Skip to main content
You are viewing the companion.
VIEW THE ORIGINAL ARTICLE
You are viewing the companion.
VIEW THE ORIGINAL ARTICLE
You are viewing the companion.
VIEW THE ORIGINAL ARTICLE
You are viewing the companion.
VIEW THE ORIGINAL ARTICLE
You are viewing the companion.
VIEW THE ORIGINAL ARTICLE
You are viewing the companion.
VIEW THE ORIGINAL ARTICLE
This article has a companion.
VIEW THE COMPANION
This article has a companion.
VIEW THE COMPANION
This article has a companion.
VIEW THE COMPANION
This article has a companion.
VIEW THE COMPANION
This article has a companion.
VIEW THE COMPANION
This article has a companion.
VIEW THE COMPANION

Abstract

Stroke is one of the leading causes of death worldwide and currently only few therapeutic options are available. Stroke is a sexually dimorphic disease contributing to the difficulty in finding efficient treatments. Poststroke neuroinflammation is geared largely by brain microglia and infiltrating peripheral immune cells and largely contributes to sex differences in the outcome of stroke. Microglia, since very early in the development, are sexually divergent, imprinting specific sex-related features. The diversity in terms of microglial density, morphology, and transcriptomic and proteomic profiles between sexes remains in the adulthood and is likely to contribute to the observed sex-differences on the postischemic inflammation. The impact of sexual hormones is fundamental: changes in terms of risk and severity have been observed for females before and after menopause underlining the importance of altered circulating sexual hormones. Moreover, aging is a driving force for changes that interact with sex, shifting the inflammatory response in a sex-dependent manner. This review summarizes the present literature on sex differences in stroke-induced inflammatory responses, with the focus on different microglial responses along lifespan.
See related articles, p 1432, 1438, 1449, 1460, 1473, 1487
Stroke is the second cause of death worldwide and one of the leading causes of disability, although the incidence and mortality show large geographic variations, and depend on a variety of risk factors present in different populations. Sex differences have been described in terms of risk, symptoms, severity, and outcome.1–3 Despite women present a lower stroke incidence than men throughout most of their lifespan, the overall prevalence of stroke is higher in women because of their increased longevity.4 Moreover, the incidence of stroke increases in women after menopause, when the cytoprotective effect of estrogen disappears.5 At the time, also the severity of stroke increases to the level comparable to men at similar ages.6
An important contributor to stroke pathology is postischemic inflammation, which is largely geared by concerted function of brain microglia and peripheral immune cells.7 It is now clear that sex differences in the inflammatory response exist also in the pathogenesis of several other diseases.8–10 This review summarizes the sex differences in microglia and their role in the inflammatory response after stroke, taking into consideration the effect of circulating hormones and age.

Sex Differences in Microglia in Healthy Brain

Microglia exists in region, time, and context-dependent states in the healthy adult central nervous system, presenting differential phenotypic characteristics and specific transcriptomic profiles in different brain areas.11,12 However, knowledge on sex differences in microglia states in healthy brain is scarce.
Most of the information on sex differences in microglia are derived from studies performed in rodents. Sex-related differences in microglial density and morphology exist already in early the development and, although these features change during the lifespan, some of the differences remain in the adult brain. Only few studies have addressed sexual dimorphism in adult rodents in different brain regions. Some studies show that females have a higher density of microglia in specific hippocampal regions compared with males,13,14 whereas others show a higher density of microglia in the cortex, amygdala, and hippocampus of male rodents.15–17 Not only the density but also microglial morphology (eg, number of branches, processes per branch, and branch thickness) is region-dependent. For instance, microglia in the dorsal hippocampi have a higher total number of processes and more processes per branch compared with the prefrontal cortex in male rats. On the contrary, microglia are reported to have a higher total number of processes and more processes per branch in the prefrontal cortex compared with the dorsal hippocampi in female mice.18 Microglia are more ramified in the prefrontal cortex in female rats compared with males,19 while microglia in hippocampus are more ramified in males compared with females.20 The contradictory results may be partly due to differences in the analyzed brain region, age, and species, but also due to the microglia markers and techniques that were used.21 This fact limits the comparison between studies, urging the researchers to use harmonized techniques to explore the sex-differences in microglia (Table 1).
Table 1. Sex Differences in Microglial Density and Morphology in Adult Brain
Species: ageDifferencesReference.
Differences in cell density
C57BL/6 mice: 3–4, 13–14, and 20–24 moDG and CA1: F>M at all the ages13
C57BL/6 mice: 3–4 moCx, Hp, Amy: M>F17
Str, Cb: M=F
SD rats: 2–3 momPFC, prelimbic region: M=F19
SD rats: 2–3 momPFC: F>M15
SD rats: 2–3 moPaCx, CA1, CA3, DG, Amy: M>F14
Differences in microglia morphology (number and complexity of microglial branches) 
C57BL/6 mice: 3 moCA3: M>F in process volume and area, number of branches and intersections.20
C57BL/6 mice: 3–4 moCx, Hp, Amy: M>F in the size of the cell soma17
C57BL/6 mice: 3 moM: more complexity in the dHIP than in the PFC18
F: more complexity in the PFC than in the dHIP
SD rats: 2–3 moPaCx, CA1, CA3, DG, Amy: M>F in the thickness and length of microglia processes14
SD rats: 2–3 moCx: F>M in primed/ramified microglia19
Amy indicates amygdala; CA1, cornus ammonis 1; CA3, cornus ammonis 3; Cb, cerebellum; Cx, cerebral cortex; DG, dentate gyrus; dHIP, dorsal hippocampus; F, female; Hp, hippocampus; M, male; mPFC, medial prefrontal cortex; PaCx, parietal cortex; PFC, prefontral cortex; SD, Sprague-Dawley; and Str, striatum.
Of note, analysis of the expression of molecular patterns or transcriptomic programs in rodents reveals sex differences that cannot be quantified by morphometric analysis. Single-cell transcriptomics emerges as a powerful tool to analyze changes in cellular populations and states, yet, studies evaluating sex-differences at the single cell level are largely missing. In the recent years, a growing number of studies have demonstrated differences in the bulk transcriptomic and proteomic profiles of microglia depending on the brain region12,22 or age.23 Focusing on sex differences, transcriptomic analysis revealed 1109 differentially expressed genes in the hippocampus, 55 in the cerebral cortex and 46 in both regions between male and female mice, demonstrating brain region to be an important factor in microglial sex-diversity.17 Whole-brain microglia showed increased expression of 204 genes related to inflammatory processes in male mice compared with females, including regulation of cell migration, chemotaxis, and cytokine production.24 In agreement with transcriptomic studies, NF-κB (nuclear factor kappa B) is transcriptionally more activated in male microglia compared with female microglia.25 This suggests that male microglia are inherently more prone for inflammatory activation in pathological conditions compared with female microglia, consistently with higher expression of interferon-stimulated genes in males.26 On the other hand, females show higher expression of genes associated with development or morphogenesis that are under the control of transcription factors linked to the inhibition of the inflammatory response.24 Female microglia also express more sensome-related genes,26 a cluster of transcripts that code for proteins involved in sensing of endogenous ligands and microbes.25 In addition, proteomic profiling of microglia has demonstrated 263 proteins with significantly higher expression in males and other 96 proteins, preferably expressed in females.17 All these studies provide evidence that female and male microglia express different protein and RNA patterns even in the healthy brain which may contribute to the sex-differential response of microglia to stroke in rodent models (Table 2).
Table 2. Sex Differences in Mouse Microglial Gene Expression at Different Ages
Species and ageRegionDifferencesReference
C57BL/6 mice: 3-to 4-mo oldCx, HpGO of genes overexpressed in M cortex: transcription factor activity, histone demethylation and deacetylation.
GO of genes overexpressed in M hippocampus: regulation of defense response to bacteria, insulin receptor pathway, glial cell differentiation, and ATP binding
GO of genes overexpressed in F dataset: GABA and glutamate receptor activity, ubiquitin protein activity, and magnesium ion transport
17
C57BL/6 mice: 3-mo oldWhole brainGO of genes overexpressed in M: inflammatory response, leucocyte migration, regulation of response to wounding, chemotaxis, regulation of cytokine production. Many genes regulated by NF-kB
GO of genes overexpressed in F: movement of cell, nervous system development, cell migration, vasculature development, anatomic structure formation, actin cytoskeleton organization. Many genes regulated by transcription factors, linked to the inhibition of the inflammatory response.
24
C57BL/6 mice: 3-, 12- and 24-mo oldHpActivation of inflammatory genes in both sexes, but more evident in old F: higher expression of genes for the complement system and the microglial sensome with the age.27
C57BL/6J mice: 2- to 3-mo oldN/IF: higher expression of interferon-stimulated genes (S100a8, S100a9, Ifit1, Ifit2, Cxcl10, Ccl2, Irf1, Ccnd3, and Gbp5).
Differential expression of sensome genes between F and M.
26
C57BL/6 background mice: 3-, 12-, and 24-mo oldForebrainThe main sex differences in gene expression are observed at 24-mo old. 37 transcripts differentially expressed between M and F at 24-mo.
F: higher levels of Spp1, Gpnmb, Lgals3, ApoE, Ccl3, Clec7a, and Ccl4.
28
ApoE: apolipoprotein E; Ccl2, C-C motif chemokine ligand 2; Ccl3, C-C motif chemokine ligand 3; Ccl4, C-C motif chemokine ligand 4; Ccnd3, cyclin D3; Clec7a, C-type lectin domain family 7 member A; Cx, cerebral cortex; Cxcl10, C-X-C motif chemokine ligand 10; GABA, gamma aminobutyric acid; GO, gene ontology; F, female; Gbp5, guanylate-binding protein 5; Gpnmb, glycoprotein Nmb; Hp, hippocampus; Ifit, interferon-induced proteins with tetratricopeptide repeats; Lgals3, lectin galactoside-binding soluble 3; M, male; N/I, not indicated; NF-kB, nuclear factor kappa B; S100a8, S100 calcium-binding protein A8; S100a9, S100 calcium-binding protein A9; and Spp1, sporulation-specific protein 1.
In addition, there are differences in microglia across species. Microglial morphology is relatively conserved between humans, macaque, and rodents, but human microglia are more heterogeneous and display an organization in different subtypes, as revealed by single-cell RNA-seq.12,29 These subtypes do not correlate with sex-specific features but add an additional layer of complexity to the analysis of sex differences.

Sex Differences in Microglia Response to Stroke

Microglia are essential in responding to brain insults and play a central role in initiating and gearing the neuroinflammatory response.7 In the presence of a stimulus, such as stroke, these cells can exhibit a wide spectrum of activation states.30 Traditionally, microglia were divided into M1 and M2 phenotypes, with M1 type cells secreting pro-inflammatory cytokines, proteins of the complement system, reactive oxygen species and reactive nitrogen species, and the highly phagocytic M2 type microglia releasing anti-inflammatory and immunosuppressing cytokines. While this classification has been used extensively in the literature, we omit from using this over-simplistic dichotomy, and rather refer to microglial states that are both stimulus and context dependent and form a continuity between different states.30,31
The abovementioned sex differences in the gene transcription contribute to their differential responses to ischemic stroke. Differential stroke outcomes (short- and long-term) between sexes, as well as sex-specific inflammatory response are reported, which are critical to understand the consequences of sex-differences in microglial response to stroke and in the related outcomes (Table 3).
Table 3. Sex Differences in Infarct Size and in the Inflammatory Response
Infarct volume
M>FNeonatal: 3 dpi,32 30 dpi,33 90 dpi34
Young/adult: 1 hpi,24 1 dpi,24 2 dpi,24,35 3 dpi,36–38 4 dpi42
M=FNeonatal: 1 dpi,32 3 dpi34,39
F>MAged: 3 dpi40
Microglial density after stroke
M>F41
M=F34,42,43
Microglial reactivity/activation after stroke
M>FRamification44 and morphology39
Leukocyte infiltration
F>M45
M>F33,35,39,46
Pro-inflammatory serum cytokines
M>FTNF-α,32,33 IL-1β33
Dpi indicates days postischemia; F, female; hpi, hours postischemia; IL-1β, interleukin 1 beta; and M, male; and TNF-α, tumor necrosis factor alpha.

Sex Differences in Infarct Volume and Microglial Activation After Stroke

According to clinical data, the age-adjusted incidence of stroke is higher in men compared women, although this has changed during the recent years.47 In line, adult female rodents display smaller infarcts compared with males24,35–38,42 (Table 3). The protection in females also exists in neonatal ischemia, but the studies are less consistent32,34,39 The protective effect observed in young/adult females in their reproductive period of life is reversed with aging,40,48 as discussed in Section Interations Between Age and Sex: the Relevance of the X Chromosome.
To correlate microglia activation to the severity of stroke, different morphological parameters have been utilized, such as number of microglia and morphological changes from ramified to amoeboid morphology. Despite the lack of sex differences in the number of microglia after stroke in most of the studies, males display a more ameboid microglial morphology compared with females that has been interpreted as an indication of more robust activation.24,44 These studies suggest that microglial activation in young females is less pronounced compared with males. However, besides the morphological features, microglia can respond in many other ways depending on the cytokine profile they secrete, and this has led to a myriad of phenotypes with different anti- and pro-inflammatory properties.31

Stroke Induces Divergent Microglial Phenotypes Between Male and Female

Several studies have had a focus on the characterization of the microglial phenotypic response after stroke. After ischemic insult, microglial inflammatory phenotype polarizes differently between sexes. The expression of markers of pro-inflammatory microglia is higher in males compared with females, where the anti-inflammatory phenotype is dominant,35,37 revealing a clear predisposition of male microglia to trigger inflammation. The studies gathered in Table 4 are evidence of distinctive molecular pathways of triggering inflammation between sexes and are in line with transcriptomics studies that showed differential expression of inflammation-related genes in males and females in healthy brain (Table 4).
Table 4. Sex Differences in Microglia Markers After Hypoxia/Ischemia and Stroke
 Marker TechniqueModelReference
Pro-inflammatory phenotypeCD16/32+M>FmRNANeonatal pMCAO39
M=FProteinCortical injury in adults41
TNF-αM>FmRNANeonatal pMCAO39
COX-2M>FmRNANeonatal pMCAO39
M=FproteinNeonatal pMCAO34
IL-1βM=FmRNANeonatal pMCAO39
NOS2M=FmRNANeonatal pMCAO39
MHCIIM>FProteinNeonatal hypoxia-ischemic32
M=FproteinNeonatal hypoxia-ischemic33
Anti-inflammatory phenotypeYM1F>MProteintMCAO in adults35
F>MProteinpMCAO in adults24
Arg1F>MProteintMCAO in adults35
F>MProteintMCAO in adults34
F=MmRNANeonatal pMCAO39
F=MproteinNeonatal pMCAO34
M>FproteinCortical injury in adults41
CD206F>MproteintMCAO in adults37
F>MproteinNeonatal hypoxia-ischemia33
F=MmRNANeonatal pMCAO39
M-trans phenotypeArg1+/COX2F>MProteinNeonatal pMCAO34
OthersCX3CR1M=FmRNANeonatal pMCAO39
CD68M=FProteinNeonatal hypoxia-ischemia43
The changes are divided depending on if they are measured based on protein levels (indicating changes in the expression per cell or changes in the number of positive cells for this protein) or mRNA levels (showing mRNA transcriptional changes in microglia). Arg1 indicates arginase 1; CD206, cluster of differentiation 206; CD68, cluster of differentiation 68; COX2, cyclooxigenase 2; CX3CR1, C-X3-C motif chemokine receptor 1; F, female; IL-1β, interleukin 1β; M, male; MHCII, major histocompatibility II; pMCAO, permanent middle cerebral artery occlusion; tMCAO, transient middle cerebral artery occlusion; and TNF-α, tumor necrosis factor alpha.
The evidence of divergent microglia responses to stroke between sexes has also been demonstrated in prenatal ischemia, when the sexual hormones remain at the same levels in both sexes.39 This suggests that part of differences between males and females arise from inherent developmental differences before the sexual maturation. In this regard, male and female microglia display differences in their gene expression during brain development,20,26 some of them are associated with the X and Y chromosomes. The number of differently expressed genes increases in adulthood.26 For instance, adult females show a higher expression of IL (interleukin)-4 receptors, a well-known inducer of the anti-inflammatory phenotype, as well as higher levels of Ifr3 (interferon regulator factor 3) compared with males, which acts as an anti-inflammatory regulator of microglia.17,46 Similarly, the higher responsiveness to immunologic stimuli observed in males may be also related to the higher levels of proteins of the TLR (toll-like receptor) pathway and to the presentation of MHC (major histocompatibility complex) I and II reported in males.17,49
These sex-related differences could arise from the specific programs activated to drive the differential sexual maturation of the brain.50 Early in the development, between post-natal day 1 and post-natal day 10, the expression of pro-inflammatory-related genes (specifically PGE2 [prostaglandin E2]) triggered by the presence of estradiol aromatized from testicular testosterone drives the brain masculinization.51 Microglia participate in this process by both producing and responding to PGE2,50 and this might lead to a priming effect persisting long in the adulthood,52 as well as to higher male vulnerability observed in early-life inflammation.
Despite the scarce number of studies that explore the sex-differential response to stroke in microglia, all the previous evidence suggest that microglia acquire a sex-linked transcriptomic signature triggered by X- and Y-associated gene expression and brain sexual differentiation. This may render microglia imprinted even after the sensitive period,24,26 leading to differential inflammatory responses between sexes reported in brain pathologies. The sex differences in microglial response are also observed in human postmortem tissues from different diseases, including stroke,53 Alzheimer disease,54 or glioma,55 evidencing that the differential sex response is a conserved feature between species that also occurs in the human brain.

Influence of Circulating Sex Hormones in Stroke Outcome

Sex hormones have a profound impact on the outcome of stroke.56 There is clinical evidence demonstrating a higher stroke incidence and severity in women after the menopause,5 and for this reason, female sex hormones, specifically estrogen, have been attributed a cytoprotective role in ischemic stroke.57,58 Several studies consistently demonstrate that estrogens drive the sex-differential protective effect in young females, which has been confirmed in ovariectomized female rodents subjected to stroke and the protection by estrogen therapies.57,59,60 While the effect of estrogens is consistent across studies, the role of testosterone remains controversial with evidence of both detrimental and beneficial effects.61,62 The fact that the beneficial effect of testosterone is dose-dependent62 suggests that it is the balance between the sex hormones that matters.63 Besides this controversy, it has been suggested that estrogens have an anti-inflammatory role after stroke in which microglia may be involved.64

Estrogens and Microglia After Stroke

Estrogens initiate most physiological and pharmacological actions by binding to the ERα and ERβ (estrogen receptors α and β), and to the recently identified novel estrogen receptor denominated GPR30. The predominant and biologically most active form of estrogen is the 17β-estradiol (E2).65 At present, little is known about ER receptors in microglia, and there is an ongoing debate whether ERα or ERβ mediate signaling in microglia.58 Whether ERβ but not ERα expressed in microglia is controversial.66,67 However, transcriptomic studies show that there are no sex differences in ER subtype expression in microglia,68 suggesting that the cytoprotective effect of estrogens may rely on the amount of hormone rather than on the differential expression of estrogen receptors between females and males. In addition, a study carried out by Elzer et al59 proposes that the estrogen-elicited cytoprotection relies on neuronal ERα expression, suggesting that the protective mechanism of estrogens is mediated by neurons rather than microglia. Immunosuppressive effect of E2 on microglia and astrocytes is apparent: it limits microglial-inducible nitric oxide synthase activity, downregulates apoptotic factors and secretion of pro-inflammatory cytokines, such as IL-1β (interleukin 1 beta), IL-6 (interleukin 6), and TNF-α (tumor necrosis factor alpha). In addition, E2 mediates upregulation of pro-survival and anti-inflammatory factors, including TREM2 (triggering receptors expressed on myeloid cells 2) and IL-10 (interleukin-10; Figure). Indeed, several publications report an inflammatory resolving effect of estrogen.57,69
Figure. Factors that contribute to the sex differences in microglial response to stroke. Yellow refers to females and blue to males. Among the factors that contribute to sex differences in microglia response are the peak of hormones. In males, this drives the brain sexualization and imprints microglia toward a pro-inflammatory predisposition (blue line). The presence of estrogen in females during adulthood results in cytoprotection after experimental stroke (orange line). The age-dependent increase of X-chromosome escapes in females, giving an extra dosage of X-related genes (gray line). Infarct volumes after stroke are smaller in adult females than in males. Microglia also adopts more anti-inflammatory phenotype in adult females compared with males. Aged females, however, shows a switch in the response and the infarct volume increases compared with young females, and microglia displays a more pro-inflammatory phenotype. E2 indicates 17β-estradiol; IL-1β, interleukin 1β; IL-6, interleukin 6; IL-10, interleukin-10; MHCII, major histocompatibility II; TNF-α, tumor necrosis factor alpha; and TREM2, triggering receptor expressed on myeloid cells 2. Created with www.BioRender.com
It is worth to highlight that, as previously mentioned, microglia display a differential response between males and females after postnatal stroke in the absence of sex hormones.39 Moreover, microglia maintains sex-specific expression of a significant number of genes in culture, in ovariectomized female mice, or after transplantation to the opposite sex.24 All this evidence supports the hypothesis of the microglia imprinting in early development, but it does not contradict the fact that estrogens play an important role in the post-ischemic inflammatory response by microglia.

Interations Between Age and Sex: the Relevance of the X Chromosome

Age is the main contributor for the outcome of stroke and has a complex and interactive effect with sex. Aging by itself is associated to changes in the immune system, such as a rise in inflammatory markers and diminished response to cytokine stimulation, which can modulate the stroke progression and outcome.70 In experimental stroke, while young females show smaller infarct volume compared with males, the outcome is inverted in aged males.16,40,48 It is still under discussion whether the worsened outcome of aged females compared with males is caused by the lack or dysregulation of estrogens, or due to a sex chromosome–linked features. Some studies highlighting the relevance of the X chromosome in aging suggest that these differences could rely on X chromosome dosage. The additional X chromosome is silenced in young female animals,71 thus no differences in the infarct volumes are observed between females with 1 and 2 X chromosomes (XX and XO).72 However, aging affects the stability of the silencing, and thus the X chromosome may escape from the inactivation, giving an extra dosage of X-related genes.73 The Four Core Genotype mouse model is a useful tool to dissect the effect of X-dosage from sex hormones along lifespan. In this model, the testis-determining gene sry is deleted from the Y chromosome and inserted on an autosome, allowing the generation of XXM mice (a gonadal male with XX chromosomal complement) and XYF mice (a gonadal female with XY chromosome complement).74 Manwani et al60 utilized this model to demonstrate that gonadal hormones are responsible for the cytoprotection in young females compared with males. The authors show that both XXF and XYF (gonadal females) display reduced infarct volumes compared with XXM and XYM (gonadal males), which is not observed when comparing males with ovariectomized females. Moreover, estradiol treatment not only reverses this but also exerts protective effect in males suggesting that circulating estrogen is the main driver for the protection during reproductive period of life. However, age is a game-changer. McCullough et al40 used the Four Core Genotype model in aged mice to demonstrate that aging changes the relative contribution of XX/XY chromosomes to ischemic damage when compared with young mice. XXF and XXM aged mice have significantly larger infarct volumes. Moreover, these mice show a more robust stroke-induced microglial activation pattern compared with XY female and XY male mice, revealing that the chromosomal complement shapes the inflammatory response as well as the outcome of stroke in aged mice.40 The combination of these 2 studies provide a wide perspective of the relative contribution of XX/XY chromosomes along lifespan.
Focusing on microglia specific features in aged animals, sex differences are scarcely studied. Transcriptomic analysis through the lifespan show activation of inflammatory genes in both males and females, but these changes are more evident in females, resulting in a sharper sex divergence in the elderly.27,28 These data suggest that the predisposition of pro-inflammatory signaling observed in young males is compensated by age in females, possibly via X chromosome escape as described above.53 In fact, microglia from aged female animals express higher levels of pro-inflammatory markers (MHCII [major histocompatibility II]) compared with male microglia, together with an increase of pro-inflammatory cytokines in the serum (TNF-α and IL-1β).40 Another example of this sexual divergence along the age is the differential expression of complement proteins in microglia of old females and males. The complement system is induced upon ischemic stroke and modulates of the poststroke inflammatory response.75 Microglia are described to release several complement effectors after stroke, such as C1qa (complement 1qa)76 and complement 3 (C3).77 Aging induces the transcription of C1qa and complement 1qc (C1qc) genes and the translation of C1q protein at higher levels in females compared with males.27 Similar to the complement system, there is a higher upregulation of the sensome genes in female microglia compared with males.27 In addition, the importance of X chromosome escape has been highlighted by a recent study on 2 X escapee genes, the lysine demethylase 5C and 6a (kdm5c and kdm6a respectively), which code for histone demethylases.53 These genes regulate microglial production of cytokines and are expressed at higher levels in microglia from aged females subjected to middle cerebral artery occlusion compared with males as well as in human postmortem tissue from stroke patients, suggesting that epigenetic modifications may play a role in poststroke sex differences.53
All these studies highlight the fact that sex differences in the inflammatory response are not static along lifespan and raise the need of more in-deep studies of the sex-differential inflammatory response after stroke in aged animals to achieve better therapies.

Sex-Specific Features of Microglia As a Therapeutic Target in Stroke

Sex differences in treatment efficacy are common in stroke, yet, only a few studies have reported sex-differences on microglia-based therapies. Minocycline and other PARP (poly [ADP-ribose] polymerase) inhibitors78 have been reported to alleviate poststroke neuroinflammation and ischemic damage only in male rats.79,80 Interestingly, minocycline was ineffective in ovariectomized female with estrogen levels similar to males. Consistent with experimental studies, Amiri-Nikpour et al81 showed that oral administration of minocycline was effective in male patients with stroke but not in women. Meta-analysis of 7 randomized-controlled clinical trials showed that although minocycline seems to be a promising cytoprotective agent in patients with stroke, further studies are needed to evaluate the efficacy and safety.82 Our cautious conclusion is that minocycline may fail to effectively modulate highly dynamic, complex and sex-specific stroke-induced microglial responses.
Indeed, to discover more effective microglia-targeted therapies for stroke, we need to gain a deeper understanding of microglia biology. An elegant work by Villa et al demonstrated a significant difference in the transcriptome profile between adult male and female microglia independent of hormonal cues. Female phenotype is retained after transfer of microglia into male brain and more importantly, female microglia protected the male brain from ischemic stroke.24 The higher expression of genes involved in cell plasticity, control of inflammation and brain repair in female mice could contribute to protection. These genes may provide novel targets for sex-related approaches for effective stroke therapy.
Going beyond the acute phases of stroke is also important. Peri-infarct cortex is highly plastic undergoing reorganization through the reactivation of neurodevelopmental mechanisms to compensate lost neuronal functions.83 Increase in dendritic spine production together with axonal sprouting is an integral part of remodeling and brain repair process. Here, stroke-induced microglia have a major role in elimination of spines and shaping neural networks in functionally relevant manner. In depth understanding of the sex differences in microglia modulation of the peri-infarct cortex during stroke repair process, similar to neurodevelopment, may offer novel therapeutic targets for long-term functional brain recovery.
Different molecular mechanisms evolve after stroke in time-dependent manner. These are potential targets for microglia-based therapies to reduce degeneration and alleviate neuroinflammation or later to enhance brain repair, when properly timed. High number experimental drugs and interventions are effective in stroke models, yet, they have not translated into clinic success.84 It is expected that emerging understanding of sexually dimorphic brain and stroke pathology improves translation of basic science results to sex-specific therapies. For translational success, both sexes and aged animals should be considered and used whenever possible as suggested by STAIR committee,85 despite budgetary constraints and complex study design. Microglial chimeric mice enable comparison of sex differences in human context.86

Conclusions and Future Directions

Sex has been shown to contribute to the progression of poststroke inflammation and outcome. Specific molecular programs are launched to drive the process of brain differentiation between sexes already in very early stages of development and specifically in microglia. The characteristic differences acquired during development will influence the inflammatory response in adulthood and particularly in case of severe brain insults such as stroke. These inherent differences need to be investigated and understood to ensure that the novel therapeutic paradigms translate into new treatments.
Recent advances in molecular biology and single-cell analysis have provided considerable insights into dynamic microglial responses in stroke. In-depth understanding of sex-dependent microglial responses in aging animals and both acute and long-term consequences aid in driving and accelerating development of sex- and microglia-specific, personalized treatment strategies for acute protection and brain repair in stroke.

Footnote

Nonstandard Abbreviations and Acronyms

C1qa
complement 1qa
ERα
estrogen-receptor α
ERβ
estrogen-receptor β
Ifr3
interferon regulator factor 3
IL-1β
interleukin 1 beta
IL-6
interleukin 6
IL-10
interleukin-10
MHCII
major histocompatibility II
PARP
poly-adp ribose polymerase
PGE2
prostaglandin E2
TLR
toll-like receptor
TNF-α
tumor necrosis factor alpha
TREM2
triggering receptor expressed on myeloid cells 2

References

1.
Lisabeth LD, Reeves MJ, Baek J, Skolarus LE, Brown DL, Zahuranec DB, Smith MA, Morgenstern LB. Factors influencing sex differences in poststroke functional outcome. Stroke. 2015;46:860–863. doi: 10.1161/STROKEAHA.114.007985
2.
Zrelak PA. Sex-based differences in symptom perception and care-seeking behavior in acute stroke. Perm J. 2018;22:18–042. doi: 10.7812/TPP/18-042
3.
Phan HT, Blizzard CL, Reeves MJ, Thrift AG, Cadilhac D, Sturm J, Heeley E, Otahal P, Konstantinos V, Anderson C, et al. Sex differences in long-term mortality after stroke in the INSTRUCT (INternational STRoke oUtComes sTudy): a meta-analysis of individual participant data. Circ Cardiovasc Qual Outcomes. 2017;10:e003436. doi: 10.1161/CIRCOUTCOMES.116.003436
4.
Krishnamurthi RV, Ikeda T, Feigin VL. Global, regional and country-specific burden of ischaemic stroke, intracerebral haemorrhage and subarachnoid haemorrhage: a systematic analysis of the global burden of disease study 2017. Neuroepidemiology. 2020;54:171–179. doi: 10.1159/000506396
5.
Lisabeth L, Bushnell C. Stroke risk in women: the role of menopause and hormone therapy. Lancet Neurol. 2012;11:82–91. doi: 10.1016/S1474-4422(11)70269-1
6.
Dehlendorff C, Andersen KK, Olsen TS. Sex disparities in stroke: women have more severe strokes but better survival than men. J Am Heart Assoc. 2015;4:e001967. doi: 10.1161/JAHA.115.001967
7.
Anrather J, Iadecola C. Inflammation and stroke: an overview. Neurotherapeutics. 2016;13:661–670. doi: 10.1007/s13311-016-0483-x
8.
Batton KA, Austin CO, Bruno KA, Burger CD, Shapiro BP, Fairweather D. Sex differences in pulmonary arterial hypertension: role of infection and autoimmunity in the pathogenesis of disease. Biol Sex Differ. 2018;9:15. doi: 10.1186/s13293-018-0176-8
9.
Ortona E, Pierdominici M, Maselli A, Veroni C, Aloisi F, Shoenfeld Y. Sex-based differences in autoimmune diseases. Ann Ist Super Sanita. 2016;52:205–212. doi: 10.4415/ANN_16_02_12
10.
Tejera D, Heneka MT. Microglia in neurodegenerative disorders. Methods Mol Biol. 2019;2034:57–67. doi: 10.1007/978-1-4939-9658-2_5
11.
Grabert K, Michoel T, Karavolos MH, Clohisey S, Baillie JK, Stevens MP, Freeman TC, Summers KM, McColl BW. Microglial brain region-dependent diversity and selective regional sensitivities to aging. Nat Neurosci. 2016;19:504–516. doi: 10.1038/nn.4222
12.
Masuda T, Sankowski R, Staszewski O, Böttcher C, Amann LSagar, Scheiwe C, Nessler S, Kunz P, van Loo G, et al. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature. 2019;566:388–392. doi: 10.1038/s41586-019-0924-x
13.
Mouton PR, Long JM, Lei DL, Howard V, Jucker M, Calhoun ME, Ingram DK. Age and gender effects on microglia and astrocyte numbers in brains of mice. Brain Res. 2002;956:30–35. doi: 10.1016/s0006-8993(02)03475-3
14.
Schwarz JM, Sholar PW, Bilbo SD. Sex differences in microglial colonization of the developing rat brain. J Neurochem. 2012;120:948–963. doi: 10.1111/j.1471-4159.2011.07630.x
15.
Bollinger JL, Salinas I, Fender E, Sengelaub DR, Wellman CL. Gonadal hormones differentially regulate sex-specific stress effects on glia in the medial prefrontal cortex. J Neuroendocrinol. 2019;31:e12762. doi: 10.1111/jne.12762
16.
Manwani B, Liu F, Scranton V, Hammond MD, Sansing LH, McCullough LD. Differential effects of aging and sex on stroke induced inflammation across the lifespan. Exp Neurol. 2013;249:120–131. doi: 10.1016/j.expneurol.2013.08.011
17.
Guneykaya D, Ivanov A, Hernandez DP, Haage V, Wojtas B, Meyer N, Maricos M, Jordan P, Buonfiglioli A, Gielniewski B, et al. Transcriptional and translational differences of microglia from male and female brains. Cell Rep. 2018;24:2773–2783.e6. doi: 10.1016/j.celrep.2018.08.001
18.
Simões-Henriques C, Mateus-Pinheiro M, Gaspar R, Pinheiro H, Mendes Duarte J, Baptista FI, Canas PM, Fontes-Ribeiro CA, Cunha RA, Ambrósio AF, et al. Microglia cytoarchitecture in the brain of adenosine A2A receptor knockout mice: Brain region and sex specificities. Eur J Neurosci. 2020;51:1377–1387. doi: 10.1111/ejn.14561
19.
Bollinger JL, Bergeon Burns CM, Wellman CL. Differential effects of stress on microglial cell activation in male and female medial prefrontal cortex. Brain Behav Immun. 2016;52:88–97. doi: 10.1016/j.bbi.2015.10.003
20.
Hanamsagar R, Alter MD, Block CS, Sullivan H, Bolton JL, Bilbo SD. Generation of a microglial developmental index in mice and in humans reveals a sex difference in maturation and immune reactivity. Glia. 2017;65:1504–1520. doi: 10.1002/glia.23176
21.
Eme-Scolan E, Dando SJ. Tools and approaches for studying microglia in vivo. Front Immunol. 2020;11:583647. doi: 10.3389/fimmu.2020.583647
22.
Ayata P, Schaefer A. Innate sensing of mechanical properties of brain tissue by microglia. Curr Opin Immunol. 2020;62:123–130. doi: 10.1016/j.coi.2020.01.003
23.
Pan J, Ma N, Yu B, Zhang W, Wan J. Transcriptomic profiling of microglia and astrocytes throughout aging. J Neuroinflammation. 2020;17:97. doi: 10.1186/s12974-020-01774-9
24.
Villa A, Gelosa P, Castiglioni L, Cimino M, Rizzi N, Pepe G, Lolli F, Marcello E, Sironi L, Vegeto E, et al. Sex-specific features of microglia from adult mice. Cell Rep. 2018;23:3501–3511. doi: 10.1016/j.celrep.2018.05.048
25.
Hickman SE, Kingery ND, Ohsumi TK, Borowsky ML, Wang LC, Means TK, El Khoury J. The microglial sensome revealed by direct RNA sequencing. Nat Neurosci. 2013;16:1896–1905. doi: 10.1038/nn.3554
26.
Thion MS, Low D, Silvin A, Chen J, Grisel P, Schulte-Schrepping J, Blecher R, Ulas T, Squarzoni P, Hoeffel G, et al. Microbiome influences prenatal and adult microglia in a sex-specific manner. Cell. 2018;172:500–516.e16. doi: 10.1016/j.cell.2017.11.042
27.
Mangold CA, Wronowski B, Du M, Masser DR, Hadad N, Bixler GV, Brucklacher RM, Ford MM, Sonntag WE, Freeman WM. Sexually divergent induction of microglial-associated neuroinflammation with hippocampal aging. J Neuroinflammation. 2017;14:141. doi: 10.1186/s12974-017-0920-8
28.
Kang SS, Ebbert MTW, Baker KE, Cook C, Wang X, Sens JP, Kocher JP, Petrucelli L, Fryer JD. Microglial translational profiling reveals a convergent APOE pathway from aging, amyloid, and tau. J Exp Med. 2018;215:2235–2245. doi: 10.1084/jem.20180653
29.
Geirsdottir L, David E, Keren-Shaul H, Weiner A, Bohlen SC, Neuber J, Balic A, Giladi A, Sheban F, Dutertre CA, et al. Cross-species single-cell analysis reveals divergence of the primate microglia program. Cell. 2019;179:1609–1622.e16. doi: 10.1016/j.cell.2019.11.010
30.
Stratoulias V, Venero JL, Tremblay MÈ, Joseph B. Microglial subtypes: diversity within the microglial community. EMBO J. 2019;38:e101997. doi: 10.15252/embj.2019101997
31.
Hammond TR, Dufort C, Dissing-Olesen L, Giera S, Young A, Wysoker A, Walker AJ, Gergits F, Segel M, Nemesh J, et al. Single-cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes. Immunity. 2019;50:253–271.e6. doi: 10.1016/J.IMMUNI.2018.11.004
32.
Mirza MA, Ritzel R, Xu Y, McCullough LD, Liu F. Sexually dimorphic outcomes and inflammatory responses in hypoxic-ischemic encephalopathy. J Neuroinflammation. 2015;12:32. doi: 10.1186/s12974-015-0251-6
33.
Al Mamun A, Yu H, Romana S, Liu F. Inflammatory responses are sex specific in chronic hypoxic-ischemic encephalopathy. Cell Transplant. 2018;27:1328–1339. doi: 10.1177/0963689718766362
34.
Charriaut-Marlangue C, Leconte C, Csaba Z, Chafa L, Pansiot J, Talatizi M, Simon K, Moretti R, Marchand-Leroux C, Baud O, et al. Sex differences in the effects of PARP inhibition on microglial phenotypes following neonatal stroke. Brain Behav Immun. 2018;73:375–389. doi: 10.1016/j.bbi.2018.05.022
35.
Xiong X, Xu L, Wei L, White RE, Ouyang YB, Giffard RG. IL-4 Is required for sex differences in vulnerability to focal ischemia in mice. Stroke. 2015;46:2271–2276. doi: 10.1161/STROKEAHA.115.008897
36.
Dotson AL, Wang J, Saugstad J, Murphy SJ, Offner H. Splenectomy reduces infarct volume and neuroinflammation in male but not female mice in experimental stroke. J Neuroimmunol. 2015;278:289–298. doi: 10.1016/j.jneuroim.2014.11.020
37.
Seifert HA, Benedek G, Liang J, Nguyen H, Kent G, Vandenbark AA, Saugstad JA, Offner H. Sex differences in regulatory cells in experimental stroke. Cell Immunol. 2017;318:49–54. doi: 10.1016/j.cellimm.2017.06.003
38.
Seifert HA, Zhu W, Vandenbark AA, Alkayed NJ, Offner H. Sex differences in the therapeutic effects of anti-PDL2 neutralizing antibody on stroke. Metab Brain Dis. 2019;34:1705–1712. doi: 10.1007/s11011-019-00476-3
39.
Villapol S, Faivre V, Joshi P, Moretti R, Besson VC, Charriaut-Marlangue C. Early sex differences in the immune-inflammatory responses to neonatal ischemic stroke. Int J Mol Sci. 2019;20:E3809. doi: 10.3390/ijms20153809
40.
McCullough LD, Mirza MA, Xu Y, Bentivegna K, Steffens EB, Ritzel R, Liu F. Stroke sensitivity in the aged: sex chromosome complement vs. gonadal hormones. Aging (Albany NY). 2016;8:1432–1441. doi: 10.18632/aging.100997
41.
Acaz-Fonseca E, Duran JC, Carrero P, Garcia-Segura LM, Arevalo MA. Sex differences in glia reactivity after cortical brain injury. Glia. 2015;63:1966–1981. doi: 10.1002/glia.22867
42.
Freitas-Andrade M, Bechberger JF, MacVicar BA, Viau V, Naus CC. Pannexin1 knockout and blockade reduces ischemic stroke injury in female, but not in male mice. Oncotarget. 2017;8:36973–36983. doi: 10.18632/oncotarget.16937
43.
Pimentel-Coelho PM, Michaud JP, Rivest S. Evidence for a gender-specific protective role of innate immune receptors in a model of perinatal brain injury. J Neurosci. 2013;33:11556–11572. doi: 10.1523/JNEUROSCI.0535-13.2013
44.
Morrison HW, Filosa JA. Sex differences in astrocyte and microglia responses immediately following middle cerebral artery occlusion in adult mice. Neuroscience. 2016;339:85–99. doi: 10.1016/j.neuroscience.2016.09.047
45.
Beckmann L, Obst S, Labusek N, Abberger H, Köster C, Klein-Hitpass L, Schumann S, Kleinschnitz C, Hermann DM, Felderhoff-Müser U, et al. Regulatory T cells contribute to sexual dimorphism in neonatal hypoxic-ischemic brain injury. Stroke. 2022;53:381–390. doi: 10.1161/STROKEAHA.121.037537
46.
Bodhankar S, Lapato A, Chen Y, Vandenbark AA, Saugstad JA, Offner H. Role for microglia in sex differences after ischemic stroke: importance of M2. Metab Brain Dis. 2015;30:1515–1529. doi: 10.1007/s11011-015-9714-9
47.
Bushnell CD, Chaturvedi S, Gage KR, Herson PS, Hurn PD, Jiménez MC, Kittner SJ, Madsen TE, McCullough LD, McDermott M, et al. Sex differences in stroke: challenges and opportunities. J Cereb Blood Flow Metab. 2018;38:2179–2191. doi: 10.1177/0271678X18793324
48.
Liu F, Yuan R, Benashski SE, McCullough LD. Changes in experimental stroke outcome across the life span. J Cereb Blood Flow Metab. 2009;29:792–802. doi: 10.1038/jcbfm.2009.5
49.
Gesuete R, Kohama SG, Stenzel-Poore MP. Toll-like receptors and ischemic brain injury. J Neuropathol Exp Neurol. 2014;73:378–386. doi: 10.1097/NEN.0000000000000068
50.
Lenz KM, Nugent BM, Haliyur R, McCarthy MM. Microglia are essential to masculinization of brain and behavior. J Neurosci. 2013;33:2761–2772. doi: 10.1523/JNEUROSCI.1268-12.2013
51.
Amateau SK, McCarthy MM. Induction of PGE2 by estradiol mediates developmental masculinization of sex behavior. Nat Neurosci. 2004;7:643–650. doi: 10.1038/nn1254
52.
Villa A, Della Torre S, Maggi A. Sexual differentiation of microglia. Front Neuroendocrinol. 2019;52:156–164. doi: 10.1016/j.yfrne.2018.11.003
53.
Qi S, Al Mamun A, Ngwa C, Romana S, Ritzel R, Arnold AP, McCullough LD, Liu F. X chromosome escapee genes are involved in ischemic sexual dimorphism through epigenetic modification of inflammatory signals. J Neuroinflammation. 2021;18:70. doi: 10.1186/s12974-021-02120-3
54.
Guillot-Sestier MV, Araiz AR, Mela V, Gaban AS, O’Neill E, Joshi L, Chouchani ET, Mills EL, Lynch MA. Microglial metabolism is a pivotal factor in sexual dimorphism in Alzheimer’s disease. Commun Biol. 2021;4:711. doi: 10.1038/s42003-021-02259-y
55.
Ochocka N, Segit P, Walentynowicz KA, Wojnicki K, Cyranowski S, Swatler J, Mieczkowski J, Kaminska B. Single-cell RNA sequencing reveals functional heterogeneity of glioma-associated brain macrophages. Nat Commun. 2021;12:1151. doi: 10.1038/s41467-021-21407-w
56.
Sohrabji F, Okoreeh A, Panta A. Sex hormones and stroke: beyond estrogens. Horm Behav. 2019;111:87–95. doi: 10.1016/j.yhbeh.2018.10.010
57.
Guo H, Yang J, Liu M, Wang L, Hou W, Zhang L, Ma Y. Selective activation of estrogen receptor β alleviates cerebral ischemia neuroinflammatory injury. Brain Res. 2020;1726:146536. doi: 10.1016/J.BRAINRES.2019.146536
58.
Rahimian R, Cordeau P, Kriz J. Brain response to injuries: when microglia go sexist. Neuroscience. 2019;405:14–23. doi: 10.1016/j.neuroscience.2018.02.048
59.
Elzer JG, Muhammad S, Wintermantel TM, Regnier-Vigouroux A, Ludwig J, Schütz G, Schwaninger M. Neuronal estrogen receptor-alpha mediates neuroprotection by 17beta-estradiol. J Cereb Blood Flow Metab. 2010;30:935–942. doi: 10.1038/jcbfm.2009.258
60.
Manwani B, Bentivegna K, Benashski SE, Venna VR, Xu Y, Arnold AP, McCullough LD. Sex differences in ischemic stroke sensitivity are influenced by gonadal hormones, not by sex chromosome complement. J Cereb Blood Flow Metab. 2015;35:221–229. doi: 10.1038/jcbfm.2014.186
61.
Cheng J, Alkayed NJ, Hurn PD. Deleterious effects of dihydrotestosterone on cerebral ischemic injury. J Cereb Blood Flow Metab. 2007;27:1553–1562. doi: 10.1038/sj.jcbfm.9600457
62.
Uchida M, Palmateer JM, Herson PS, DeVries AC, Cheng J, Hurn PD. Dose-dependent effects of androgens on outcome after focal cerebral ischemia in adult male mice. J Cereb Blood Flow Metab. 2009;29:1454–1462. doi: 10.1038/jcbfm.2009.60
63.
Choi JW, Ryoo IW, Hong JY, Lee KY, Nam HS, Kim WC, Oh SH, Kang J, Lee HY, Na SJ, et al. Clinical impact of estradiol/testosterone ratio in patients with acute ischemic stroke. BMC Neurol. 2021;21:91. doi: 10.1186/s12883-021-02116-9
64.
Vegeto E, Benedusi V, Maggi A. Estrogen anti-inflammatory activity in brain: a therapeutic opportunity for menopause and neurodegenerative diseases. Front Neuroendocrinol. 2008;29:507–519. doi: 10.1016/j.yfrne.2008.04.001
65.
Mahmoodzadeh S, Dworatzek E. The Role of 17β-Estradiol and estrogen receptors in regulation of Ca2+ channels and mitochondrial function in cardiomyocytes. Front Endocrinol (Lausanne). 2019;10:310. doi: 10.3389/fendo.2019.00310
66.
Wua WF, Tana XJ, Daia YB, Krishnan V, Warner M, Gustafsson JÅ. Targeting estrogen receptor β in microglia and T cells to treat experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA. 2013;110:3543–3548. doi: 10.1073/PNAS.1300313110
67.
Sierra A, Gottfried-Blackmore A, Milner TA, McEwen BS, Bulloch K. Steroid hormone receptor expression and function in microglia. Glia. 2008;56:659–674. doi: 10.1002/glia.20644
68.
Crain JM, Nikodemova M, Watters JJ. Microglia express distinct M1 and M2 phenotypic markers in the postnatal and adult central nervous system in male and female mice. J Neurosci Res. 2013;91:1143–1151. doi: 10.1002/jnr.23242
69.
Thakkar R, Wang R, Wang J, Vadlamudi RK, Brann DW. 17β-Estradiol regulates microglia activation and polarization in the hippocampus following global cerebral ischemia. Oxid Med Cell Longev. 2018;2018:4248526. doi: 10.1155/2018/4248526
70.
Sykes GP, Kamtchum-Tatuene J, Falcione S, Zehnder S, Munsterman D, Stamova B, Ander BP, Sharp FR, Jickling G. Aging immune system in acute ischemic stroke: a transcriptomic analysis. Stroke. 2021;52:1355–1361. doi: 10.1161/STROKEAHA.120.032040
71.
Brockdorff N, Turner BM. Dosage compensation in mammals. Cold Spring Harb Perspect Biol. 2015;7:a019406. doi: 10.1101/cshperspect.a019406
72.
Turtzo LC, Siegel C, McCullough LD. X chromosome dosage and the response to cerebral ischemia. J Neurosci. 2011;31:13255–13259. doi: 10.1523/JNEUROSCI.0621-11.2011
73.
Peeters SB, Cotton AM, Brown CJ. Variable escape from X-chromosome inactivation: identifying factors that tip the scales towards expression. Bioessays. 2014;36:746–756. doi: 10.1002/bies.201400032
74.
Arnold AP, Chen X. What does the “four core genotypes” mouse model tell us about sex differences in the brain and other tissues? Front Neuroendocrinol. 2009;30:1–9. doi: 10.1016/j.yfrne.2008.11.001
75.
Ma Y, Liu Y, Zhang Z, Yang GY. Significance of complement system in ischemic stroke: a comprehensive review. Aging Dis. 2019;10:429–462. doi: 10.14336/AD.2019.0119
76.
Fonseca MI, Chu SH, Hernandez MX, Fang MJ, Modarresi L, Selvan P, MacGregor GR, Tenner AJ. Cell-specific deletion of C1qa identifies microglia as the dominant source of C1q in mouse brain. J Neuroinflammation. 2017;14:48. doi: 10.1186/s12974-017-0814-9
77.
Walker DG, Kim SU, McGeer PL. Complement and cytokine gene expression in cultured microglial derived from postmortem human brains. J Neurosci Res. 1995;40:478–493. doi: 10.1002/jnr.490400407
78.
Chen Y, Won SJ, Xu Y, Swanson RA. Targeting microglial activation in stroke therapy: pharmacological tools and gender effects. Curr Med Chem. 2014;21:2146–2155. doi: 10.2174/0929867321666131228203906
79.
Li J, McCullough LD. Sex differences in minocycline-induced neuroprotection after experimental stroke. J Cereb Blood Flow Metab. 2009;29:670–674. doi: 10.1038/jcbfm.2009.3
80.
Yrjänheikki J, Tikka T, Keinänen R, Goldsteins G, Chan PH, Koistinaho J. A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc Natl Acad Sci USA. 1999;96:13496–13500. doi: 10.1073/pnas.96.23.13496
81.
Amiri-Nikpour MR, Nazarbaghi S, Hamdi-Holasou M, Rezaei Y. An open-label evaluator-blinded clinical study of minocycline neuroprotection in ischemic stroke: gender-dependent effect. Acta Neurol Scand. 2015;131:45–50. doi: 10.1111/ane.12296
82.
Malhotra K, Chang JJ, Khunger A, Blacker D, Switzer JA, Goyal N, Hernandez AV, Pasupuleti V, Alexandrov AV, Tsivgoulis G. Minocycline for acute stroke treatment: a systematic review and meta-analysis of randomized clinical trials. J Neurol. 2018;265:1871–1879. doi: 10.1007/s00415-018-8935-3
83.
Sandvig I, Augestad IL, Håberg AK, Sandvig A. Neuroplasticity in stroke recovery. The role of microglia in engaging and modifying synapses and networks. Eur J Neurosci. 2018;47:1414–1428. doi: 10.1111/ejn.13959
84.
O’Collins VE, Macleod MR, Donnan GA, Horky LL, van der Worp BH, Howells DW. 1,026 experimental treatments in acute stroke. Ann Neurol. 2006;59:467–477. doi: 10.1002/ana.20741
85.
Lyden P, Buchan A, Boltze J, Fisher M; STAIR XI Consortium*. Top priorities for cerebroprotective studies-a paradigm shift: report from STAIR XI. Stroke. 2021;52:3063–3071. doi: 10.1161/STROKEAHA.121.034947
86.
Xu R, Li X, Boreland AJ, Posyton A, Kwan K, Hart RP, Jiang P. Human iPSC-derived mature microglia retain their identity and functionally integrate in the chimeric mouse brain. Nat Commun. 2020;11:1577. doi: 10.1038/S41467-020-15411-9

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Stroke
Stroke
Pages: 1500 - 1509
PubMed: 35468000

History

Published online: 25 April 2022
Published in print: May 2022

Permissions

Request permissions for this article.

Keywords

  1. age
  2. hormones
  3. inflammation
  4. microglia
  5. sex differences
  6. stroke

Subjects

Authors

Affiliations

A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (I.F.U., C.P., P.K., M.G.-B., V.S., P.K., I.S., J.J., T.M.).
Department of Pharmacology, School of Medicine, Tulane University, New Orleans, LA (I.F.U.).
A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (I.F.U., C.P., P.K., M.G.-B., V.S., P.K., I.S., J.J., T.M.).
A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (I.F.U., C.P., P.K., M.G.-B., V.S., P.K., I.S., J.J., T.M.).
Mireia Gómez-Budia, MSc https://orcid.org/0000-0002-2194-2510
A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (I.F.U., C.P., P.K., M.G.-B., V.S., P.K., I.S., J.J., T.M.).
A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (I.F.U., C.P., P.K., M.G.-B., V.S., P.K., I.S., J.J., T.M.).
A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (I.F.U., C.P., P.K., M.G.-B., V.S., P.K., I.S., J.J., T.M.).
Iveta Stanová, MSc
A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (I.F.U., C.P., P.K., M.G.-B., V.S., P.K., I.S., J.J., T.M.).
A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (I.F.U., C.P., P.K., M.G.-B., V.S., P.K., I.S., J.J., T.M.).
A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (I.F.U., C.P., P.K., M.G.-B., V.S., P.K., I.S., J.J., T.M.).

Notes

*
I.F. Ugidos and C. Pistono contributed equally.
For Sources of Funding and Disclosures. see page 1507.
Correspondence to: Tarja Malm, PhD, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland. Email [email protected]

Disclosures

Disclosures None.

Sources of Funding

This study was supported by Academy of Finland.

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. Profiling X chromosome genes expression relevant to sex dimorphism in stroke: insights from transcriptomics landscape analysis, Frontiers in Genetics, 16, (2025).https://doi.org/10.3389/fgene.2025.1479270
    Crossref
  2. Sex differences in age-associated neurological diseases—A roadmap for reliable and high-yield research, Science Advances, 11, 10, (2025).https://doi.org/10.1126/sciadv.adt9243
    Crossref
  3. Salidroside attenuates cognitive deficits induced by chronic cerebral hypoperfusion via modulating microglial phenotypic transformation in mice, Journal of Neuroimmunology, 400, (578544), (2025).https://doi.org/10.1016/j.jneuroim.2025.578544
    Crossref
  4. Characterisation of GPR17 ‐expressing oligodendrocyte precursors in human ischaemic lesions and correlation with reactive glial responses , The Journal of Pathology, 265, 2, (226-243), (2024).https://doi.org/10.1002/path.6381
    Crossref
  5. From acute lung injury to cerebral ischemia: a unified concept involving intercellular communication through extracellular vesicle-associated miRNAs released by macrophages/microglia, Clinical and Experimental Immunology, 219, 1, (2024).https://doi.org/10.1093/cei/uxae105
    Crossref
  6. Glial ‘omics in ischemia: Acute stroke and chronic cerebral small vessel disease, Glia, 73, 3, (495-518), (2024).https://doi.org/10.1002/glia.24634
    Crossref
  7. Concurrent fibrous dysplasia is associated with a high incidence of cystic formation in vestibular schwannomas, Neurosurgical Review, 47, 1, (2024).https://doi.org/10.1007/s10143-024-03061-0
    Crossref
  8. Cerebellar microglia: On the edge between neuroinflammation and neuroregulation, Neural Regeneration Research, 21, 1, (156-172), (2024).https://doi.org/10.4103/NRR.NRR-D-24-00550
    Crossref
  9. Microglial heterogeneity in the ischemic stroke mouse brain of both sexes, Genome Medicine, 16, 1, (2024).https://doi.org/10.1186/s13073-024-01368-7
    Crossref
  10. Sepsis compromises post-ischemic stroke neurological recovery and is associated with sex differences, Life Sciences, 349, (122721), (2024).https://doi.org/10.1016/j.lfs.2024.122721
    Crossref
  11. See more
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

Sex Differences in Poststroke Inflammation: a Focus on Microglia Across the Lifespan
Stroke
  • Vol. 53
  • No. 5

Purchase access to this journal for 24 hours

Stroke
  • Vol. 53
  • No. 5
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Figures

Tables

Media

Share

Share

Share article link

Share

Comment Response