli-induced abdominal sepsis in mice.
). Utilization of Fab fragments or monoclonal antibodies instead of polyclonal antibodies used here could represent a practical advantage as they would not bind to other scavenger receptors sharing similar epitopes. A necessary step for development of a clinical protocol based on immune neutralization of RAGE, in this context, is the rational design of a selective RAGE antibody for human utilization in a clinical trial. Besides, the delivery of an anti-RAGE molecule to specific brain regions should be considered another essential step, which could demand a surgical approach. A clinical trial with a synthetic RAGE inhibitor, PF-04494700 (Azeliragon or TTP488), indicated no consistent effect on plasma levels of Aβ, inflammatory biomarkers, or secondary cognitive outcomes in phase II (
72- Sabbagh M.N.
- Agro A.
- Bell J.
- Aisen P.S.
- Schweizer E.
- Galasko D.
PF-04494700, an oral inhibitor of receptor for advanced glycation end products (RAGE), in Alzheimer disease.
). However, a follow-up examination, conducted after treatment was suspended, suggested a possible clinical benefit for a low dose, but as these data became evident only long after discontinuation of the treatment based on preliminary results, they remain inconclusive (
73- Galasko D.
- Bell J.
- Mancuso J.Y.
- Kupiec J.W.
- Sabbagh M.N.
- van Dyck C.
- Thomas R.G.
- Aisen P.S.
- Alzheimer's Disease Cooperative Study
Clinical trial of an inhibitor of RAGE-Aβ interactions in Alzheimer disease.
). Currently, a phase III clinical trial is ongoing for efficacy and safety in mild AD (NCT02080364). Nonetheless, in an experimental context, blocking of RAGE with polyclonal antibodies has been successfully demonstrated in different rodent models of systemic inflammation, and effects caused by unspecific binding have not been observed (
74- Origlia N.
- Righi M.
- Capsoni S.
- Cattaneo A.
- Fang F.
- Stern D.M.
- Chen J.X.
- Schmidt A.M.
- Arancio O.
- Yan S.D.
- Domenici L.
Receptor for advanced glycation end product-dependent activation of p38 mitogen-activated protein kinase contributes to amyloid-β-mediated cortical synaptic dysfunction.
,
75- Kuhla A.
- Norden J.
- Abshagen K.
- Menger M.D.
- Vollmar B.
RAGE blockade and hepatic microcirculation in experimental endotoxaemic liver failure.
76- Xia P.
- Deng Q.
- Gao J.
- Yu X.
- Zhang Y.
- Li J.
- Guan W.
- Hu J.
- Tan Q.
- Zhou L.
- Han W.
- Yuan Y.
- Yu Y.
Therapeutic effects of antigen affinity-purified polyclonal anti-receptor of advanced glycation end-product (RAGE) antibodies on cholestasis-induced liver injury in rats.
). Systemic blocking of RAGE previous to sepsis attenuated the effects of endotoxemic shock (
23- Lutterloh E.C.
- Opal S.M.
- Pittman D.D.
- Keith Jr, J.C.
- Tan X.Y.
- Clancy B.M.
- Palmer H.
- Milarski K.
- Sun Y.
- Palardy J.E.
- Parejo N.A.
- Kessimian N.
Inhibition of the RAGE products increases survival in experimental models of severe sepsis and systemic infection.
,
25- van Zoelen M.A.
- van der Poll T.
Targeting RAGE in sepsis.
,
77Antibodies against RAGE in sepsis and inflammation: implications for therapy.
). Here, RAGE
ab was used to evaluate the role of RAGE in brain dysfunction occurring 30 days after CLP. Importantly, the hippocampal administration of RAGE
ab was performed from 15 to 19 days after CLP, to establish the importance of CLP-induced up-regulation of RAGE in changes emerging after the acute pro-inflammatory phase of sepsis. Blocking RAGE prior to CLP, or using constitutive RAGE knock-out models, would result in an overall inhibition of systemic inflammation, as demonstrated previously (
77Antibodies against RAGE in sepsis and inflammation: implications for therapy.
), whereas the main goal of the current approach was to identify a molecular link connecting systemic inflammation and brain dysfunction that may be involved in long-term brain impairment caused by sepsis. This allowed identifying RAGE as an important molecular component in the course of CNS signaling events often associated with the onset and progression of neurodegeneration, such as Aβ accumulation, Tau phosphorylation, astrocyte/microglia activation, and modulation of Akt and mTOR. Also, the participation of RAGE in cognitive impairment as animals recover from an episode of sepsis could also be identified.
In conclusion, the results presented here demonstrate a prominent role of RAGE in the development of biochemical and behavioral changes commonly associated with the onset of neurodegenerative processes. Although the limitations of the current model do not allow an extrapolation of a more important role for RAGE in the evolution of characteristic neurodegenerative conditions (such as AD and other tauopathies, for instance), this study provided valuable insight to the understanding of mechanistic links that could associate episodes of acute systemic inflammation to the activation of neurodegenerative-associated signaling in the CNS in later periods of life. Besides, the development of a clinical protocol for RAGE immune neutralization in a specific brain region could be designed to be applied in early steps of neurodegenerative diseases, such as AD, in future clinical studies. The results presented here indicate that inhibition of RAGE in the hippocampus and prefrontal cortex by injection of RAGEab in hippocampus is able to counteract the degenerative process triggered by sepsis. Clinical strategies that treat the cause of neuronal death in neurodegenerative diseases are currently scarce, as most therapeutic approaches focus on alleviating or retarding the symptoms of neurodegeneration.