Battle in the Brain - the two Sides of Neuroinflammation

 

Overview

Introduction

Neuroinflammation, defined as an inflammatory response within the central nervous system (CNS), is a process triggered by different factors, including infection (e.g. COVID-19, toxoplasmosis, malaria), toxic metabolites, trauma, bleeding or autoimmunity processes (Tohidpour et al., 2017). Inflammation in the CNS occurs as a result of cells sensing pathogen-associated molecular patterns (PAMPs, e.g., Lipopolysaccharide (LPS) or lipoteichoic acid (LTA) from bacteria) or damage-associated molecular patterns (DAMPs, e.g., nuclear or cytosolic proteins or nucleic acids from damaged neurons, extracellular ATP or parasite hemozoin) via pattern-recognition receptors (PRRs).
Neuroinflammation is considered neuroprotective in its normally innate response when the inflammatory activity is for a shorter period; however, prolonged or maladaptive neuroinflammation is a key pathological driver of many neurological diseases, including neurodegenerative (e.g., Alzheimer’s and Parkinson’s diseases, epilepsy and motor degenerative disease) and psychiatric diseases (like schizophrenia and depression), as well as stroke, brain cancer, pain syndromes and traumatic brain injury (Chaney et al., 2021). 
Immune activation in the CNS involves microglia and astrocytes, as well as endothelial and perivascular cells. It triggers a transient, self-limiting immune system response and initiates tissue repair. When the normal resolution mechanism fails, persistent inflammatory responses develop that are detrimental and inhibit regeneration. Persistent inflammation can be triggered by endogenous (e.g., genetic mutation, protein aggregates such as α-synuclein or amyloid β) or environmental (e.g., infection, trauma) factors. An unhealthy lifestyle, obesity, diabetes, mental stress, sleep loss and aging also increase the likelihood of neuroinflammation.

3D rendered image of acute inflammation in the central nervous system (CNS) is accompanied by neurodegeneration and activation of glial cells

Figure 1: Acute inflammation in the central nervous system (CNS) is accompanied by neurodegeneration and activation of glial cells. 

 

Glial cells and macrophages in the first line of defense

Microglia, astrocytes, and oligodendrocytes, the glial cells ubiquitously distributed within the brain parenchyma, together with brain-associated macrophages (BAMs) contribute in a coordinated manner to initiate and modulate the brain's inflammatory response. 
Previously, it was believed that macrophages, microglia, and astrocytes underwent a simplistic polarization into either a pro-inflammatory or anti-inflammatory phenotype. These phenotypes were referred to as M1/M2 for microglia and macrophages (The M1 and M2 Paradigm of Macrophage and Microglia Polarization), or A1/A2 for astrocytes. According to this view, immune cells in the pro-inflammatory phenotype would initiate inflammation by releasing pro-inflammatory cytokines, recruiting other immune cells, and inducing inflammatory responses in neighboring cells. Conversely, cells in the anti-inflammatory phenotype would regulate and potentially halt the inflammatory process by releasing anti-inflammatory cytokines and promoting tissue repair through the release of trophic factors.
However, recent research, especially through genome-wide expression profiling studies, has shed light on the complexity of these cell phenotypes. It has been revealed that these cells can undergo a spectrum of phenotypic states, indicating a more nuanced and finely regulated neuroinflammatory process than previously expected. These new findings challenge the simplistic view of polarization and highlight the multifaceted nature of these cell types during inflammation.

Microglia are the resident innate immune cells of the CNS and account for approximately 10% of the cells in the CNS (Webers et al., 2020) and 20% of the glial cell population of the brain (Troncoso-Escudero et al., 2018). Microglial cells are characterized by highly dynamic processes, which aid in the detection of pathogens and subtle changes in the microenvironment when they are at rest. They act as sentinels that monitor the brain via surface receptors that recognize complement fragments, immunoglobulins, adhesion molecules, chemokine receptors (CCR), toll-like receptors (TLR), purinoreceptors, and scavenger and Fc receptors (Jurga et al., 2020; Prinz et al., 2021; Dermitzakis et al., 2023).
The resting state is transformed into an activated state by danger signals, including PAMPs and DAMPs. Activated microglia communicate by releasing cytokines, which alert surrounding cells and affect their function. They produce both proinflammatory (e.g., tumor necrosis factor-alpha (TNF-α), interleukins IL-1β, IL-6, IL-12, and IL-18, interferon gamma (IFN-γ), inducible nitric oxide synthase (iNOS) and monocyte chemotactic protein 1 (MCP-1)), as well as anti-inflammatory mediators (e.g., interleukins IL-4, IL-10 and IL-13, and transforming growth factor-beta (TGF-β)) (Angiulli et al., 2021). Through the release of these cytokines, microglia cells help to recruit other immune cells to the site and initiate the immune defence against pathogens or promote tissue repair.

P2Y12 is expressed in resting microglia. Indirect immunostaining of PFA fixed mouse cortex section with anti-P2Y12 (Cat. 476 011, 1:2000, red) and anti-IBA1 (cat. no. 234 308, 1:500, green). Nuclei were stained with hematoxlin.
 Indirect immunostaining of (A): PFA fixed wild-type and (B): triple transgenic Alzheimer’s disease mouse brain cortex sections with guinea pig anti-CD11c

Figure 2: P2Y12 is expressed in resting microglia. Indirect immunostaining of PFA fixed mouse cortex section with mouse anti-P2Y12 (Cat. 476 011, 1:2000, red) and guinea pig anti-IBA1 (cat. no. 234 308, 1:500, green). Nuclei were stained with DAPI (blue).

Figure 3: Indirect immunostaining of (A): PFA fixed wild-type and (B): triple transgenic Alzheimer’s disease mouse brain cortex sections with guinea pig anti-CD11c (cat. no. HS-375 004, dilution 1:1000; red) and rabbit anti-IBA1 (cat. no. HS-234 013, dilution 1:1000; green). The red signal shows CD11c expression only in activated microglia. Nuclei were stained with DAPI (blue).

 

Remarkably, activation of microglia is accompanied by changes in protein expression. Proteins characteristic of resting microglia, such as P2Y12 (Figure 2), CX3CR1, HexB, or TMEM119, are downregulated, whereas other proteins, such as CD11c (Figure 3), CD86 (Figure 4), CD11b, IBA1, CD68, F4/80 (mouse specific) and Galectin-3 (Figure 5), are upregulated or expressed only upon microglial activation (Benmamar-Badel et al., 2020, Jurga et al., 2020, Almolda et al., 2015, Garcia-Revilla et al., 2022).

In addition to these acute changes, microglia exhibit a long-lasting change after exposure to inflammatory stimuli and become more sensitive to potentially milder stimuli. On the one hand, microglial priming prepares microglia cells for later immune changes; on the other hand, constant exposure of microglia to infections throughout life may contribute to neurodegenerative diseases (Lima et al., 2022). 
Microglial transcriptome changes have been identified in aged brain leading to enhanced inflammation, impaired phagocytosis, and profound morphological changes that reduce immune surveillance. In mouse models of neurodegenerative diseases, microglia cells with a “disease-associated microglial” (DAM) profile have been described (Finger et al., 2022).

Activated microglia in the ipsilateral hippocampus of a mouse stroke brain highly express CD86 (green) and CD11b (red)
Activated microglia in the lesion area of a mouse stroke brain highly express Galectin-3 (pink), CD11b (red) and IBA1 (green)

Figure 4: (A): Activated microglia in the ipsilateral hippocampus of a mouse stroke brain highly express CD86 (green) and CD11b (red). (B): Higher magnification of the hippocampal region reveals co-localization of both markers in a subset of activated microglia. Indirect immunostaining of a formalin fixed paraffin embedded mouse stroke brain section (14 days after middle cerebral artery occlusion (MCAO) using Tyramide signal amplification with rabbit anti-CD86 (cat. no. HS-466 003, 1:600, green) and rat anti-CD11b (cat. no. HS-384 117, 1:250, red). Nuclei were stained with DAPI (blue).

Figure 5: Activated microglia in the lesion area of a mouse stroke brain highly express Galectin-3 (pink), CD11b (red) and IBA1 (green). Indirect immunostaining of PFA fixed mouse brain section (14 days after MCAO) with (A): rat anti-Galectin-3 (cat. no. HS-477 017, 1:500 pink), (B): rabbit anti-CD11b (cat. no. HS-384 008, 1:500, red) and (C): guinea pig anti-IBA1 (cat. no. 234 308, 1:500, green). (D): Higher magnification of the lesion area reveals different microglial phenotypes expressing either IBA1 alone or in combination with CD11b and/or Galectin-3. Nuclei were stained with DAPI (blue).

 
Products microglia in inflammation
Cat. No. Product Description Application Quantity Price Cart
HS-397 004
F4/80, Guinea pig, polyclonal, antiserumantiserumIHC IHC-P 100 µl$350.00
HS-397 008
F4/80, rabbit, monoclonal, recombinant IgGrecombinant IgGWB IHC IHC-P 100 µl$415.00
HS-397 017
F4/80, rat, monoclonal, purified IgG IgGWB IHC IHC-P IHC-Fr 200 µl$415.00
HS-397 017BT
F4/80, rat, monoclonal, purified IgG IgG, biotinWB IHC-P 100 µg$465.00
HS-477 017
Galectin-3, rat, monoclonal, purified IgG IgG K.O.
mouse specific
WB IHC IHC-P IHC-Fr 100 µg$415.00
234 008IBA1, rabbit, monoclonal, recombinant IgGrecombinant IgGWB IP ICC IHC IHC-P 50 µg$420.00
234 009IBA1, chicken, monoclonal, recombinant IgYrecombinant IgYWB ICC IHC IHC-P ExM 50 µg$425.00
234 011IBA1, mouse, monoclonal, purified IgG IgGWB IP ICC IHC IHC-P 100 µg$420.00
234 013IBA1, rabbit, polyclonal, affinity purifiedaffinity K.O.WB ICC IHC IHC-P 50 µg$380.00
234 017IBA1, rat, monoclonal, purified IgG IgGWB ICC IHC IHC-P 100 µg$415.00
234 308IBA1, Guinea pig, monoclonal, recombinant IgGrecombinant IgGWB IP ICC IHC IHC-P 50 µg$425.00
234-0PIBA1, control peptidecontrol peptide100 µg$105.00
HS-234 008
IBA1, rabbit, monoclonal, recombinant IgGrecombinant IgGWB ICC IHC IHC-P 100 µl$420.00
HS-234 013
IBA1, rabbit, polyclonal, affinity purifiedaffinity K.O.WB IHC IHC-P 200 µl$380.00
HS-234 017
IBA1, rat, monoclonal, purified IgG IgGWB ICC IHC IHC-P 200 µl$415.00
Result count: 48
 

Border-associated macrophages (BAMs) are macrophages located in the distinct anatomic specializations of the CNS and peripheral nervous system (PNS) interfaces, particularly in the leptomeninges (pia mater, arachnoid, and dura mater) surrounding the brain and spinal cord, the perivascular plexus, and the choroid plexus. Although classically identified as an integral component of the CNS immune system, the profile of BAMs and their function in maintaining brain defenses and homeostasis are only recently being elucidated using advanced research tools (Mildenberg, 2022).

BAMs originate from a CD206+ macrophage population derived from the early erythro-myeloid progenitors in the yolk sac (being CD206 the unique general marker for BAMs). These cells constitute a stable cell population capable of self-maintenance, and they can even be partially replenished by bone marrow-derived monocytes (at least, at the choroid plexus). On the one hand, the subpopulations of BAMs generally vary in morphology and motility. However, unlike microglia, BAMs do not possess extensive branching processes and display limited motility. On the other hand, like microglia, BAMs possess a high antigen-presenting capacity and can efficiently phagocytose invading pathogens and foreign substances. They secrete cytokines and chemokines that influence the local microenvironment in response to injury or inflammation. Given their location at the border of the CNS/PNS, BAMs play a pivotal role in recruiting circulating immune cells from the periphery. Consequently, they exhibit shared gene upregulation associated with crucial processes like blood vessel development, lipid and cholesterol metabolism, immune response, and antigen presentation. BAMs can be differentiated from microglia by expression of CD206 (Figure 6) and CD163 (Figure 7). Additionally, BAMs express characteristic macrophage markers, including integrin αM (CD11b), IBA1 (Aif1), the receptor for macrophage-colony stimulating factor (Csf1R), and F4/80, reinforcing their macrophage identity (Gerganova, 2022).

CD206 positive BAMs are located at blood vessels
CD163 co-localized with IBA1 in perivascular macrophages.

Figure 6: CD206 positive BAMs are located on blood vessels. Indirect immunostaining of a formalin fixed paraffin embedded mouse brain section using rabbit anti-CD206 (cat. no. HS-488 003, DAB, brown) and rat anti-IBA1 antibody (cat. no. HS-234 017, AP-RED, red). Nuclei have been visualized by haematoxylin staining (blue).

Figure 7: CD163 co-localized with IBA1 in perivascular macrophages. SMA highlights the blood vessel borders. Indirect immunostaining of PFA fixed mouse brain section with rabbit anti-CD163 (cat. no. HS-455 003, 2 µg/ml, red), mouse anti-IBA1 (cat. no. 234 011, 1:500, green) and guinea pig anti-α-smooth muscle Actin (SMA) (cat. no. 449 004, 1:500, light blue). Nuclei have been visualized by DAPI (blue).

 
Products border-associated macrophages - BAMs
Cat. No. Product Description Application Quantity Price Cart
234 017IBA1, rat, monoclonal, purified IgG IgGWB ICC IHC IHC-P 100 µg$415.00
234 308IBA1, Guinea pig, monoclonal, recombinant IgGrecombinant IgGWB IP ICC IHC IHC-P 50 µg$425.00
234-0PIBA1, control peptidecontrol peptide100 µg$105.00
HS-234 008
IBA1, rabbit, monoclonal, recombinant IgGrecombinant IgGWB ICC IHC IHC-P 100 µl$420.00
HS-234 013
IBA1, rabbit, polyclonal, affinity purifiedaffinity K.O.WB IHC IHC-P 200 µl$380.00
HS-234 017
IBA1, rat, monoclonal, purified IgG IgGWB ICC IHC IHC-P 200 µl$415.00
HS-234 017BT
IBA1, rat, monoclonal, purified IgG IgG, biotinWB IHC-P 100 µg$465.00
HS-234 308
IBA1, Guinea pig, monoclonal, recombinant IgGrecombinant IgGWB ICC IHC IHC-P 50 µg$425.00
Result count: 23
 

Astrocytes constitute ∼50% of all brain cells (Alghamri et al., 2021) and are the most abundant glial cells in the CNS. Astrocytes have multiple functions, including mechanical support of the nervous system, regulation of brain metabolism, uptake and release of neurotransmitters, and participation in the formation of functional synapses, thus helping in the maintenance of a healthy brain (Chiareli et al., 2021).

Under pathological conditions, astrocytes undergo reactive astrogliosis, with morphological and functional changes. Reactive astrocytes are characterized by an increase in glial fibrillary acidic protein (GFAP) and vimentin expression (Singh, 2022) (Figure 8), as well as upregulation of S100B protein synthesis (Higashino et al., 2009). Recent publications describe new promising astrocytic biomarkers in brain injury and neurological diseases, e.g. Aldolase C (ALDOC) (Haddadi et al., 2022), excitatory amino acid transporter 1 (EAAT1, GLAST) (Beschorner et al., 2007) and glutamine synthetase (GS) (Sandhu et al., 2021). Similarly, to microglia, astrocytes become reactive after recognizing signals in their surroundings, such as the accumulation of Aβ aggregates (Chiarini, 2020), and release both pro- or anti-inflammatory cytokines (Tang et al., 2021; Liddelow et al., 2017; Escartin et al., 2021). Astrocytes and microglia play interconnected roles in the inflammatory response within the CNS. Astrocytes can trigger microglia activation by releasing pro-inflammatory cytokines, while activated microglia, in turn, can further stimulate inflammatory responses through cytokine receptors expressed by astrocytes. Reactive astrocytes can also have direct negative effects in the brain. For example, EAAT2, a glutamate importer in astrocytes, is often downregulated in reactive astrocytes which then leads to higher extracellular glutamate concentrations and subsequently endangers neurons via excitotoxicity (Dahlmanns et al, 2023). Consequently, astrocytes exert a modulatory influence on neighboring cells throughout the CNS (Reid and Kuipers, 2021; Rothhammer and Quintana, 2015).

Activated astrocytes in the ipsilateral cortex of a mouse stroke brain highly express GFAP and vimentin.

Figure 8: Activated astrocytes in the ipsilateral cortex of a mouse stroke brain expressing high levels of GFAP and vimentin. (A): Indirect immunostaining of PFA fixed mouse brain section (14 days after MCAO) with rabbit anti-GFAP (cat. no. 173 208, 1:500, red) and guinea pig anti-Vimentin (cat. no. 172 004, 1:500, green). (B): Astrocytes in the contralateral hypothalamus are vimentin negative and show significantly lower GFAP expression than astrocytes in (C): ipsilateral hypothalamus. Nuclei have been visualized by DAPI staining (blue).

 

Astrocytes not only control the immune response, but also influence the tissue regeneration process after injury by controlling the process of scar formation as well as neurogenesis and synaptogenesis (Chiareli et al., 2021). Moreover, astrocytes contribute to the integrity and regulation of the blood-brain barrier, which helps to protect the brain from harmful substances. Dysfunctional astrocytes carrying mutations in ubiquitously expressed genes or disease-susceptible polymorphisms can cause or contribute to neurodegenerative disorders such as Huntington’s or Alzheimer’s disease.

Oligodendrocytes are glial cells that initiate myelinization, enable the propagation of electrical potentials, and assist neurons in metabolism. Oligodendrocytes are the main target of the inflammatory response in the CNS, which is triggered by harmful cytokines released by infiltrating macrophages and microglia, cytotoxicity of T lymphocytes, or by antibodies inducing antibody-mediated cytotoxicity (Kölliker-Frers et al., 2021). However, oligodendrocytes also actively influence inflammatory responses by producing a wide range of immunoregulatory factors (e.g., CXCL10, CCL2, CXCR2, CCL3). Oligodendrocytes also express receptors for interleukins (e.g., IL-4, IL-6, IL-10, IL-12) and other cytokines during inflammation and infection, suggesting that they recruit microglia to damaged tissue (Kölliker-Frers et al., 2021). It has been shown that oligodendrocyte progenitor cells (OPCs, also identified as NG2 glia) are not just bystanders in multiple sclerosis, but have the ability to present antigens to T cells and play a much more important role in neuroinflammation and neurodegeneration (Psenicka et al., 2021, Poggi et al., 2023).

 
Products Astrocytes & Oligodendrocytes in inflammation
Cat. No. Product Description Application Quantity Price Cart
173 011BTGFAP, mouse, monoclonal, purified IgG IgG, biotin K.O.WB IP ICC IHC IHC-P 100 µg$470.00
173 111GFAP, mouse, monoclonal, purified IgM IgMIP ICC IHC 100 µg$415.00
173 208GFAP, rabbit, monoclonal, recombinant IgGrecombinant IgG K.O.WB ICC IHC IHC-P 50 µg$415.00
173 211GFAP, mouse, monoclonal, purified IgG IgG K.O.WB IP ICC IHC IHC-P ELISA 100 µg$415.00
173 211BTGFAP, mouse, monoclonal, purified IgG IgG, biotin K.O.WB IP ELISA 100 µg$465.00
173 308GFAP, Guinea pig, monoclonal, recombinant IgGrecombinant IgG K.O.WB ICC IHC IHC-P 50 µg$415.00
173-0PGFAP, control proteincontrol protein100 µg$110.00
N3802-AF647-LGFAP sdAb, camelid, monoclonal, FluoTag-X2FluoTag-X2, Alexa 647ICC IHC 200 µl$515.00
N3805-250ugGFAP sdAb, camelid, monoclonal, single domain antibodysingle domain antibody250 µg$355.00
N3805-BiotinGFAP sdAb, camelid, monoclonal, single domain antibodysingle domain antibody, biotinICC IHC 250 µg$770.00
N3805-DBCOGFAP sdAb, camelid, monoclonal, single domain antibodysingle domain antibody, DBCOICC IHC 250 µg$770.00
367 004Glutamine synthetase, Guinea pig, polyclonal, antiserumantiserumWB IHC 100 µl$365.00
367 005Glutamine synthetase, Guinea pig, polyclonal, affinity purifiedaffinity WB IHC IHC-P 50 µg$465.00
367 008Glutamine synthetase, rabbit, monoclonal, recombinant IgGrecombinant IgGWB ICC IHC IHC-P 50 µg$415.00
367 011Glutamine synthetase, mouse, monoclonal, purified IgG IgGWB ICC IHC IHC-P 100 µg$415.00
Result count: 38
 

Blood-brain-barrier as the brain’s firewall

Visual graphic of a blood brain barrier. Graphic elements such as microglia depict the healthy state and the inflamed state (right hand side).

Figure 9: Under physiological conditions, brain endothelial cells are connected by tight and adherent junctions such as claudins and occludins, which are responsible for the low paracellular permeability of the BBB. Activation of astrocytes and microglia under inflammatory conditions leads to increased production of cytokines, chemokines and matrix metalloproteinases (MMPs). Glial cell-derived reactive oxygen species (ROS) and MMP-9 affect BBB integrity. ROS and MMP-9 modulate and digest tight junction proteins, leading to BBB impairment. Increased expression of ICAM1 and VCAM1 facilitates leukocyte recruitment into the brain. The infiltration of immune cells and other substances from the blood enhances the activation of glial cells.

 

The Blood-brain barrier (BBB) is the firewall of the brain and as such maintains brain homeostasis and protects neurons. The BBB is a highly specialized, multicellular structure that functions as a selective diffusion barrier between the peripheral circulation and the CNS. The BBB consists of endothelial cells of the capillary wall termed as brain microvascular endothelial cells (BMECs), pericytes, and astrocytes (astroglial basement membranes covered by astrocytic endfeet) (Figure 9). BMECs are connected by complex tight junctions (TJs) and adherent junctions (AJs), e.g., Claudin 5 and Occludin (Figure 10), to restrict the paracellular and transcellular diffusion of molecules in the CNS. The BBB is not a wall in the true sense, but a selective barrier between blood circulation and the brain, that strictly regulates the transfer of various substances and also blood cells from blood to brain. Because the brain is the most energy-consuming organ in the body, various kinds of highly expressed receptors and transporters play a critical role in supplying the brain with essential nutrients, including transferring receptor (TfR), LDL receptor (LDLR) and glucose transporter-1 (GLUT1) (Han and Jiang et al., 2020). Alteration of one property, e.g., transcytosis or transport, can significantly alter the neuronal environment and lead to seizures, autism spectrum, and psychomotor retardation syndromes.

Occludin plays a role in the formation and regulation of the tight junction paracellular permeability barrier.
Aquaporin-4 (AQP4) is expressed on perivascular astrocytes end feet that surround blood vessels.

Figure 10: Occludin plays a role in the formation and regulation of the tight junction paracellular permeability barrier. Indirect immunostaining of PFA fixed mouse cortex section with guinea pig anti-Occludin (Cat. 447 005, 1:500, red) and mouse anti-GFAP (Cat. 173 011, 1:500, green). Nuclei have been visualized by DAPI staining (blue).

Figure 11: Aquaporin-4 (AQP4) is expressed on perivascular astrocytes end feet that surround blood vessels. Indirect immunostaining of PFA fixed mouse cortex section with mouse anti-AQP4 (Cat. 429 011, 1:500, red) and rat anti-CD31 (Cat. HS-351 117, 1:500, green). Nuclei have been visualized by DAPI staining (blue).

 

Aging leads to changes in the structure and function of the BBB, which makes the brain more susceptible to neuronal impairment or even neurodegeneration (Hussain et al., 2021). These changes include age-related pericyte dysfunction and vascular cell adhesion molecule 1 (VCAM1) up-regulation. Several studies link for example BBB dysfunction and AD pathology (Profaci et al., 2020). In conditions of stroke, pericytes constrict brain capillaries and then die, which may lead to a long-lasting decrease of blood flow and loss of BBB function, increasing the death of nerve cells. (Hall et al., 2014). Other factors influencing BBB permeability are, e.g., intestinal microbiota changes through unhealthy eating or systemic inflammation (Sun et al., 2022).

Aquaporin-4 (AQP4) is the most abundant bidirectional water channel in the CNS and shows the highest density on perivascular astrocytes endfeet that surround blood vessels. (Figure 11). AQP4 plays a fundamental role in BBB disruption. Abnormal expression and dysfunction of AQP4 has been observed in numerous cognitive disorders, such as AD, vascular dementia and Creutzfeldt-Jakob disease (CJD) (Wang et al., 2022). BBB dysfunction in various brain diseases includes also impaired transport mechanisms. Under physiological conditions, endogenous Aβ is transported bidirectionally across the BBB, with influx receptor RAGE (receptor for advanced glycosylation end products) and efflux receptors LRP1 (low-density lipoprotein receptor-related protein 1) and transporter P-glycoprotein (P-gp) working in tandem. Under AD conditions, increased RAGE and decreased LRP1 and transporter P-gp expression promote Aβ accumulation in the brain. Reduced GLUT1 expression leads to impaired glucose transport and is associated with BBB breakdown in AD pathology. Enhanced transcellular transport is characteristic of the early phase of stroke mediated by increased endothelial caveolin-1 (Cav-1) expression (Han and Jiang, 2020). Through degradation of matrix metalloproteinases (MMPs), extracellular matrix (ECM), and TJ proteins, Cav-1 also directly determines the endothelial barrier integrity following stroke and is responsible to regulate thrombo-inflammatory activity in the peri-infarct area (Zhang et al., 2022).

The integrity of the ECM, and in particular the basement membrane (BM), is essential for the maintenance of proper brain function. In the normal brain, the ECM is composed of chondroitin sulfate proteoglycans (CSPGs), heparan sulphate proteoglycans (HSPGs), hyaluronan, laminins, collagen, and fibronectin, all of which are produced by the cell types found in the brain – neurons, glial cells, and endothelial cells (George et al., 2018). The BM is composed of various ECM molecules, with collagen IV being the most abundant component. The BM forms a thin acellular layer that contributes significantly to vascular barrier function (Xu et al., 2018). Increased expression of ECM components has been described in various brain injuries. For example, the CSPG protein nerve/glial-antigen 2 (NG2), expressed by oligodendrocyte progenitor cells (OPCs), microglial cells, and pericytes, has been found to be increased in glial scars following experimental traumatic brain injury (TBI) (George et al., 2018). High expression of collagen IV and fibronectin has been observed in experimental rodent stroke models and in human stroke tissue (Michalski et al., 2020).

Damage to the BBB allows immune cells or plasma proteins to enter the brain parenchyma, leading to inflammatory processes in the brain. Intercellular adhesion molecule (ICAM) 1, VCAM1 and platelet-endothelial cell adhesion molecule 1 (PECAM 1, CD31) are constitutively expressed at low levels in endothelial cells. Under hypoxic conditions or through cytokine release of IL-1β and TNF-α, ICAM1 and VCAM1 are dramatically increased (Figure 12). ICAM 1 regulates leukocyte recruitment into the brain during neuroinflammation and ischemia, whereas the role of VCAM1 is less clear (Yang et al., 2019). Limiting immune cell trafficking across the BBB has proven to be effective in MS. However, leukocyte trafficking is required at low levels in order to limit infections. The infiltration of immune cells and other substances from the blood enhances the activation of glial cells. Thus, BBB dysfunction enhances the development of neuroinflammation (Takata et al., 2021).

VCAM 1 expression is increased in blood vessels near the stroke lesion site.

Figure 12: VCAM1 expression is increased in blood vessels near the stroke lesion site. Indirect immunostaining of a formalin fixed paraffin embedded mouse stroke brain section (14 days after MCAO) using Tyramide signal amplification with guinea pig anti-VCAM1 (cat. no. HS-470 004, 1:750, green) and rat anti-CD31 (cat. no. HS-351 117, 1:1500, red). Nuclei have been visualized by DAPI staining (blue).

 
Products Blood-brain barrier (BBB) in inflammation
Cat. No. Product Description Application Quantity Price Cart
482 005Claudin5, Guinea pig, polyclonal, affinity purifiedaffinity WB ICC IHC-P IHC-Fr 50 µg$460.00
462 004Collagen type IV, Guinea pig, polyclonal, antiserumantiserumWB IHC IHC-P IHC-Fr 100 µl$365.00
462 017Collagen type IV, rat, monoclonal, purified IgG IgGIHC IHC-P IHC-Fr 100 µg$415.00
419 005GLUT1, Guinea pig, polyclonal, affinity purifiedaffinity IHC IHC-P IHC-Fr 50 µg$460.00
447 005Occludin, Guinea pig, polyclonal, affinity purifiedaffinity WB ICC IHC IHC-P IHC-Fr 50 µg$460.00
HS-470 004
VCAM1, Guinea pig, polyclonal, antiserumantiserum
mouse specific
WB IHC IHC-P 100 µl$350.00
Result count: 21
 

Infiltrating peripheral immune cells form the cavalry

Under normal conditions, the BBB strictly limits the infiltration of peripheral immune cells into the CNS. Only a few subsets of immune cells required for immune surveillance and detected in the cerebrospinal fluid (CSF) migrate through the BBB in a multi-step extravasation process. In the absence of CNS antigens presented by APCs (antigen presenting cells), T cells cross only the endothelium of the BBB but not the glial layer and remain in the CSF. During inflammation, T cells recognize their cognate antigens on perivascular APCs and are reactivated behind the BBB. Matrix metalloproteinases produced by myeloid cells and astrocytes cleave the astrocyte endfeet and allow T cells to enter the CNS parenchyma. Changes in the expression of adhesion molecules and decreased expression of junctional molecules during inflammation facilitate the recruitment of peripheral immune cells across the BBB (Marchetti and Engelhardt, 2020). Peripheral immune cells play a dual role in neuroinflammation and can produce either protective or pathogenic effects (Figure 13).

The good and the dark side of peripheral immune cells: Infiltration of peripheral immune cells during inflammation induces protective or pathogenic effects in the brain. 

Figure 13: The good and the dark side of peripheral immune cells: Infiltration of peripheral immune cells during inflammation induces protective (shown in blue) or pathogenic effects (marked in grey) in the brain. 

 

Neutrophils are the first cells attracted by chemical mediators or DAMPs to the site of injury or infection during the acute phase of CNS pathophysiology. Chil3 (YM-1) is a rodent-specific chitinase-like protein (CLP) expressed by neutrophils (Figure 14), but also alveolar marophages, alternatively activated macrophages and microglia, and osteoclasts (Kang et al., 2022). Release from free oxygen radicals and MMP-9 by neutrophils leads to BBB breakdown and increased infiltration of peripheral immune cells (Manda-Handzlik et al., 2019). Accumulating neutrophils release extracellular web-like structures composed of DNA and proteins called neutrophil extracellular traps (NETs), which protect injured brain from bacterial, parasitic, viral and fungal attack. However, excessive NET formation is implicated in brain disorders and neurological pathologies (Manda-Handzlik et al., 2019).

T cells are important immune cells of the adaptive immune system and are often considered pathogenic when localized to the CNS. There are two main types of T cells: cytotoxic CD8+ cells and CD4+ helper T-cells (Figure 15). Recent studies have shown that T cells play an active role in limiting inflammation and CNS damage. Leptomeningeal macrophages and choroid plexus APCs present antigens to CD4 T cells contained in the CSF under normal conditions, allowing T cells to enter the CNS. In the short term, T cells are neuroprotective but can become pathogenic if the inflammation is not subsided. Pathogenic T cells are described in MS and chronic neurodegenerative diseases like PD or AD. Tissue-resident memory T cells (TRM cells), which are indispensable for local protection against pathogens, have been identified in the parenchyma of mouse and human brains (Smolders et al., 2018). In the healthy brain, CD4 T cells are involved in brain development by helping microglia to mature (Pasciuto et al., 2020).

Chil3 (YM-1) is a rodent-specific neutrophil granule protein that visualizes neutrophile infiltration.
CD4 positive (red) helper T cells and CD8 positive cytotoxic T cells are detected in a mouse brain infected with Toxoplasma (T.) gondii.

Figure 14: Chil3 (YM-1) is a rodent-specific neutrophil granule protein that visualizes neutrophile infiltration. Indirect immunostaining of formalin fixed paraffin embedded brain sections from a K18-hACE2 transgenic mouse infected with SARS-CoV2 using Tyramide signal amplification with (A): rat anti-Chil3 (cat. no. HS-442 017, 1:1500, red) and rabbit anti-CD45 (cat. no. HS-427 008, 1:2000, green) or (B): with rat anti-Chil3 (cat. no. HS-442 017, 1:1500, red) and rabbit anti-IBA1 (cat. no. HS-234 008, 1:2000, green). Nuclei have been visualized by DAPI staining (blue).

Figure 15: CD4 positive (red) helper T cells and CD8 positive cytotoxic T cells are detected in a mouse brain infected with Toxoplasma (T.) gondii. Indirect immunostaining of a formalin fixed paraffin embedded mouse brain section infected with T. gondii using Tyramide signal amplification with rat anti-CD4 (cat. no. HS-360 117, 1:200, red) and rabbit anti-CD8a (cat. no. HS-361 003, 1:250, green). Nuclei have been visualized by DAPI staining (blue).

 

Monocyte-derived macrophages (MDMs). Classical CCR2-positive monocytes are recruited to the CNS only under pathological conditions, where they differentiate into MDMs. MDMs have higher phagocytic activity than microglia and also have a greater ability to induce antigen-specific T cell proliferation due to higher expression of MHCII (Chang et al., 2021). In stroke, infiltrating macrophages and mononuclear cells are thought to initiate debris clearance and tissue repair in concert with resident cells. Immigration of anti-inflammatory monocytes is associated with improved outcomes after experimental stroke. However acute inflammation can become to chronic, and inflammatory mononuclear cells and macrophages have been found to persist for years in chronic lesions (Spiteri et al., 2022).

Dendritic cells. CD11c-positive brain dendritic cells represent 1 % of immune cells found in the brain and are present in the meninges, choroid plexus, cerebrospinal fluid, and perivascular spaces. They are the predominant antigen-presenting cells of the brain and act as sentinels under steady-state conditions. Their number increases during neuroinflammation. By producing  IFN, dendritic cells can directly target viruses or contribute to neuroinflammation by activating T cells (Constant et al., 2022). Monocyte-derived dendritic cells (moDCs) emerge in the setting of active inflammation and are not present in the healthy parenchymal tissue. In contrast to brain-resident DCs, moDCs are poor APCs and are suggested to play a role in the modulation of the inflammatory milieu and clearance of myelin debris (Giles et al., 2018). 

B cells can be identified by their expression of the B cell lineage-specific antigens CD19 and CD20 and contribute to many aspects of the adaptive immune system, most notably the production and secretion of antibodies. In healthy brains, B cells are rarely found in the CNS parenchyma. More commonly, B cells are found in the meninges, particularly in the dura mater. In viral or bacterial CNS infections, B cells may be recruited and exert a protective function. However, secretion of antibodies can lead to autoimmune CNS diseases (Jain et al., 2021). B cells also play a role in neurodegenerative diseases through antibody-independent mechanisms. It is suggested that B cell-derived cytokines may play a role in the development of MS (Ahn et al., 2021).

Natural Killer (NK) cells. The Innate Lymphoid Cell (ILC) family is a relatively new immune cell population and consists of the three family members ILC1, ILC2 and ILC3. NK cells  are considered part of the ILC1 family. NK cells are bone marrow (BM)-derived haematopoietic cells that are widely distributed in peripheral lymphoid organs and the circulatory system. Under pathological conditions such as stroke, NK cells are recruited by chemokines to the CNS, where they can have a protective function (Wang et al., 2023). However, NK cells may have also pathogenic functions, depending on the NK cell subset, microenvironment, disease type or stage (Liu et al., 2021). Residential NK cells have been identified in the brain parenchyma, that may play a critical role in controlling the extent of CNS inflammation (Hao et al., 2010).

 
Products infiltrating peripheral immune cells
Cat. No. Product Description Application Quantity Price Cart
HS-427 017
CD45, rat, monoclonal, purified IgG IgG
mouse specific
IHC IHC-P IHC-Fr 200 µl$415.00
HS-427 117
CD45, rat, monoclonal, purified IgG IgG
human specific
IHC-P 100 µg$415.00
HS-361 003
CD8a, rabbit, polyclonal, affinity purifiedaffinity
mouse specific
WB IHC IHC-P IHC-Fr 200 µl$375.00
HS-361 008
CD8a, rabbit, monoclonal, recombinant IgGrecombinant IgG
mouse specific
WB IHC-P 100 µl$415.00
HS-361 017
CD8a, rat, monoclonal, purified IgG IgG
mouse specific
WB IHC-P IHC-Fr 200 µl$415.00
442 004Chil3 / YM1, Guinea pig, polyclonal, antiserumantiserumWB ICC IHC IHC-P 100 µl$365.00
442-0PChil3 / YM1, control peptidecontrol peptide100 µg$105.00
HS-442 017
Chil3 / YM1, rat, monoclonal, purified IgG IgG
mouse specific
WB ICC IHC IHC-P 100 µg$415.00
HS-442 117
Chil3 / YM1, rat, monoclonal, purified IgG IgG
mouse specific
WB ICC IHC IHC-P 100 µg$415.00
234 008IBA1, rabbit, monoclonal, recombinant IgGrecombinant IgGWB IP ICC IHC IHC-P 50 µg$420.00
234 009IBA1, chicken, monoclonal, recombinant IgYrecombinant IgYWB ICC IHC IHC-P ExM 50 µg$425.00
234 011IBA1, mouse, monoclonal, purified IgG IgGWB IP ICC IHC IHC-P 100 µg$420.00
234 013IBA1, rabbit, polyclonal, affinity purifiedaffinity K.O.WB ICC IHC IHC-P 50 µg$380.00
234 017IBA1, rat, monoclonal, purified IgG IgGWB ICC IHC IHC-P 100 µg$415.00
234 308IBA1, Guinea pig, monoclonal, recombinant IgGrecombinant IgGWB IP ICC IHC IHC-P 50 µg$425.00
Result count: 36
 

Neuropeptides modulate the battle

Neuropeptides (Neuropeptides) are small endogenous protein messengers synthesized and secreted by neurons that affect development, reproduction, physiology, and behavior (Yeo et al., 2022). However, neuropeptides have been shown to modulate the inflammatory response in the brain by controlling immune cell and glia cell function and BBB permeability (Figure 16). Receptors for several neuropeptides have been detected on microglial cells, such as receptors for NPY (neuropeptide Y), VIP (Vasoactive intestinal peptide) and PACAP (pituitary adenylate cyclase-activating polypeptide). VIP is known to play a critical immunoregulatory role in the periphery and is not only expressed by nerve endings but also by CD4 and CD8 T cells. VIP exhibits anti-inflammatory activity by inhibiting macrophage and microglia activation and acts on mast cells, dendritic cells and synovial fibroblasts. In the brain, VIP promotes neuron proliferation, survival and axon regeneration and is therefore a promising therapeutic target in MS and Parkinson’s disease (Ganea et al., 2015; Martinez et al., 2019). Like VIP, PACAP inhibits inflammation and promotes neuronal survival, and the administration of exogenous PACAP has been shown to be beneficial in a mouse model of T. gondii-induced neuroinflammation (Figueiredo et al., 2022).

Visual grapgic of immunomodulatory roles of the neuropeptides PACAP, VIP and Substance P in the brain.

Figure 16: Immunomodulatory roles of the neuropeptides PACAP, VIP and Substance P in the brain.

 

Substance P and corticotropin-releasing hormone (CRH) have been shown to activate microglia and mast cells, leading to the release of pro-inflammatory cytokines, which promote permeability of the BBB. By increasing the permeability of BBB, upregulation of substance P, as well as bradykinin and neurotensin, exacerbates stroke pathology (Yeo et al., 2022).
The dysfunction of the neuropeptide activity contributes to several neurological diseases. NPY and substance P have been reported to be reduced in the cerebral cortex of AD patients. Since NPY is reported to attenuate the toxic effects of Aβ accumulation, NPY is discussed as a potential therapeutic option in AD (Yeo et al., 2022). Neuropeptides involved in inflammation also play a central role in the pain response. Migraines and headaches emerge with the vasodilation of blood vessels in the brain following the release of calcitonin gene-related peptide (CGRP) and substance P by afferent nerve terminals (Carr and Frings, 2019).

NPY is a neuropeptide with proinflammatory properties that is produced abundantly in the central and peripheral nervous system.

Figure 17: NPY is a neuropeptide with proinflammatory properties that is abundantly produced in the central and peripheral nervous system. NPY modulates inflammation by regulating key immune cell functions and is upregulated around the area of stroke injury. Indirect immunostaining of a PFA fixed mouse brain section (14 days after MCAO) with chicken anti-NPY antibody (cat. no. 394 006, 1:500, green). Nuclei have been visualized by DAPI staining (blue).

 
Products neuropeptides in inflammation
Cat. No. Product Description Application Quantity Price Cart
414 004CGRP, Guinea pig, polyclonal, antiserumantiserumIHC IHC-P iDISCO Clarity 100 µl$365.00
394 004Neuropeptide Y, Guinea pig, polyclonal, antiserumantiserumICC IHC IHC-P 100 µl$365.00
394 006Neuropeptide Y, chicken, polyclonal, affinity purifiedaffinity K.O.ICC IHC 200 µl$380.00
418 005Neurotensin, Guinea pig, polyclonal, affinity purifiedaffinity IHC IHC-P 50 µg$460.00
459 005Substance P, Guinea pig, polyclonal, affinity purifiedaffinity IHC 50 µg$460.00
443 005VIP, Guinea pig, polyclonal, affinity purifiedaffinity ICC IHC IHC-P 50 µg$465.00
Result count: 6
 

The Three-Act Symphony of Neuroinflammation: Insights from Acute Ischemic Stroke

An example of the complexity of neuroinflammation involving multiple agents is the inflammatory process after acute ischemic stroke. It has been shown that the inflammatory process after stroke occurs in three stages: acute phase (1): elimination of necrotic cells by microglia and macrophages and initial neutrophil influx in the first minutes to hours,  subacute phase (2) resolution of inflammatory processes by microglial activation and leukocyte infiltration during the first days after the ischemic insult, and late phase (3) astrocytic and microglial reparative processes with contribution of inflammatory cells (Figure 18) (Jurcau and Simion, 2022).

Visual graphic of a inflammatory process in ischemic stroke adapted from Jurcau and Simion

Figure 18: Inflammatory process in ischemic stroke adapted from Jurcau and Simion., 2022. In the acute phase immediately after ischemic stroke, damaged neurons release DAMPs, which lead to microglial and endothelial activation and infiltration of neutrophils. Cytotoxic microglia release pro-inflammatory interleukins, MMPs and ROS, which weaken the BBB. Pericytes and astrocytic endfeet are lifted from the BBB basement membrane. In the subacute phase, leukocytes and monocytes infiltrate through the “leaky” BBB, where they produce pro-inflammatory factors and exacerbate neuronal damage. In the late subacute phase, microglia switch to a phagocytic M2 phenotype and clear tissue debris. The release of anti-inflammatory mediators and neurotrophic factors promotes BBB repair, neurogenesis, astrogenesis, oligodendrogenesis and angiogenesis in the late phase.

 

Products

 
All mentioned products
Cat. No. Product Description Application Quantity Price Cart
HS-384 117
CD11b, rat, monoclonal, purified IgG IgG
mouse specific
WB IHC IHC-P IHC-Fr 200 µl$415.00
HS-384 117BT
CD11b, rat, monoclonal, purified IgG IgG, biotinWB IHC-P 100 µg$465.00
HS-384 308
CD11b, Guinea pig, monoclonal, recombinant IgGrecombinant IgG
mouse specific
WB IHC IHC-P IHC-Fr 100 µl$415.00
375-0P
CD11c, control peptidecontrol peptide100 µg$105.00
HS-375 003
CD11c, rabbit, polyclonal, affinity purifiedaffinity WB IHC IHC-P 200 µl$370.00
HS-375 004
CD11c, Guinea pig, polyclonal, antiserumantiserumWB IHC IHC-P 100 µl$350.00
HS-375 017
CD11c, rat, monoclonal, purified IgG IgGIHC IHC-P IHC-Fr 100 µg$415.00
HS-455 003
CD163, rabbit, polyclonal, affinity purifiedaffinity
mouse specific
WB IHC IHC-P 200 µl$370.00
HS-455 013
CD163, rabbit, polyclonal, affinity purifiedaffinity
human specific
IHC-P 50 µg$370.00
HS-439 003
CD19, rabbit, polyclonal, affinity purifiedaffinity
mouse specific
WB ICC IHC IHC-P 200 µl$370.00
HS-439 017
CD19, rat, monoclonal, purified IgG IgG
mouse specific
WB IHC IHC-P IHC-Fr 100 µg$415.00
HS-439 103
CD19, rabbit, polyclonal, affinity purifiedaffinity
human specific
WB IHC-P 50 µg$370.00
HS-439 117
CD19, rat, monoclonal, affinity purifiedaffinity
human specific
IP IHC-P 100 µg$415.00
HS-488 003
CD206, rabbit, polyclonal, affinity purifiedaffinity WB IHC IHC-P 50 µg$370.00
351 004CD31, Guinea pig, polyclonal, antiserumantiserum
discontinued, replacement: HS-351 004
WB ICC IHC-P 100 µl
 

Authors: Dr. Christel Bonnas and Dr. Roser Ufartes

Christel and Roser have a solid background in histopathology and are very interested in understanding the origin and development of human diseases, especially the immune system and cancer. They are responsible for antibody development, validation and quality control of our HistoSure Cancer & Immunology product line.

 

Literature

Ahn et al., 2021. B Cells in Neuroinflammation: New Perspectives and Mechanistic Insights. PMID: 34206848

Alghamri et al., 2021. Targeting Neuroinflammation in Brain Cancer: Uncovering Mechanisms, Pharmacological Targets, and Neuropharmaceutical Developments. PMID: 34084145

Almolda et al., 2015. Alterations in microglial phenotype and hippocampal neuronal function in transgenic mice with astrocyte-targeted production of interleukin-10. PMID: 25449577

Angiulli et al., 2021. Blood-Based Biomarkers of Neuroinflammation in Alzheimer's Disease: A Central Role for Periphery? PMID: 34573867

Benmamar-Badel et al., 2020. Protective Microglial Subset in Development, Aging, and Disease: Lessons From Transcriptomic Studies. PMID: 32318054

Beschorner et al., 2007. Expression of EAAT1 reflects a possible neuroprotective function of reactive astrocytes and activated microglia following human traumatic brain injury. PMID: 17330806

Carr and Frings, 2019. Neuropeptides in sensory signal processing. PMID: 30377783

Chaney et al., 2021. PET Imaging of Neuroinflammation. doi: 10.1016/B978-0-12-816386-3.00047-8.

Chang et al., 2021. Divergent Functions of Tissue-Resident and Blood-Derived Macrophages in the Hemorrhagic Brain. PMID: 33840225

Chiareli et al., 2021. The Role of Astrocytes in the Neurorepair Process. PMID: 34113618

Constant et al., 2022. Role of Dendritic Cells in Viral Brain Infections. PMID: 35529884

Dahlmanns et al., 2023. Glial Glutamate Transporter-Mediated Plasticity: System xc-/xCT/SLC7A11 and EAAT1/2 in Brain Diseases. PMID: 37005761

Dermitzakis et al., 2023. CNS Border-Associated Macrophages: Ontogeny and Potential Implication in Disease PMID: 37232741

Escartin et al., 2021. Reactive astrocyte nomenclature, definitions, and future directions. PMID: 33589835

Figueiredo et al., 2022. The neuropeptide PACAP alleviates T. gondii infection induced neuroinflammation and neuronal impairment. PMID: 36403002

Finger et al., 2022. Age-related immune alterations and cerebrovascular inflammation. PMID: 34711943

Ganea et al., 2015. The neuropeptide vasoactive intestinal peptide: direct effects on immune cells and involvement in inflammatory and autoimmune diseases. PMID: 25422088

Garcia-Revilla et al., 2022. Galectin-3, a rising star in modulating microglia activation under conditions of neurodegeneration. PMID: 35859075

George et al., 2018. Extracellular Matrix and Traumatic Brain Injury. PMID: 29344975

Gerganova et al., 2022. CNS border-associated macrophages in the homeostatic and ischaemic brain. PMID: 35667516

Giles et al., 2018. CNS-resident classical DCs play a critical role in CNS autoimmune disease. PMID: 30226829

Haddadi et al., 2022. Aldolase C Profiling in Serum after Mild Traumatic Brain Injury: A Prospective Cohort Study. PMID: 35017775

Hall et al., 2014. Capillary pericytes regulate cerebral blood flow in health and disease. PMID: 24670647

Han and Jiang et al., 2020. Evolution of blood-brain barrier in brain diseases and related systemic nanoscale brain targeting drug delivery strategies. PMID: 34522589

Hao et al., 2010. Central nervous system (CNS)–resident natural killer cells suppress Th17 responses and CNS autoimmune pathology. PMID: 20696699

Higashino et al., 2009. Immunohistochemical analysis of brain lesions using S100B and glial fibrillary acidic protein antibodies in arundic acid- (ONO-2506) treated stroke-prone spontaneously hypertensive rats. PMID: 19657585

Hussain et al., 2021. Blood–Brain Barrier Breakdown: An Emerging Biomarker of Cognitive Impairment in Normal Aging and Dementia. PMID: 34489623

Jain et al., 2021. B cells in central nervous system disease: diversity, locations and pathophysiology. PMID: 34903877

Jäkel and Dimou, 2017. Glial Cells and Their Function in the Adult Brain: A Journey through the History of Their Ablation. PMID: 28243193

Jurcau and Simion, 2022. Neuroinflammation in Cerebral Ischemia and Ischemia/Reperfusion Injuries: From Pathophysiology to Therapeutic Strategies. PMID: 35008440

Jurga et al., 2020. Overview of General and Discriminating Markers of Differential Microglia Phenotypes. PMID: 32848611

Kang et al., 2022. An update on Ym1 and its immunoregulatory role in diseases. PMID: 35967383

Kölliker-Frers et al., 2021. Neuroinflammation: An Integrating Overview of Reactive-Neuroimmune Cell Interactions in Health and Disease. PMID: 34158806

Liddelow et al., 2017. Reactive Astrocytes: Production, Function, and Therapeutic Potential. PMID: 28636962

Lima et al., 2022. Microglial Priming in Infections and Its Risk to Neurodegenerative Diseases. PMID: 35783096

Liu et al., 2021. NK Cells in Autoimmune Diseases: Protective or Pathogenic? PMID: 33777006

Manda-Handzlik et al., 2019. The Brain Entangled: The Contribution of Neutrophil Extracellular Traps to the Diseases of the Central Nervous System. PMID: 31766346

Marchetti and Engelhardt, 2020. Immune cell trafficking across the blood-brain barrier in the absence and presence of neuroinflammation. PMID: 32923970

Martinez et al., 2019. A Clinical Approach for the Use of VIP Axis in Inflammatory and Autoimmune Diseases. PMID: 31861827

Michalski et al., 2020. Increased Immunosignals of Collagen IV and Fibronectin Indicate Ischemic Consequences for the Neurovascular Matrix Adhesion Zone in Various Animal Models and Human Stroke Tissue. PMID: 33192578

Mildenberger et al., 2022. Diversity and function of brain-associated macrophages. PMID: 35462276

Pasciuto et al., 2020. Microglia Require CD4 T Cells to Complete the Fetalto-Adult Transition. PMID: 32702313

Poggi et al., 2023. NG2-glia: rising stars in stress-related mental disorders? PMID: 36280754

Prinz et al., 2021. Microglia and Central Nervous System–Associated Macrophages—From Origin to Disease Modulation. PMID: 33556248

Profaci et al., 2020. The blood–brain barrier in health and disease: Important unanswered questions. PMID: 32211826

Psenicka et al., 2021. Connecting Neuroinflammation and Neurodegeneration in Multiple Sclerosis: Are Oligodendrocyte Precursor Cells a Nexus of Disease? PMID: 34234647

Reid and Kuipers, 2021. She Doesn’t Even Go Here: The Role of Inflammatory Astrocytes in CNS Disorders. PMID: 34539348

Rothhammer and Quintana, 2015. Control of autoimmune CNS inflammation by astrocytes. PMID: 26223505

Sandhu et al., 2021. Astroglial Glutamine Synthetase and the Pathogenesis of Mesial Temporal Lobe Epilepsy. PMID: 33927688

Singh, 2022. Astrocytic and microglial cells as the modulators of neuroinflammation in Alzheimer’s disease. PMID: 35978311

Smolders et al., 2018. Tissue-resident memory T cells populate the human brain. PMID: 30389931

Spiteri et al., 2022. Microglia and monocytes in inflammatory CNS disease: integrating phenotype and function. PMID: 34853891

Sun et al., 2022. Inflammation From Peripheral Organs to the Brain: How Does Systemic Inflammation Cause Neuroinflammation? PMID: 35783147

Takata et al., 2021. Blood-Brain Barrier Dysfunction Amplifies the Development of Neuroinflammation: Understanding of Cellular Events in Brain Microvascular Endothelial Cells for Prevention and Treatment of BBB Dysfunction. PMID: 34588955

Tang et al., 2021. Inflammatory neuropsychiatric disorders and COVID-19 neuroinflammation. PMID: 33926589

Tohidpour et al., 2017. Neuroinflammation and Infection: Molecular Mechanisms Associated with Dysfunction of Neurovascular Unit. PMID: 28676848

Troncoso-Escudero et al., 2018. Outside in: Unraveling the Role of Neuroinflammation in the Progression of Parkinson’s Disease. PMID: 30459700

Wang et al., 2022. Aquaporin-4 and Cognitive Disorders. PMID: 35111362

Wang et al., 2023. Brain endothelial CXCL12 attracts protective natural killer cells during ischemic stroke. PMID: 36631780

Webers et al., 2020. The role of innate immune responses and neuroinflammation in amyloid accumulation and progression of Alzheimer’s disease. PMID: 31654430

Xu et al., 2019. Basement membrane and blood-brain barrier. PMID: 31338215

Yang et al., 2019. Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. PMID: 30379577

Yeo et al., 2022. Potentials of Neuropeptides as Therapeutic Agents for Neurological Diseases. PMID: 35203552

Zhang et al., 2022. Endothelial caveolin-1 regulates cerebral thrombo-inflammation in acute ischemia/reperfusion injury. PMID: 36152520