1. Introduction
Alzheimer's disease (AD), the most prevalent form of dementia around the globe, develops over a period of several years, as a disease with both early and late onset and an increasing severity of cognitive decline [
1]. AD affects more than 6.5 million people over the age of 65 years in the United States alone [
2] and 50 million people worldwide [
3]. The United Nations estimates are that the global AD population will increase to nearly 152 million by 2050 [
3]. The main neuropathological characteristics of AD are the accumulation of amyloid-β (Aβ) peptide in brain parenchyma and perivascular regions as senile plaques, the presence of hyper-phosphorylated (phospho) tau protein as neurofibrillary tangles, and an accompanying synaptic and neuronal loss mainly in the hippocampal and cortical regions of the brain [
4]. The early clinical signs of AD manifest as neurobehavioral symptoms such as difficulty remembering names, events, and recent conversations, followed by impaired communication skills, disorientation, confusion, and lack of judgment. In the later stages of the disease, progressive symptoms include difficulty in speaking, swallowing, and walking, together with dementia [
5].
Understanding the molecular mechanisms of AD and targeting therapies for preclinical and clinical studies are indispensably aided by the use of transgenic animal models of AD that accurately recapitulate clinical pathology and cognitive decline with the symptomatic phase of AD [
6,
7]. The 5xFAD transgenic mouse model, developed in 2006, is still widely used because its amyloid plaque pathology is analogous to that found in human AD. 5xFAD mice overexpress humanized sequences of five AD-linked mutations: the Swedish (K670N/M671L), Florida (I716V), and London (V717I) mutations in APP and the M146L and L286V mutations in PSEN1 under the control of the neuron-specific thy1 promoter [
6]. The APP transgene includes the 5' untranslated region and thus contains a putative interleukin-1beta translational enhancer element [
6]. The most prominent feature of 5xFAD mice is that they present AD amyloid pathology and generate amyloid-β plaque formation with an accelerated rate of Aβ42 deposition in the cerebral cortex, subiculum, and deep cortical layers at an age as early as 2 months. This aggressive amyloid deposition of Aβ42 leads to memory deficits at 4 months and neuron loss, as indicated by a decrease in synaptic markers, such as synaptophysin, syntaxin, and postsynaptic density protein 95, at 9 months of age [
6,
8]. A significant loss of pyramidal neurons is evident in cortical Layer 5 and the subiculum from 9 months to 12 months of age [
6], together with increases in amyloid Aβ42 plaque numbers at 6, 9, and 12 months of age in brain regions adjacent to Layer 5 and the subiculum, such as CA1/2, CA3, the dentate gyrus, Layers 4 and 6 of the cortex, and the midbrain [
9]. 5xFAD mice show cardinal symptoms of AD, including cognitive deficits (learning and memory deficits) in Y-maze and Morris water maze test partially at 6 months and completely at 9 months of age [
10,
11] Some motor deficits, such as missing reflexes and rigidity of the fore and hind paws, also begin to appear at 9 months [
12].
AD has a decade-long clinically silent prodromal phase associated with subtle changes in biochemical and cellular functions, including neuronal dystrophy [
13], astrogliosis [
14] , early onset of neurovascular dysfunction [
15], and recruitment of microglia [
16]. A need to shift focus to this silent prodromal phase has recently been acknowledged to prevent early loss of synapses in the brain and for exploration of potential targets for therapeutic AD interventions [
17]. AD progression is significantly influenced by the neuroinflammatory state triggered by Aβ accumulation [
18]. The innate immune response controls the Aβ levels in the brain [
19]. Aβ can activate an inflammatory response involving escalations in microglial and astrocyte numbers for the uptake and clearance of Aβ from brains[
19], even though Aβ primarily produced by neurons [
20,
21]
Microglial cells are highly dynamic and surveille the brain. They migrate toward dense-core Aβ plaques, and microgliosis occurs in the vicinity of Aβ [
22]. Neuroinflammation is one of the major contributing factors to neurodegeneration in AD, and it starts as early as 2–3 months of age in 5xFAD mice [
6]. Inflammatory M1-phenotype microglia contribute to and signify this neuroinflammation [
23,
24,
25]. Microglia at the resting stage, as resident immune cells of the central nervous system, are constantly monitoring the microenvironment of the brain [
26]. Upon injury, microglia can be activated to assume the M1 phenotype, becoming amoeboid and highly phagocytic in nature and expressing CD68 [
27,
28,
29,
30], MHC-II, and/or OX-6 as markers of inflammation [
31,
32]. By contrast, alternative M2 microglia help in immuno-resolution and repair processes in an injury and have neuroprotective effects [
33,
34]. The switching between M1 and M2 phenotypes depends on the severity and stage of the disease [
35].
Astrocytes play a vital role in protecting and repairing neuronal damage in brains [
36,
37]. Astrocytes perform various essential functions including ion homeostasis, neurotransmitter buffering, secretion of neuroactive agents, synaptogenesis, and regulation of the blood brain barriers (BBBs). Astrocytes also use their end feet connections with the brain vasculature to vasodilate or constrict brain blood vessels to accommodate nutrient–waste exchange [
38] through a waste clearance system known as the glymphatic system [
38,
39]. Recent research has also suggested that astrocytes may play a role in the development and progression of AD [
40] by clearing the brain of amyloid-beta (Aβ), the peptide that forms the plaques that are a hallmark of AD. Astrocytes can normally phagocytose Aβ and are able to degrade it [
38]; however, this ability is impaired in AD astrocytes, leading to Aβ accumulation in the brain [
41]. Patients with AD have been reported to have reactive astrocytes in the hippocampus and entorhinal cortex, as indicated by a significantly higher mean number of glial fibrillary acidic protein positive (GFAP
+) cells in the hippocampus [
42,
43]. These reactive astrocytes have been shown to produce high levels of inflammatory molecules, such as TNF-α and interleukin-1β, which may contribute to neuronal damage. AD is also associated with an additional GFAP-high state astrocyte type, termed disease-associated astrocytes (DAAs) [
44], which exhibit an increased expression of pan-reactive and inflammatory A1 signatures in 5xFAD mice. DAAs are associated with diverse molecular pathways and are found positioned in proximity to Aβ plaques in AD-affected brains [
6,
45].
Reactive astrocytes display either detrimental or beneficial effects [
46], depending on their level of reactivity. In mouse models of infection-induced by LPS and in middle cerebral artery occlusion, quiescent (A0) astrocytes undergo a transformation into reactive forms through the upregulation or downregulation of specific genes [
47]. Gene expression profiles indicate the existence of reactive astrocytes in two distinct forms: A1 (inflammatory, neurotoxic) and A2 (inflammation-resolving, neuroprotective) [
48,
49]. Furthermore, the deposition of Aβ plaque triggers a response in A1 astrocytes, leading to an increased production of inflammatory mediators, such as chemokines, pro-inflammatory cytokines, and reactive oxygen species (ROS) that, in turn, damage neurons [
50]. The presence of these pro-inflammatory signals creates a feedback loop that further activates astrocytes, thereby perpetuating the chronic inflammatory environment that is instrumental in the development and progression of AD. The A1 astrocytes, found surrounding amyloid plaques, release pro-inflammatory cytokines, such as TNF-α, IL-1, and IL-6, which contribute to the pathogenesis and progression of AD [
51].
Recent studies have shed light on the molecular mechanisms underlying the formation and function of A2 astrocytes in AD. For instance, microglia-mediated signaling pathways have been implicated in the induction of A2 phenotype following brain injury [
52]. Additionally, nuclear factor IA, a transcription factor, has been identified as a molecular switch that triggers the transition of astrocytes into the neuroprotective A2 state [
53]. Furthermore, A2 astrocytes have been shown to upregulate various neuroprotective factors, including prokineticin-2, chitin-like 3, Frizzled class receptor 1, arginase 1, NF-E2-related factor 2 (Nrf2), pentraxin 3, sphingosine kinase 1, and transmembrane 4 L6 family member [
54]. These factors contribute to synaptic maintenance, growth, and protection against neurodegenerative processes associated with AD. Conversely, other mediators undergo upregulation or downregulation in these distinct forms of reactive astrocytes. A2 astrocytes are characterized by their ability to dampen neuroinflammation, promote synaptic repair, and enhance neuronal survival. Studies have highlighted the induction of A2 astrocytes in various neurodegenerative conditions, including traumatic brain injury [
55] and spinal cord injury [
56]. However, their role in AD is still an emerging area of research. In the present study we characterized the A1 and A2 astrocytes and found that MarL1 treatment shifts the polarization of A1 (inflammatory, neurotoxic) to A2 (inflammation-resolving, neuroprotective) astrocytes demonstrating neuroprotective properties of MarL1 in 5xFAD mice model of AD.
Another cell type potentially involved in AD is the neutrophil, the most abundant circulating leukocyte, which plays a critical role as first responder to inflammation [
57]. Recently, multiple reports have supported a role for the peripheral immune system, and especially neutrophils in AD [
58,
59]. Neutrophils can swarm to sites of inflammation but are not believed to cross the BBB [
60,
61,
62]. A recent report has indicated an increase in the neutrophil population in AD brains and in a murine APP/PS1 model of AD [
63]. Another study using 2-photon microscopy imaging reported that neutrophils infiltrate the brain parenchyma and migrate toward amyloid plaques in the 5xFAD mouse model, with a peak neutrophil infiltration occurring at 4 months of age in 5xFAD mice and at 6 months of age in the 3xTg-AD mouse model. This infiltration of neutrophils coincides with the onset of memory loss in cognitive tests [
58]. A review article has summarized the role of a neutrophil granule protein, CAP37, as well as neutrophil elastase and cathepsin G, in neuroinflammation, with an emphasis on AD [
64]. Analogous to the conventional categorization of macrophages or microglia into two major types, M1 and M2 phenotypes, neutrophils have been into categorized into pro-inflammatory N1 [
65] and anti-inflammatory N2 subpopulations [
66]. The N1 and N2 neutrophil populations reportedly have distinct transcriptomic profiles and functions [
65]. Pro-inflammatory N1 neutrophils exhibit increased production of inflammatory cytokines/chemokines with elevated levels of ROS and NO, increased activity of protein and matrix-degrading enzymes, amplified chemotactic responses, and enhanced phosphorylated forms of ERK1/2 and p65 signaling molecules associated with an inflammatory phenotype. By contrast, N2 neutrophils demonstrate increased expression of CD206, Ym1 and Arg1, and have similar ROS and NO levels and equivalent chemotactic response to unstimulated controls but no changes in ERK1/2 and p65 signaling molecules [
65]. We have determined the possible occurrence of N1 and N2 neutrophils in brains of 5xFAD mice with and without treatment as well as of wildtype control mice in this study.
The recognition of the importance of neuroinflammation in AD has led to increased focus on specialized pro-resolving mediators (SPMs) that effectively promote the resolution of inflammation [
67]. One class of these compounds are the maresins, first discovered in 2009 by Serhan et al [
68]. Maresins [
68,
69] and the related maresin -like (MarL) mediators [
70] are derived from essential ω-3 docosahexaenoic acid (DHA) by the action of endogenous enzymatic systems [12- or 15-lipoxgenase (LO) for maresins; and 12/15-LO plus a cytochrome P450 CYP4F3 for MarL]. Maresin-1 (7
R,14
S-dihydroxy-4
Z,8
E,10
E,12
Z,16
Z,19
Z-DHA) reduces neuroinflammation, mitochondrial damage, and neuronal death, while enhancing neural functional recovery, macrophage phagocytosis of Aβ, and the switch in macrophage phenotype to the anti-inflammatory M2 type [
71,
72]. Maresin 1 also ameliorates the physiopathology of experimental autoimmune encephalomyelitis [
73] and promotes neuroprotection and functional recovery after spinal cord injury by resolving inflammation [
71]. Maresin 1 treatment has recently been reported to ameliorate Aβ42-induced cognitive decline and neuroinflammation in C57BL/6 mice [
74]. Similarly, the intranasal instillation of maresin 1 in combination with resolvins and neuroprotectin/protectin D1 ameliorated memory deficits and gamma oscillation deficits, concurrently with a dramatic decrease in microglial activation in the App
NL-G-F/NL-G-F mouse model for AD [
75]. Despite the protective effects of maresin 1 on the brain in 5xFAD transgenic mice, the underlying mechanisms remain abstruse.
By contrast, the maresin-like compound MarL1 (14
S,22-dihydroxy-docosa-4
Z,7
Z,10
Z,12
E,16
Z,19
Z-hexaenoic acid) promotes the production of the regenerative angiogenic growth factor HGF and attenuates the production of the pro-inflammatory cytokine TNFα
in vitro [
70] in macrophages. MarL1 also enhances the macrophage-induced promotion of migration of epithelial cells and fibroblasts from scratch-wounded monolayer cultures, as well as stem cell transmigration [
70]. Despite the growing evidence for pro-resolving and neuroprotective effects of maresin 1 and MarLs, the underlying mechanisms require further investigation for their therapeutic development. In the present study, we have hypothesized that long-term administration of MarL1 administration could mitigate AD-related brain neuropathogenesis by curbing neuroinflammation and neurodegeneration.
2. Materials and Methods
2.1. Animals
The Louisiana State University Health Science Center (LSUHSC) IACUC committee approved all the animal procedures, which are consistent with American Veterinary Medical Association guidelines. Mice were maintained at LSUHSC at a controlled temperature of 25 ± 2°C and 50%–65% humidity with a fixed 12:12 h light-dark cycle. We used a widely used model of AD—5x familial Alzheimer’s Disease (5xFAD) transgenic mice (Jackson Laboratory, Bar Harbor, ME, USA) with a C57BL/6J genetic background (
MMRRC Strain #034840-JAX)—and compared them with wildtype (WT) control mice (C57BL/6J) [
76].
2.2. Intranasal Treatment with Maresin-like 1
Male mice at the age of 1.5 months were randomly selected and equally divided into the following three groups: 1) WT (C57BL/6J mice), 2) 5xFAD mice treated with vehicle, and 3) 5xFAD mice treated with MarL1 plus vehicle. The MarL1 or vehicle was administered by the intranasal route 3 times per week to each mouse from 1.5 months to 9 months of age. Briefly, each mouse was temporarily immobilized for about 60 seconds by a slight anesthetization using isoflurane inhalation. During the immobilization, the mouse was held in the supine position and its nostrils were then instilled with 3 µL/nostril of MarL1 (100 ng per mouse for each administration) dissolved in the vehicle (0.05% dimethyl sulfoxide [DMSO] in sterile 0.9% saline) or with 3 µL the vehicle alone. The mouse was held at the same position for 20 more seconds to ensure liquid intranasal intake [
75,
77,
78,
79]. The mice were sacrificed at the age of 12.5 months.
2.3. Harvesting of Murine Brains for Immunohistology
The mice were anesthetized with 5% isoflurane and then transcardially perfused with 4% paraformaldehyde (PFA). The color change in the liver from a deep red to a paler shade was used as an indicator of a sufficient perfusion. The mice were then decapitated using a pair of sharp scissors and the brains were quickly removed from the cranium and immersed in 4% paraformaldehyde for fixing overnight at 4°C. Sections from the cortex and hippocampus regions were chosen for immunohistochemistry. After 24 h, the brains were transferred to 15% for 12 hours, then to 30% sucrose solution. After the brain tissue finally sank in the 30% sucrose solution, brain tissue blocks were made using optimal cutting temperature compound (OCT) liquid and cryo-molds for cryo-sectioning. Coronal section of brains were cut rostral-to-caudal serially at 20 μm thickness on a HM550 cryostat (Microm-HM 550, Thermo Fisher Scientific), mounted on Superfrost Plus glass slides (VWR, Radnor, PA, USA), and stored.
2.4. Immunofluorescence Staining
The mounted brain sections (containing cortex and hippocampus) were washed twice in phosphate-buffered saline (PBS), followed by two washes in PBS-T (PBS containing 0.05% Triton-X). The sections were then incubated in blocking buffer (1% BSA+0.5% Trition-X+0.02% Tween-20 in 1X PBS) at room temperature for 60 min. The sections were then treated with the following antibodies: NeuN (1:500, rabbit, cell signaling-12943S), amyloid-β (MOAB-2, 1:500, mouse host, monoclonal 6C3, Millipore-MABN254), Gr-1 (Ly-6G/Ly-6C monoclonal antibody (1:200, rat host, Invitrogen), ionized calcium-binding adapter molecule-1 (Iba-1, 1:500, rabbit, Fujifilm Wako-019-19741), iNOs (1:200, rat, Invitrogen eBioscience), Arg-1 (1:500, goat, Abcam-ab60176), GFAP (1:500, rabbit, Sigma-G9269), TNF-α (1:500, goat, Invitrogen-PA5-46945), anti-choline acetyltransferase (1:500, ChAT, goat, Millipore-sigma-AB144P), cleaved caspase-3 (cleaved-caspase-3, 1:500, rabbit, Cell Signaling-9661) antibody, CD68 (1:500, mouse, Santa Cruz Biotechnology-70761), C3 (1:500, rabbit, Invitrogen-PA5-21349), S100A10 (reconstituted in sterile PBS, 0.2 mg/mL, mouse, Biotechne-AF2377), and claudin-5 (1:500, mouse, Santa Cruz Biotechnology, sc-374221). The sections were then incubated with appropriate secondary antibodies (Alexa Fluor 488, 568, or 594; Invitrogen) compatible with the aforementioned primary antibodies, followed by the incubation with DAPI (1:10,000 dilution). A total of three to four sections of brain per slide from each of five to eight mice per group were used for the histological study. Co-stained (yellow-colored) cells by two protein/peptide biomarkers (green-colored and red-colored) were quantified by cell counting or Pearson’s coefficients obtained in ImageJ analysis (1.54J National Institutes of Health, Bethesda, MD).
2.5. Image Quantification
After immunofluorescence staining, fluorescent images were captured on ECHO Revolve fluorescent microscope using 4X, 10X, 20X, and 40X objectives, and mean intensity of fluorescence, number of immunoreactive cells, were quantified using the ImageJ software. The results were expressed in mean fluorescence intensity (MFI) as we conducted previously [
80]. The counting of co-localized immune-positive cells for two markers was performed manually using the cell counter application of ImageJ. All quantifications were performed blinded to the experimental groups with the help of the ImageJ image analysis software.
2.6. Thioflavin S Staining and Analysis of Plaques
Thioflavin S (Sigma-Aldrich, T1892) is a fluorescent staining used for visualizing Aβ plaques in the brain [
6]. The thioflavin S stained Aβ plaques appear bright green by fluorescence microscopy, making them easily identified. Briefly, brain sections (20 μm thickness) were incubated in 1% thioflavin S solution in deionized water for 10 min and washed with running water carefully for 5 min, then incubated in 1% acetic acid for 15 min and wash again in running water. The slides were dried by placing them on paper towels for few minutes and then dehydrated in an alcohol series of 70%, 80%, 95%, 100% ethanol, transferred to xylene, and then mounted with Permount mounting medium and dried overnight in the dark. Thioflavin-S-positive plaques were determined from the images taken by Discover-ECHO Revolve fluorescence microscope in a single plain at 4× magnification. The images were subjected to threshold processing (Otsu) using ImageJ, and the total number of plaques based on size (less than and greater than 100 μm) were analyzed in the cortical and hippocampal regions. For each animal, 6 fields from the cortex and from the hippocampus were imaged and analyzed.
2.7. Statistical Analysis
Results were expressed in Mean ± standard error of mean (SEM). Kruskal–Wallis one-way analysis of variance (ANOVA, non-parametric test) followed by Tukey’s multiple comparison post-hoc test was used to determine the statistical significance of differences between mouse groups of wildtype, 5xFAD, and 5xFAD + MarL1 (*p < 0.05, **p < 0.01, and ***p < 0.001). For all experiments at least three replicates were performed. The definition of the significance for various p values are described in the figure legends, together with the number of biological replicates (n) for each experiment. GraphPad Prism 9.0 (GraphPad, Boston, MA) was used for graphs and statistical analyses.
4. Discussion
AD is characterized as a progressive neurodegenerative disorder and the accumulation of amyloid beta plaques and tau tangles in the brain are considered to be key pathological features of AD [
123]; however, recent studies have shown that AD has a multifactorial etiology and that neuroinflammation plays a central role in its etiopathogenesis, owing to its capacity to exacerbate Aβ and Tau pathologies [
123,
124,
125]. Evidence of increased microglia activation (inflammation) in the brains of AD patients with elevated levels of pro-inflammatory cytokines in serum and in post-mortem brains supports the link between neuroinflammation and AD [
126,
127]. In the present study, we examined the effect of MarL1 on the extent of inflammation in brains of transgenic 5xFAD mice.
We evaluated the effect of intranasal instillation of the MarL1 mediator from the age of 1.5 to 9 months in 5xFAD mice. Intranasal instillation of drugs is well recognized to partly bypass the BBB to deliver drugs to brains more efficiently than
ip or
iv methods, thereby increasing drug bioavailability in the brain, while also delivering drugs noninvasively and to the blood circulation as well [
77,
128,
129,
130]. At the age of 12.5 months, 5xFAD mice show a significant reduction in levels of oligomeric Aβ42 and Aβ plaques in cortex and hippocampal regions of the brain and in the loss of NeuN
+ neurons in CA3 and dentate gyrus regions of hippocampus (
Figure 1). Quantification of the Aβ plaque numbers based on sizes greater than and less than 100 μm
2 area [
83,
84,
85] using thioflavin-S staining revealed a marked reduction in the number of Aβ plaques following MarL1 treatment of 5xFAD mice (
Figure S1). The neuroprotective effect of MarL1 was mediated by restoring the cholinergic neurons in striatum and decreasing the apoptotic cleaved caspase-3
+ neurons in brain. Other studies have shown increased activation of cleaved caspase-3 in the hippocampus of AD patients and significant increases in synaptic pre-caspase-3 and active caspase-3 (i.e., cleaved caspase-3) expression levels in the postsynaptic density fractions [
131]. These findings suggest that MarL1 has a neuroprotective effect in the brain.
Neuroinflammation is thought to play an essential role in neurodegeneration by contributing to neuronal damage and synaptic loss, with microglia and astrocytes as the key players [
132,
133,
134]. Microglia, as the resident phagocytes in the brain, constantly use their highly motile processes to surveille brain regions for the presence of pathogens and cellular debris and simultaneously provide factors that support tissue maintenance [
134]. The M2 phenotype microglia are also involved in the protection and remodeling of synapses for proper maintenance of the plasticity of neuronal circuits and release of trophic factors, including brain-derived neurotrophic factors, which contribute to memory formation [
135,
136]. Upon activation due to pathological triggers, such as neuronal death or protein aggregates, microglia change phenotypically to an amoeboid state and begin to migrate to the lesion, where they initiate an innate immune response [
35]. Activated microglia (M1) are “pro-inflammatory” in nature and increase the expressions of markers involved in microglial activation, such as CD68 [
27,
28,
29,
30], CD86, CD45, CX3CR1, CD11b, and MHC-II [
137].
Histological examinations of AD brains show that microglial cells are found in close association with Aβ deposits and are associated with dense-core plaques [
138,
139,
140]; however, fewer microglial cells are found in the vicinity of the diffuse Aβ deposits in the cerebral cortex of AD patients [
141,
142]. The number of microglia and their size directly increases in proportion to plaque dimension, and Aβ deposition has been reported to recruit a microglial population to allow for the accumulation of these cells at the periphery of amyloid deposits [
143,
144]. Microglia have the ability to attach themselves to soluble oligomers and fibrils of Aβ by means of various receptors, such as CD36, α6β1 integrin, CD47, CD14, and class A scavenger receptor A1, as well as toll-like receptors (TLR2, TLR4, TLR6, and TLR9) [
143,
144]. Our results demonstrate enhanced microglial phagocytosis of Aβ plaques (
Figure S2), and we found microglia in clusters in 5xFAD mice. These aggregates showed that microglial accumulation and proliferation in the brain could cause neuroinflammation, while MarL1 treatment reduced the Aβ accumulation thereby decreasing the microglial activation.
Enhanced microglial activation leads to increases in the expression of pro-inflammatory markers, such as interleukin-1β, TNF-α, and iNOs, thereby exacerbating neuroinflammation [
126,
145]. Our data demonstrate that MarL1 treatment attenuated microglial activation by decreasing the population of Iba-1 positive microglia and CD68 expression in the hippocampus (CA1, dentate gyrus) of the 5xFAD mouse brain. We phenotypically characterized microglia and calculated the number of microglia in different states as: 1) ramified, 2) partially- ramified, 3) partially-amoeboid, 4) and fully-amoeboid microglia [
146,
147]. We found that MarL1 reduced the level of amoeboid microglia, suggesting its inflammation-resolving and anti-inflammatory properties are a consequence of suppression of microglial activation (
Figure 3).
Specialized pro-resolving lipid mediators are reported to modulate immunity and inflammation by resolving inflammation by triggering a biochemical paradigm shift commonly referred to as the “lipid mediator class switch” and skewing the M1/M2 macrophage balance toward the anti-inflammatory M2 phenotype, with replacement of injured cells and restoration of the normal functions of tissues [
67]. Consistent with these reports, MarL1 treatment caused a shift in the M1/M2 population in brain as indicated by a surging Arg-1
+ microglial population in the brain (
Figure 4). These finding suggest that MarL1 is a potent immunoresolvent that is effective in brains
We found hypertrophy of astrocytes interacting with Aβ plaques in hippocampus in 5xFAD mice (
Figure S2), as well as increased expression of TNFα in astrocytes in 5xFAD mice (
Figure 5). These responses suggest that the mutations present in 5xFAD mice disrupt the mechanisms that would normally protect against and repair neuronal damage in the brain—functions that are typically carried out by astrocytes [
36,
37]. The MarL1 treatment attenuated the GFAP-high state astrocytes in brains of 5xFAD mice and reduced Aβ accumulation. Our characterization of A1 and A2 astrocytes in the 5xFAD mouse model of AD revealed that MarL1 treatment shifts the polarization of A1 (neurotoxic) astrocytes to the A2 (neuroprotective) form, thereby demonstrating a potential reason for the neuroprotective action of MarL1 (
Figure 6 and
Figure 7). These results demonstrate that MarL1 mitigates the astrocyte hypertrophy and inflammatory cytokine production and increases Aβ clearance in brains.
In this study we explored the concept of neutrophil migration in brain parenchyma. Here, we observed the phenomenon of “neutrophil swarming” [
148] in the cortex, as shown in
Figure 8, as evidence of neutrophil aggregation in the 5xFAD mouse brain. This is the first immunohistochemical report of neutrophil swarming in the brain cortex in the literature. Co-saining of Gr-1 (a marker for neutrophils) and claudin-5 (a marker for vasculature) showed that Gr-1
+ neutrophils migrated into the brain and formed swarms, as neutrophils are nonresident cells in brains. We could not find neutrophil swarms in the brains of MarL1-treated 5xFAD mice, although neutrophils aggregate were evident than wildtype mice. No swarms were detected in the brains of the wildtype littermates. These findings suggests that neutrophil infiltration into brains could contribute to AD pathogenesis since neutrophils produce reactive oxygen species and degradation enzymes that can cause neuroinflammation and neurodegeneration[
57,
58,
59]. Furthermore, our findings revealed that MarL1 treatment inhibited the AD-linked neutrophil infiltration and swarm in the brains of 5xFAD mice.
We also characterized neutrophils into pro-inflammatory N1 [
65] and anti-inflammatory N2 subpopulations [
66]. The N1 and N2 neutrophil populations reportedly have distinct transcriptomic profiles and functions [
65]. Pro-inflammatory N1 neutrophils exhibit increased production of inflammatory cytokines/chemokines with elevated levels of ROS and NO, increased activity of protein and matrix-degrading enzymes, amplified chemotactic responses, and enhanced phosphorylated forms of ERK1/2 and p65 signaling molecules associated with an inflammatory phenotype. By contrast, N2 neutrophils demonstrate increased expression of CD206, Ym1 and Arg1, and have similar ROS and NO levels and equivalent chemotactic response to unstimulated controls but no changes in ERK1/2 and p65 signaling molecules [
65]. In our study, we found that Gr-1
+iNOS
+ N1 neutrophils increased in 5xFAD mice. We also found that MarL1 treatment dampens the elevation of N1 neutrophils in brain (
Figure 9). Furthermore, Gr-1
+Arg1
+ N2 neutrophils were amplified with MarL1 treatment and were lower in 5xFAD mice than in wildtype controls (
Figure 10). These results demonstrate that chronic MarL1 treatment polarizes a shift from N1 to N2 in 5xFAD mice, suggesting resolution of inflammation in the brain.
Taken together, our results provide the first evidence that MarL1 was effective in inhibiting Aβ pathology in brain. However, the 5xFAD mouse model is an aggressive early onset transgenic AD model [
8,
149,
150,
151,
152], therefore, the protection effects of MarL1 need to be explored using less aggressive AD models, including late-onset AD models [
152,
153]. Furthermore, the different doses of MarL1 and their safety should be evaluated for future clinical studies. Further research is needed to fully understand the mechanisms involved in the neuroprotection imparted by MarL1 in AD. MarL1-specific receptor targeted research could be a promising strategy for developing new treatments for this debilitating disease.
Author Contributions
Conceptualization, S.H.; Methodology, S.H., Y.L., P.S., Y.K., Y.Z., W.L; Validation, P.S., Y.L., S.S., Y.K., Y.Z., N.L., A.-R.M. and S.H.; Formal analysis P.S., Y.L., N.L. and S.H.; Investigation, P.S., Y.L., Y.K., Y.Z., N.L. and S.H.; Resources, Y.K., Y.Z., W.L., and S.H.; Data curation, P.S., Y.L., Y.K., Y.Z., N.L. and S.H.; Writing - original draft, P.S., Y.L., S.S. and S.H.; Writing - review & editing, P.S., Y.L., S.S., Y.K., Y.Z., N.L., A.-R.M. and S.H.; Visualization, P.S., Y.L. and S.H.; Supervision: Y.K. and S.H.; Project administration, Y.K. and S.H.; Funding acquisition, Y.K. and S.H.
Figure 1.
MarL1 treatment ameliorated AD neuropathology in brains of 5xFAD mice. (A) Immunostaining of NeuN (green) and Amyloid-β1-42 (red) in CA3 and dentate gyrus (DG) of hippocampus. White arrows mark some Aβ1-42 deposition in hippocampal regions. Scale bar: 180 μm. (B) Quantification of NeuN+ and Amyloid-β1-42+ staining intensities of hippocampus (Mean fluorescence intensity, MFI). Data are Means ± SEM. ***p < 0.001.
Figure 1.
MarL1 treatment ameliorated AD neuropathology in brains of 5xFAD mice. (A) Immunostaining of NeuN (green) and Amyloid-β1-42 (red) in CA3 and dentate gyrus (DG) of hippocampus. White arrows mark some Aβ1-42 deposition in hippocampal regions. Scale bar: 180 μm. (B) Quantification of NeuN+ and Amyloid-β1-42+ staining intensities of hippocampus (Mean fluorescence intensity, MFI). Data are Means ± SEM. ***p < 0.001.
Figure 2.
MarL1 protected cholinergic neurons (ChAT+) and inhibited apoptotic cleaved-caspase-3 activity in brains of 5xFAD mice. (A) Immunostaining of ChAT (green) and cleaved-caspase-3 (red) in striatum (Panels a-c: 10X magnification; scale bar: 180 µm. White arrows mark cleaved-caspase-3+ cholinergic neurons in zoomed-in images (Panels d-f). Scale bar: 35 µm. (B) Quantification of ChAT and caspase-3 in striatum. Left: Mean fluorescence intensity MFI for ChAT+; middle: MFI for cleaved-caspase-3+; right: count of cells stained positive for both ChAT and cleaved-caspase-3. Data are Means ± SEM. *** p < 0.001, and *p < 0.05.
Figure 2.
MarL1 protected cholinergic neurons (ChAT+) and inhibited apoptotic cleaved-caspase-3 activity in brains of 5xFAD mice. (A) Immunostaining of ChAT (green) and cleaved-caspase-3 (red) in striatum (Panels a-c: 10X magnification; scale bar: 180 µm. White arrows mark cleaved-caspase-3+ cholinergic neurons in zoomed-in images (Panels d-f). Scale bar: 35 µm. (B) Quantification of ChAT and caspase-3 in striatum. Left: Mean fluorescence intensity MFI for ChAT+; middle: MFI for cleaved-caspase-3+; right: count of cells stained positive for both ChAT and cleaved-caspase-3. Data are Means ± SEM. *** p < 0.001, and *p < 0.05.
Figure 3.
MarL1 suppressed pro-inflammatory M1 phenotype polarization of microglia in brains of 5xFAD mice. (A) Immunostaining of microglia with Iba-1 (green) and CD68 (red) in CA1 region of hippocampus from 5xFAD transgenic mice (Panels a-c: 10X magnification; scale bar: 180 µm. Panels d-f: zoomed-in images; scale bar: 30 µm). White arrows mark Iba-1+CD68+ microglia. (B) Quantification of Iba-1+ and CD68+ in hippocampus. Left: mean fluorescence intensity MFI of Iba-1+; middle: MFI of CD68+; right: count of microglia stained positive for both Iba-1+ and CD68+; (C) Quantification of microglia based on phenotype characterization (ramified, partially ramified, partially amoeboid, amoeboid) in hippocampus. Data are Means ± SEM. *** p < 0.001, ** p <0.01, and * p < 0.05.
Figure 3.
MarL1 suppressed pro-inflammatory M1 phenotype polarization of microglia in brains of 5xFAD mice. (A) Immunostaining of microglia with Iba-1 (green) and CD68 (red) in CA1 region of hippocampus from 5xFAD transgenic mice (Panels a-c: 10X magnification; scale bar: 180 µm. Panels d-f: zoomed-in images; scale bar: 30 µm). White arrows mark Iba-1+CD68+ microglia. (B) Quantification of Iba-1+ and CD68+ in hippocampus. Left: mean fluorescence intensity MFI of Iba-1+; middle: MFI of CD68+; right: count of microglia stained positive for both Iba-1+ and CD68+; (C) Quantification of microglia based on phenotype characterization (ramified, partially ramified, partially amoeboid, amoeboid) in hippocampus. Data are Means ± SEM. *** p < 0.001, ** p <0.01, and * p < 0.05.
Figure 4.
MarL1 promoted anti-inflammatory M2 phenotype polarization of microglia in brains of 5xFAD mice. (A) Immunostaining of microglia with Iba-1 (green) and Arg1 (red) in cortex (Panels a-c: 20X magnification; scale bar: 90 µm. Panels d-f: zoomed-in images; scale bar: 30 µm). White arrows mark Iba1+Arg1+ microglia. Red arrows mark microglial aggregation in cortex of 5xFAD mice. (B) Quantification of Iba-1 and Arg1 in cortex. Left: mean fluorescence intensity MFI of Iba1+; middle: MFI of Arg1+; right: count of microglia stained positive for both Iba1 and Arg1 in cortex. Data are Means ± SEM. *** p < 0.001, and ** p < 0.01.
Figure 4.
MarL1 promoted anti-inflammatory M2 phenotype polarization of microglia in brains of 5xFAD mice. (A) Immunostaining of microglia with Iba-1 (green) and Arg1 (red) in cortex (Panels a-c: 20X magnification; scale bar: 90 µm. Panels d-f: zoomed-in images; scale bar: 30 µm). White arrows mark Iba1+Arg1+ microglia. Red arrows mark microglial aggregation in cortex of 5xFAD mice. (B) Quantification of Iba-1 and Arg1 in cortex. Left: mean fluorescence intensity MFI of Iba1+; middle: MFI of Arg1+; right: count of microglia stained positive for both Iba1 and Arg1 in cortex. Data are Means ± SEM. *** p < 0.001, and ** p < 0.01.
Figure 5.
MarL1 treatment mitigated AD-augmented TNF-α expression and astrogliosis in brains of 5xFAD mice. (A) Immunostaining of GFAP (green) and TNF-α (red) in cortex. White arrows mark GFAP+TNF-α+ astrocytes in cortex (Panels a-c: 10X magnification; scale bar: 180 µm. Panels d-f: zoomed-in images; scale bar: 30 µm). (B) Quantification of GFAP+ and TNF-α+ in mean fluorescence intensity MFI in cortex. Data are Means ± SEM. *** p < 0.001, and ** p < 0.01.
Figure 5.
MarL1 treatment mitigated AD-augmented TNF-α expression and astrogliosis in brains of 5xFAD mice. (A) Immunostaining of GFAP (green) and TNF-α (red) in cortex. White arrows mark GFAP+TNF-α+ astrocytes in cortex (Panels a-c: 10X magnification; scale bar: 180 µm. Panels d-f: zoomed-in images; scale bar: 30 µm). (B) Quantification of GFAP+ and TNF-α+ in mean fluorescence intensity MFI in cortex. Data are Means ± SEM. *** p < 0.001, and ** p < 0.01.
Figure 6.
MarL1 treatment suppressed AD-associated pro-inflammatory A1 phenotypic polarization of astrocytes in brains of 5xFAD mice. (A) Immunostaining of GFAP (green) and Complement C3 (red) in dentate gyrus of hippocampus. Panels a-c: 20X magnification; scale bar: 90 µm. White arrows mark GFAP and complement C3 astrocytes co-expression in zoomed-in images (panels d-f); scale bar: 30 µm. (B) Quantification of GFAP+ and C3+ in mean fluorescence intensity MFI in dentate gyri. Left: MFI of GFAP+; middle: MFI of C3+; right: Pearson’s coefficient for quantification of co-localization of GFAP and C3. Data are Means ± SEM. *** p < 0.001, and ** p < 0.01.
Figure 6.
MarL1 treatment suppressed AD-associated pro-inflammatory A1 phenotypic polarization of astrocytes in brains of 5xFAD mice. (A) Immunostaining of GFAP (green) and Complement C3 (red) in dentate gyrus of hippocampus. Panels a-c: 20X magnification; scale bar: 90 µm. White arrows mark GFAP and complement C3 astrocytes co-expression in zoomed-in images (panels d-f); scale bar: 30 µm. (B) Quantification of GFAP+ and C3+ in mean fluorescence intensity MFI in dentate gyri. Left: MFI of GFAP+; middle: MFI of C3+; right: Pearson’s coefficient for quantification of co-localization of GFAP and C3. Data are Means ± SEM. *** p < 0.001, and ** p < 0.01.
Figure 7.
MarL1 treatment reversed AD-associated diminish of anti-inflammatory A2 phenotypic polarization of astrocytes in brains of 5xFAD mice. (A) Immunostaining of GFAP (green) and A2 marker S100A10 (red) in dentate gyrus of hippocampus. White arrows mark GFAP and S100A10 positive astrocytes in dentate gyri (Panels a-c: 20X magnification; scale bar, 90 µm. Panels d-e: zoomed-in images; scale bar: 30 µm). (B) Quantification of GFAP+ and S100A10+ in mean fluorescence intensity MFI in dentate gyri. Left: MFI of GFAP+; middle: MFI of s100A10+; right: Pearson’s coefficient for quantification of co-localization of GFAP and s100A10. Data are Means ± SEM. *** p < 0.001, and *p < 0.05.
Figure 7.
MarL1 treatment reversed AD-associated diminish of anti-inflammatory A2 phenotypic polarization of astrocytes in brains of 5xFAD mice. (A) Immunostaining of GFAP (green) and A2 marker S100A10 (red) in dentate gyrus of hippocampus. White arrows mark GFAP and S100A10 positive astrocytes in dentate gyri (Panels a-c: 20X magnification; scale bar, 90 µm. Panels d-e: zoomed-in images; scale bar: 30 µm). (B) Quantification of GFAP+ and S100A10+ in mean fluorescence intensity MFI in dentate gyri. Left: MFI of GFAP+; middle: MFI of s100A10+; right: Pearson’s coefficient for quantification of co-localization of GFAP and s100A10. Data are Means ± SEM. *** p < 0.001, and *p < 0.05.
Figure 8.
MarL1 attenuated the AD-associated compromise of blood–brain-barrier tight-junctions as well as neutrophil infiltration into brains of 5xFAD mice. (A) Immunostaining of Gr-1 (green) for neutrophils and Claudin-5 (red) for tight-junctions of the vasculatures in cortex. Panels a-c show images from cortex (4X magnification; scale bar: 460 µm). Panels d-f show zoomed-in images; scale bar: 65 µm. White arrows mark some Gr-1+ cells outside the vasculature in parenchyma in zoomed-in images. Yellow arrows mark some claudin-5+ vasculatures. Neutrophil swarming is evident in panels b and e. (B) Quantification of Gr-1+ and Claudin-5+ in MFI in cortex. Data are Means ± SEM. *** p < 0.001, and *p < 0.05.
Figure 8.
MarL1 attenuated the AD-associated compromise of blood–brain-barrier tight-junctions as well as neutrophil infiltration into brains of 5xFAD mice. (A) Immunostaining of Gr-1 (green) for neutrophils and Claudin-5 (red) for tight-junctions of the vasculatures in cortex. Panels a-c show images from cortex (4X magnification; scale bar: 460 µm). Panels d-f show zoomed-in images; scale bar: 65 µm. White arrows mark some Gr-1+ cells outside the vasculature in parenchyma in zoomed-in images. Yellow arrows mark some claudin-5+ vasculatures. Neutrophil swarming is evident in panels b and e. (B) Quantification of Gr-1+ and Claudin-5+ in MFI in cortex. Data are Means ± SEM. *** p < 0.001, and *p < 0.05.
Figure 9.
MarL1 treatment suppressed pro-inflammatory N1 polarization of neutrophils infiltrated into AD-pathogenic brains in 5xFAD mice. (A) Immunostaining of Gr-1 (green) for neutrophils and iNOs (red), an inflammatory marker. Panels a-c show hippocampus (4X magnification, scale bar: 460 µm). Panels d-f show zoomed-in images; scale bar: 40 µm. White arrows mark some Gr-1+iNOs+ cells and yellow arrows mark only Gr-1 positive cells in zoomed-in panels. (B) Quantification of Gr-1+ and iNOs+ in hippocampus. Left: MFI of Gr-1+; middle: MFI of iNOs+; right: Pearson’s coefficient for quantification of co-localization of Gr-1 and iNOs. *** p < 0.001, ** p <0.01, and *p < 0.05.
Figure 9.
MarL1 treatment suppressed pro-inflammatory N1 polarization of neutrophils infiltrated into AD-pathogenic brains in 5xFAD mice. (A) Immunostaining of Gr-1 (green) for neutrophils and iNOs (red), an inflammatory marker. Panels a-c show hippocampus (4X magnification, scale bar: 460 µm). Panels d-f show zoomed-in images; scale bar: 40 µm. White arrows mark some Gr-1+iNOs+ cells and yellow arrows mark only Gr-1 positive cells in zoomed-in panels. (B) Quantification of Gr-1+ and iNOs+ in hippocampus. Left: MFI of Gr-1+; middle: MFI of iNOs+; right: Pearson’s coefficient for quantification of co-localization of Gr-1 and iNOs. *** p < 0.001, ** p <0.01, and *p < 0.05.
Figure 10.
MarL1 treatment induced anti-inflammatory N2 phenotypic polarization of neutrophils infiltrated into AD-pathogenic brains in 5xFAD mice. (A) Immunostaining of Gr-1 (green) for neutrophils and Arg1(red), an anti-inflammatory marker. Panels a-c show hippocampus (4X magnification, scale bar: 460 µm). Panels d-f show zoomed-in images; scale bar: 40 µm. White arrows mark Gr-1+ cells and yellow arrows mark Gr-1+Arg1+ cells in zoomed-in panels. (B) Quantification of Gr-1+ and Arg1+ in hippocampus. Left: MFI of Gr-1+; middle: MFI of Arg1+; right: Pearson’s coefficient for quantification of co-localization of Gr-1 and Arg1. Data are Means ± SEM. *** p < 0.001, ** p < 0.01, and *p < 0.05.
Figure 10.
MarL1 treatment induced anti-inflammatory N2 phenotypic polarization of neutrophils infiltrated into AD-pathogenic brains in 5xFAD mice. (A) Immunostaining of Gr-1 (green) for neutrophils and Arg1(red), an anti-inflammatory marker. Panels a-c show hippocampus (4X magnification, scale bar: 460 µm). Panels d-f show zoomed-in images; scale bar: 40 µm. White arrows mark Gr-1+ cells and yellow arrows mark Gr-1+Arg1+ cells in zoomed-in panels. (B) Quantification of Gr-1+ and Arg1+ in hippocampus. Left: MFI of Gr-1+; middle: MFI of Arg1+; right: Pearson’s coefficient for quantification of co-localization of Gr-1 and Arg1. Data are Means ± SEM. *** p < 0.001, ** p < 0.01, and *p < 0.05.