Astrocytes are thought to play a pivotal role in coupling neural activity and cerebral blood flow. However, it has been shown that astrocytes undergo morphologic changes in response to brain metastasis, switching to a reactive phenotype, which has the potential to significantly compromise cerebrovascular function and contribute to the neurological sequelae associated with brain metastasis. Given that STAT3 is a key regulator of astrocyte reactivity, we aimed here to determine the impact of STAT3-mediated astrocyte reactivity on neurovascular function in brain metastasis. Rat models of brain metastasis and ciliary neurotrophic factor were used to induce astrocyte reactivity. Multimodal imaging, electrophysiology, and IHC were performed to determine the relationship between reactive astrocytes and changes in the cerebrovascular response to electrical and physiological stimuli. Subsequently, the STAT3 pathway in astrocytes was inhibited with WP1066 to determine the role of STAT3-mediated astrocyte reactivity, specifically, in brain metastasis. Astrocyte reactivity associated with brain metastases impaired cerebrovascular responses to stimuli at both the cellular and functional level and disrupted astrocyte–endothelial interactions in both animal models and human brain metastasis samples. Inhibition of STAT3-mediated astrocyte reactivity in rats with brain metastases restored cerebrovascular function, as shown by in vivo imaging, and limited cerebrovascular changes associated with tumor growth. Together these findings suggest that inhibiting STAT3-mediated astrocyte reactivity may confer significant improvements in neurological outcome for patients with brain metastases and could potentially be tested in other brain tumors.

Significance:

These findings demonstrate that selectively targeting STAT3-mediated astrocyte reactivity ameliorates the cerebrovascular dysfunction associated with brain metastasis, providing a potential therapeutic avenue for improved patient outcome.

Neurovascular coupling matches local cerebral blood flow (CBF) to neuronal energy use and, thus, ensures normal functioning of the brain (1). While neuronal-, astrocyte-, and pericyte-mediated pathways have been implicated in neurovascular coupling to varying degrees (2), the significant contribution of astrocytes remains the most widely recognized (1–3).

Astrocyte processes cover most of the cerebrovascular surface and connect neurons with blood vessels (3–5), playing essential roles in recycling ions and neurotransmitters (3), supplying energy substrates to neurons (6), maintaining endothelial tight junctions to support the blood–brain barrier (BBB; ref. 7), and releasing vasoactive molecules that regulate vascular tone (1). However, in response to disease or injury, astrocytes become reactive, displaying hypertrophy, upregulation of intermediate filament proteins (e.g., GFAP; ref. 8), and metabolic changes (9). The consequences of this astrocyte reactivity, however, on the control of CBF remains unknown (3).

Changes in astrocyte function may be particularly pertinent in brain metastasis, where tumor cells interact closely with blood vessels. Such perivascular growth enables tumor cells to use the brain's vasculature for nutrient supply (10–13), but also disrupts astrocyte-vascular coupling and BBB integrity (12). At the same time, brain metastases trigger astrocyte reactivity from the earliest stages (10–13), and gradually expel astrocyte end-feet to the border of the tumor (14). Notably, STAT3-mediated astrocyte reactivity, specifically, has been shown to be important in maintaining the vascular co-option profile of brain metastases (15). Consequently, brain metastases may impact on cerebrovascular function not only through mechanical dissociation of astrocyte end-feet from vessels (12), but also through astrocyte reactivity per se and, as such, may contribute to the neurologic sequelae associated with brain metastases. However, a lack of suitable animal models and imaging techniques that are able to separate these two processes, to date, have limited our ability to fully understand their relative contributions.

The aim of this study, therefore, was to determine the impact of reactive astrocytes, specifically, on cerebrovascular function using animal models of brain metastasis and ciliary neurotrophic factor (CNTF)-induced astrocyte reactivity (16, 17), together with human brain metastasis biopsies. The CNTF model induces astrocyte reactivity, as observed in disease, in the absence of any other confounding pathology. Subsequently, the potential of inhibiting STAT3-mediated astrocyte reactivity to maintain vascular function was determined, as a potential adjuvant therapy for brain metastasis. Cortical vascular and neuronal function were measured using a multimodal imaging strategy involving MRI, laser speckle contrast imaging (LSCI) and local field potential (LFP) measurements, in combination with postmortem IHC analysis.

All animal experiments were approved by the University of Oxford Clinical Medicine Ethics Review Committee and UK Home Office [Animals (Scientific Procedures) Act 1986], and conducted in accordance with the University of Oxford Policy on the Use of Animals in Scientific Research, the ARRIVE Guidelines and Guidelines for the Welfare and Use of Animals in Cancer Research (18). No overt clinical signs or weight loss were evident, in any cohort, over the experimental period. For in vivo experiments, blood gases were within the normal ranges for animals under isoflurane anesthesia (Supplementary Table S1). No effects of WP1066 treatment on blood gases and mean arterial blood pressure were observed (Supplementary Table S1).

Metastatic tumor cell line, lentivirus, and drug

For the brain metastasis model, an ENU1564 tumor cell line was used (kind gift from G. Stoica, Texas A&M University, Texas), which originated from an N-ethyl-N-nitrosourea–induced mammary adenocarcinoma in a female Berlin-Druckrey IX (BD-IX) rat, and is highly metastatic to brain and bone tissues. As a rat cell line, there is no publicly available short tandem repeat profile for the cells, thus they have not been externally validated. Cells were tested for Mycoplasma immediately prior to freezing each stock vial using MycoAlert Kit (Lonza). Stock vials were revived, grown for up to 1 week in DMEM supplemented with 10% FBS and 1% 200 mmol/L glutamine, harvested by trypsinization, washed and then injected, as described previously (19).

For the reactive astrocyte model, lentivirus-mediated gene transfer of CNTF (CNTF-Lv) was used to induce sustained production of CNTF by neurons, which is released and then gives rise to stable activation of astrocytes via the STAT3 pathway, as described previously (9, 16, 17). To this end, self-inactivated lentiviruses encoding either the human CNTF gene or the (control) β-galactosidase gene (LacZ-Lv), under the control of the mouse phosphoglycerate kinase I promoter, were used (9, 17). In the CNTF-Lv model, transfected neurons display a normal phenotype in terms of morphology, expression of neuronal proteins and spontaneous electrophysiological activity (9, 17).

For STAT3 inhibition, a potent inhibitor that can cross the BBB, WP1066 (Millipore) was injected intraperitoneal daily (3 mg/kg) using DMSO as a vehicle.

Experimental models

All experimental procedures used in this study were approved by the UK Home Office. Two cohorts of female BD-IX rats (Charles River), 6–12 weeks old, were injected intracortically in the whisker barrel somatosensory cortex (2 mm posterior, 5 mm lateral to Bregma, and 1 mm from the cortical surface). One cohort of rats was injected with either 103 metastatic ENU1564 cells in PBS (n = 8) or PBS alone (n = 6). The second cohort of rats was injected with either CNTF-Lv (n = 9) or LacZ-Lv (n = 7) diluted in PBS at a concentration of 100 ng p24/μL. The third cohort of rats was injected with 103 metastatic ENU1564 cells (n = 10). Animals injected with ENU1564 cells were randomly distributed for intraperitoneal injection of either 50 μL WP1066 in DMSO (3 mg/kg, n = 5) or 50 μL DMSO (n = 5).

All animals were transcardially perfusion-fixed under terminal anesthesia at the end of the experiment for histologic analysis. For full details, please see Supplementary Methods.

MRI

All MRI experiments were performed on a horizontal bore 9.4T spectrometer driven by an Agilent DirectDrive console (Agilent Technologies). Animals were anesthetized with 1%–2% isoflurane in 70%N2/30%O2 and positioned in a 72-mm quadrature volume transmit coil with 4-channel phased-array surface receiver (RAPID Biomedical GmbH). In some animals, a cannula was positioned in the tail vein prior to the MRI experiment for gadolinium-DTPA (Gd-DTPA) injection to identify potential BBB breakdown.

Multimodal acquisitions included T1- and T2-weighted MRI and arterial spin labeling (ASL) MRI. For technical details, please visit the Supplementary Methods section.

LSCI

Through the cranial window, superficial (ascending venules and pial vessels) and deep blood vessels (penetrating arterioles and capillary beds) within the cortex were visible (0.5–1 mm deep). Animals underwent LSCI on the day following MRI measurements. For full details regarding LSCI setup and surgery, see Supplementary Data (20, 21).

LFPs

In a subset of animals, LFPs were recorded to measure neuronal responses to the same electrical stimulation of the whisker pad (see above). In this case, burr holes were drilled over both barrel cortices and an electrode (0.155 mm diameter, Teflon insulated platinum; Bilaney Consultants Ltd.) inserted to a depth of 0.5 mm under stereotaxic control, as described previously (22). Electrode signals were recorded using a CED 1902 isolated pre-amplifier and signal processor (Cambridge Electronic Design limited), which was controlled using Spike2.7 data acquisition and analysis package software (Cambridge Electronic Design limited). Electrode signals were acquired with a waveform sampling rate of 10 kHz, 2 μs per time unit resolution and without notch filtering. Low-frequency noise was eliminated from the electrode signal using a 50/60 Hz noise eliminator (Hum Bug, Quest scientific).

MRI data analysis

Signal intensity on T1- and T2-weighted images in the ipsilateral and contralateral cortices of animals was quantified using ImageJ software package (NIH; ref. 23). CBF maps were quantified with nonlinear perfusion signal modelling (24) using the BASIL (Bayesian Inference for Arterial Spin Labeling MRI) toolset in FSL version 5.0 (FMRIB Software Library). For more details regarding data analysis, please see Supplementary Data (24).

LSCI data analysis

LSCI datasets were redefined and downsampled to 1 Hz for data analysis. To quantify the time course of the CBF response after electrical stimuli, datasets were averaged over 10 trials and regions of interest (ROI) drawn around areas corresponding to the whisker barrel cortex. The CBF response and its associated time series were extracted from the ROIs in both ipsilateral (left) and contralateral (right) whisker barrel cortices. More information regarding data acquisition can be found in Supplementary Data.

LFP data analysis

At the end of the LFP recording period, waveform and sampling times were converted into a MATLAB file using Spike 2.7 software. The data were aligned with the stimulation time series and binned at 100Hz. LFP responses to whisker pad stimulation were extracted from 0 to 16 seconds after stimulus using in-house MATLAB scripts. To quantify neuronal response magnitudes, amplitude (mV) was calculated from the average response to the first pulse in the stimulation train (over a 50 ms period following stimulation onset), as described previously (22).

IHC and immunofluorescence

All animals were transcardially perfusion fixed under terminal anesthesia with 0.9% heparinized saline followed by periodate lysine paraformaldehyde (PLP) containing only 0.025% glutaraldehyde (PLPlight) at the end of LSCI and LFP measurements. The brains were postfixed, cryoprotected, embedded in tissue-tek (Sakura Finetek Europe) and frozen in isopentane at −40°C. Frozen, 20 μm thick, serial sections spanning the somatosensory cortex were cut from fixed tissue and mounted on gelatin-coated glass slides. IHC was performed with antibodies against markers of activated astrocytes (1:300, GFAP, Dako), STAT3 (1:300, STAT3α, Cell Signaling Technology), neurons (1:500, NeuN, Millipore), blood vessels (1:100, CD31, Abcam), microglia [1:200, OX42 (cd11b/c), Abcam], cyclooxygenase-1 and 2 (1:100, COX-1/2; Abcam), inducible isoform of nitric oxide synthetase (1:50, iNOS, Abcam), nitrotyrosine (1:400, Millipore), alpha-smooth muscle actin (1:100, αSMA; Abcam), β-dystroglycan (1:200, Abcam), Claudin-5 (1:200, Abcam) and cytochrome p450-4A (1:100, Abcam), the enzyme responsible for the biosynthesis of 20-hydroxyeicosatetraenoic acid (20-HETE).

Immunofluorescent microscopy was performed to identify reactive astrocytes (GFAP), microglia (OX42), neurons (NeuN), blood vessels (CD31), cyclooxygenase-1 and 2 (COX-1/2), inducible isoform of nitric oxide synthase (iNOS), cytochrome p450-4A, αSMA, β-dystroglycan, and Claudin-5. More details about the performed protocols can be found in Supplementary Methods.

IHC and immunofluorescence analysis

Photomicrographs of each brain section were obtained by scanning each section with Aperio Image Analysis (Leica), or using a Nikon Microscope (Nikon E800) coupled to a RoHS camera (RoHS). Subsequently, images were analyzed using ImageScope (Leica) and Qcapture Pro software (Qimaging), respectively.

Immunofluorescent images were acquired using an inverted confocal microscope (LSM-710, Carl Zeiss Microimaging) or Leica DM IRBE (Leica). Detection ranges were set to eliminate cross-talk between fluorophores: 409–485 nm for AMCA, 494–553 nm for Alexa Fluo 488 and 561–595 nm for Cy3/Texas Red. Images were analyzed using in ImageJ software package (NIH; ref. 23). For full information regarding the colocalization studies, please see Supplementary Data.

Statistical analysis

All data are given as mean ± SEM. Statistical analysis was performed using GraphPad Prism (GraphPad Software, Inc.). Unpaired or paired t tests were used to compare data between ipsilateral and contralateral cortices. Unpaired t tests were used to identify differences between DMSO and WP1066 groups. The relationships between ASL-CBF maps and areas of reactive astrocytes were tested by a bivariate correlation analysis, including calculation of the Pearson correlation coefficient (r).

Perivascular growth of focally induced brain metastases

One week after intracortical injection of ENU1564 cells, 94% of all brain metastases were associated with blood vessels (Fig. 1A–C; P < 0.001 compared with nonassociated), indicating that focally induced metastases were predominantly perivascular. Of the blood vessels associated with ENU1564 cells, the majority (74%) were capillaries or small veins (<8.5 μm diameter (Fig. 1D; ref. 25), rather than arterioles, arteries, and large veins (>8.5 μm; ref. 25; P < 0.001 small vs. large).

Figure 1.

Brain metastasis growth disrupts both neurovascular coupling and astrocyte–blood vessel interaction. A, Schematic of the experimental design. B, Photomicrograph showing IHC detection of blood vessels (CD31, brown) in a rat injected with 103 ENU1564 cells (arrows), in both cortices (ipsilateral and contralateral). C, Graph showing percentage of metastatic ENU1564 cells attached to the vasculature as quantified by presence of CD31 staining within a metastasis. N = 4 animals; unpaired t test; mean ± SEM; ***, P < 0.001. D, Graph showing classification of vessels in the tumor area according to the 8.5-μm-diameter threshold in animals injected with ENU1564 cells (N = 4 animals; 4 sections per animal). Unpaired t test; mean ± SEM; ***, P < 0.001. E, LSCIs showing cortical blood flow in an animal injected with ENU1564 cells before and during whisker barrel stimulation [*, injection site; anterior-to-posterior axis (A) and left-to-right (R) axes]. F, Graphs showing CBF response and ΔCBF amplitude [arbitrary unit (a.u); see Supplementary Methods] in both cortices of ENU1564-injected animals (N = 8). Mean ± SEM; paired t test, *, P < 0.05. G, Graphs showing LFP responses and LFP amplitude (mV; see Supplementary Methods) to whisker barrel stimulation in both cortices of ENU1564-injected animals (N = 6; mean ± SEM). H, Representative immunofluorescence images showing dissociation of astrocyte end-feet from blood vessels co-opted by perivascular metastases [arrows in (i)] and at lower magnification changes in cellular morphology of astrocytes in the area associated with brain metastases [within dotted line in (ii)]. I, Immunofluorescence images taken from ipsilateral and contralateral cortices of animals injected with either ENU1564 cells or PBS and showing astrocytes (GFAP, blue), blood vessels (CD31, red), and β-dystroglycan (βDG, green). To assess the interaction of astrocytes with blood vessels, quantitation of red and blue pixels within the green channel was measured in both cortices. Graphs showing percentage colocalization of CD31, GFAP, and β-dystroglycan staining on both sides of animals injected with either ENU1564 cells (top; N = 7) or PBS (bottom; N = 3); four sections per animal; paired t test; mean ± SEM; *, P < 0.05. Ipsilateral and injected cortex represented by I and contralateral (C) cortex used as control.

Figure 1.

Brain metastasis growth disrupts both neurovascular coupling and astrocyte–blood vessel interaction. A, Schematic of the experimental design. B, Photomicrograph showing IHC detection of blood vessels (CD31, brown) in a rat injected with 103 ENU1564 cells (arrows), in both cortices (ipsilateral and contralateral). C, Graph showing percentage of metastatic ENU1564 cells attached to the vasculature as quantified by presence of CD31 staining within a metastasis. N = 4 animals; unpaired t test; mean ± SEM; ***, P < 0.001. D, Graph showing classification of vessels in the tumor area according to the 8.5-μm-diameter threshold in animals injected with ENU1564 cells (N = 4 animals; 4 sections per animal). Unpaired t test; mean ± SEM; ***, P < 0.001. E, LSCIs showing cortical blood flow in an animal injected with ENU1564 cells before and during whisker barrel stimulation [*, injection site; anterior-to-posterior axis (A) and left-to-right (R) axes]. F, Graphs showing CBF response and ΔCBF amplitude [arbitrary unit (a.u); see Supplementary Methods] in both cortices of ENU1564-injected animals (N = 8). Mean ± SEM; paired t test, *, P < 0.05. G, Graphs showing LFP responses and LFP amplitude (mV; see Supplementary Methods) to whisker barrel stimulation in both cortices of ENU1564-injected animals (N = 6; mean ± SEM). H, Representative immunofluorescence images showing dissociation of astrocyte end-feet from blood vessels co-opted by perivascular metastases [arrows in (i)] and at lower magnification changes in cellular morphology of astrocytes in the area associated with brain metastases [within dotted line in (ii)]. I, Immunofluorescence images taken from ipsilateral and contralateral cortices of animals injected with either ENU1564 cells or PBS and showing astrocytes (GFAP, blue), blood vessels (CD31, red), and β-dystroglycan (βDG, green). To assess the interaction of astrocytes with blood vessels, quantitation of red and blue pixels within the green channel was measured in both cortices. Graphs showing percentage colocalization of CD31, GFAP, and β-dystroglycan staining on both sides of animals injected with either ENU1564 cells (top; N = 7) or PBS (bottom; N = 3); four sections per animal; paired t test; mean ± SEM; *, P < 0.05. Ipsilateral and injected cortex represented by I and contralateral (C) cortex used as control.

Close modal

Neurovascular coupling is disrupted in areas of brain metastasis

LSCI measurement of CBF responses to electrical stimulation of the whisker pad in vivo, revealed a focal region of hyperaemia in the contralateral (normal) cortex during stimulation, but a considerably reduced response in the ipsilateral (metastasis bearing) cortex of ENU1564-injected animals (Fig. 1E). Quantitatively, the CBF response was significantly reduced in the ipsilateral versus contralateral cortex (∼50%; Fig. 1F; P < 0.05). Simultaneous LFP recordings of neuronal responses showed no significant differences between ipsilateral and contralateral cortices (Fig. 1G). No significant differences between ipsilateral and contralateral cortices were seen in PBS-injected animals for either CBF or LFP responses (Supplementary Fig. S1A–S1C). Immunohistochemically, no differences in the number of NeuN-positive neurons between cortices in either group (excluding metastatic foci) were evident (Supplementary Fig. S1D). Together, these findings suggest that neurovascular coupling is disrupted in the presence of brain metastases.

Brain metastases alter BBB-astrocyte morphology and induce reactive astrocytes

Within the metastatic foci, astrocyte end-feet appeared to be displaced from blood vessels owing to the perivascular growth pattern of the metastatic cells [mean displacement = 31.3 ± 2.4 μm; Fig. 1H (i)]. At the same time, a widespread area of reactive astrocytes was evident around the metastatic foci in the ipsilateral cortex [Fig. 1H (ii), dotted line, 0.54 ± 0.11 mm2]. Moreover, a significant decrease in colocalization between β-dystroglycan, GFAP and CD31 was evident throughout the area of reactive astrocytes in ENU1564 animals (Fig. 1I; Table 1; P < 0.05), suggesting detachment of astrocyte end-feet from the vascular endothelium unrelated to the direct mechanical effects of the metastatic cells. No decrease in colocalization was observed in PBS-injected animals (Fig. 1I; Table 1).

Table 1.

Percentage colocalization of CD31, GFAP, and β-dystroglycan staining in ipsilateral and contralateral cortices of animals injected with either ENU1564 cells (N = 7), PBS (N = 3), CNTF lentivirus (N = 7), or control lentivirus (N = 3); four sections per animal.

CD31/βDG GFAP/βDG GFAP/CD31
% colocalization Ipsi Contra Ipsi Contra Ipsi Contra
ENU cells  13 ± 2*  23 ± 4  71 ± 2  72 ± 2  40 ± 7  46 ± 3 
PBS  29 ± 2  32 ± 3  41 ± 5  47 ± 6  41 ± 6  37 ± 4 
Lv-CNTF  18 ± 3**  40 ± 5  66 ± 9  51 ± 4  45 ± 2*  57 ± 3 
Lv-LacZ  42 ± 5  45 ± 3  44 ± 5  34 ± 10  35 ± 5  31 ± 3 
CD31/βDG GFAP/βDG GFAP/CD31
% colocalization Ipsi Contra Ipsi Contra Ipsi Contra
ENU cells  13 ± 2*  23 ± 4  71 ± 2  72 ± 2  40 ± 7  46 ± 3 
PBS  29 ± 2  32 ± 3  41 ± 5  47 ± 6  41 ± 6  37 ± 4 
Lv-CNTF  18 ± 3**  40 ± 5  66 ± 9  51 ± 4  45 ± 2*  57 ± 3 
Lv-LacZ  42 ± 5  45 ± 3  44 ± 5  34 ± 10  35 ± 5  31 ± 3 

Note: Paired t test; mean ± SEM; *, P < 0.05; **, P < 0.01.

Abbreviations: Ipsi, ipsilateral; Contra, contralateral; Lv-CNTF, CNTF lentivirus, Lv-CNTF; Lv-LacZ, control lentivirus.

Despite the apparent dissociation of astrocyte end-feet from the vasculature, BBB integrity remained intact, as determined by post-Gd-DTPA T1-weighted MRI (Supplementary Fig. S1E) and IHC detection of Claudin-5, a key tight junctional protein in BBB paracellular permeability (Supplementary Fig. S1F and S1G). Moreover, although blood vessels associated with metastatic cells were abnormal in appearance (Supplementary Fig. S1H), no change in blood vessel number was evident in either metastatic foci or areas of reactive astrocytes (Supplementary Fig. S1I).

CNTF-induced reactive astrocytes

To assess the effect of reactive astrocytes per se on neurovascular function, we used a model of CNTF-induced reactive astrocytes (Fig. 2A; ref. 17). Six weeks after intracortical injection, an area of reactive astrocytes was observed in CNTF-Lv animals (Fig. 2B; 0.46 ± 0.09 mm2), while negligible astrocyte reactivity was observed in control LacZ-Lv animals (Fig. 2B). Reactive astrocytes induced by CNTF-Lv did not alter BBB integrity, as determined by post-Gd-DTPA T1-weighted MRI (Fig. 2C) and Claudin-5/CD31 (vessel marker) colocalization (Supplementary Fig. S2A), neuronal number (Supplementary Fig. S2B), or blood vessel numbers (Supplementary Fig. S2C).

Figure 2.

Reactive astrocytes disrupt both neurovascular coupling and astrocyte–blood vessel interaction. A, Schematic of the experimental design. B, Photomicrographs showing IHC detection of reactive astrocytes (GFAP; brown) in the ipsilateral cortex of animals injected with either CNTF-Lv (left) or the control LacZ-Lv (right). C, T2- and gadolinium-enhanced T1-weighted magnetic resonance images showing that BBB integrity remained intact in the cortex of animals injected with CNTF-Lv at 6 weeks postinjection. Graph shows signal intensity in the cortices (paired t test; mean ± SEM, n = 3). Box indicates region of reactive astrocytes. D, LSCIs showing cortical blood flow in an animal injected with CNTF-Lv prestimulation and poststimulation of the whisker barrel [*, injection site; anterior-posterior (A) axis and left-to-right (R) axes]. E, Graphs showing CBF responses and ΔCBF amplitudes (a.u; see Supplementary Methods) to whisker barrel stimulation in cortices of CNTF-Lv–injected animals (N = 8). Mean ± SEM; paired t test; **, P < 0.01. F, Graphs showing LFP responses and LFP amplitudes (mV; see Supplementary Methods) to whisker barrel stimulation in both sides of CNTF-Lv–injected animals (N = 7; mean ± SEM). G, Immunofluorescence images of animals injected with either CNTF-Lv or the control LacZ-Lv showing astrocytes (GFAP, blue, AMCA), blood vessels (CD31, red, Texas Red) and β-dystroglycan (β-DG, green, A488). Graphs show percentage colocalization of CD31, GFAP, and β-dystroglycan staining of animals injected with either CNTF (top; N = 7) or control LacZ-Lv (bottom; N = 4); four sections per animal; paired t test; mean ± SEM; *, P < 0.05. Ipsilateral/injected cortex represented by I and contralateral (C) cortex used as control.

Figure 2.

Reactive astrocytes disrupt both neurovascular coupling and astrocyte–blood vessel interaction. A, Schematic of the experimental design. B, Photomicrographs showing IHC detection of reactive astrocytes (GFAP; brown) in the ipsilateral cortex of animals injected with either CNTF-Lv (left) or the control LacZ-Lv (right). C, T2- and gadolinium-enhanced T1-weighted magnetic resonance images showing that BBB integrity remained intact in the cortex of animals injected with CNTF-Lv at 6 weeks postinjection. Graph shows signal intensity in the cortices (paired t test; mean ± SEM, n = 3). Box indicates region of reactive astrocytes. D, LSCIs showing cortical blood flow in an animal injected with CNTF-Lv prestimulation and poststimulation of the whisker barrel [*, injection site; anterior-posterior (A) axis and left-to-right (R) axes]. E, Graphs showing CBF responses and ΔCBF amplitudes (a.u; see Supplementary Methods) to whisker barrel stimulation in cortices of CNTF-Lv–injected animals (N = 8). Mean ± SEM; paired t test; **, P < 0.01. F, Graphs showing LFP responses and LFP amplitudes (mV; see Supplementary Methods) to whisker barrel stimulation in both sides of CNTF-Lv–injected animals (N = 7; mean ± SEM). G, Immunofluorescence images of animals injected with either CNTF-Lv or the control LacZ-Lv showing astrocytes (GFAP, blue, AMCA), blood vessels (CD31, red, Texas Red) and β-dystroglycan (β-DG, green, A488). Graphs show percentage colocalization of CD31, GFAP, and β-dystroglycan staining of animals injected with either CNTF (top; N = 7) or control LacZ-Lv (bottom; N = 4); four sections per animal; paired t test; mean ± SEM; *, P < 0.05. Ipsilateral/injected cortex represented by I and contralateral (C) cortex used as control.

Close modal

Neurovascular coupling is disrupted in areas of reactive astrocytes

LSCI measurements in CNTF-Lv–injected animals indicated a significant reduction in the amplitude of the CBF response to whisker barrel stimulation in the ipsilateral cortex (∼70%) compared with the contralateral cortex (Fig. 2D and E; P < 0.01). No significant difference was observed in control LacZ-Lv–injected animals (Supplementary Fig. S2D and S2E). As for the metastasis model, LFP responses to electrical stimulation were unaltered in both groups of animals (Fig. 2F; Supplementary Fig. S2F), supporting the concept that neurovascular coupling is disrupted when astrocytes become reactive.

Reactive astrocytes alter BBB-astrocyte morphology

Again, a significant decrease in colocalization of β-dystroglycan, GFAP and CD31 was observed (Fig. 2G, Table 1; P < 0.05), which was not seen in LacZ-Lv animals (Fig. 2G; Table 1). When assessing colocalization of β-dystroglycan and GFAP alone, no change was found in either ENU1546 or CNTF-Lv models (Table 1), indicating that reactive astrocytes exhibit reduced adhesion to the vascular endothelium rather than reduced expression of β-dystroglycan per se.

Inhibition of STAT3 reduces astrocyte reactivity in response to brain metastasis

Animals injected with ENU1564 cells and treated with WP1066 (Fig. 3A), a selective inhibitor of the JAK/STAT3 pathway mediating astrocyte reactivity (15), showed a significant decrease in the area of astrocyte reactivity compared with vehicle (DMSO)-treated animals for both GFAP (∼55%; Fig. 3B and C; P < 0.05) and STAT3 staining (∼77%; Fig. 3B and C; P < 0.05). However, no significant reduction was evident in the area of metastatic foci (0.63 ± 0.12 vs. 0.55 ± 0.27 mm2; Fig. 3C). In WP1066-treated animals, nuclear STAT3 expression in reactive astrocytes was decreased compared with DMSO-treated animals (Fig. 3B).

Figure 3.

Inhibition of STAT3 in reactive astrocytes improves cerebrovascular function. A, Schematic of the experimental design. B, Photomicrographs showing IHC detection of GFAP (top) and STAT3 (bottom)-positive cells (brown) in the ipsilateral cortices (dotted line) of ENU1564 animals treated with DMSO (control) or WP1066 (STAT3 inhibitor). Arrows, STAT3. C, Graphs showing area of GFAP (top left; n = 5), STAT3 (bottom left; n = 5), and tumor cells (right) in ipsilateral cortices. Unpaired t test; mean ± SEM; *, P < 0.05. D, LSCIs showing cortical blood flow in an ENU1564 animal treated with DMSO (control) before and during whisker barrel stimulation [*, injection site; anterior-posterior (A) and left-to-right (R) axes]. Graphs showing CBF response and ΔCBF amplitude (a.u; see Supplementary Methods) in animals treated with DMSO (control; bottom; N = 4; mean ± SEM; paired t test, *, P < 0.05). E, LSCIs showing cortical blood flow in animals treated with WP1066 before and during whisker barrel stimulation (*, injection site). Graphs showing CBF response and ΔCBF amplitude (a.u; see Supplementary Methods) in animals treated with WP1066 (bottom; N = 4; mean ± SEM). F, Graphs showing LFP responses and LFP amplitude (mV; see Supplementary Methods) to whisker barrel stimulation in animals treated with DMSO (control; N = 3; mean ± SEM). G, Graphs showing LFP responses and LFP amplitude (mV; see Supplementary Methods) to whisker barrel stimulation in animals treated with WP1066 (bottom; N = 4; mean ± SEM). For all graphs, cortices of ENU1564 animals are referred to as ipsilateral (I) and contralateral (C).

Figure 3.

Inhibition of STAT3 in reactive astrocytes improves cerebrovascular function. A, Schematic of the experimental design. B, Photomicrographs showing IHC detection of GFAP (top) and STAT3 (bottom)-positive cells (brown) in the ipsilateral cortices (dotted line) of ENU1564 animals treated with DMSO (control) or WP1066 (STAT3 inhibitor). Arrows, STAT3. C, Graphs showing area of GFAP (top left; n = 5), STAT3 (bottom left; n = 5), and tumor cells (right) in ipsilateral cortices. Unpaired t test; mean ± SEM; *, P < 0.05. D, LSCIs showing cortical blood flow in an ENU1564 animal treated with DMSO (control) before and during whisker barrel stimulation [*, injection site; anterior-posterior (A) and left-to-right (R) axes]. Graphs showing CBF response and ΔCBF amplitude (a.u; see Supplementary Methods) in animals treated with DMSO (control; bottom; N = 4; mean ± SEM; paired t test, *, P < 0.05). E, LSCIs showing cortical blood flow in animals treated with WP1066 before and during whisker barrel stimulation (*, injection site). Graphs showing CBF response and ΔCBF amplitude (a.u; see Supplementary Methods) in animals treated with WP1066 (bottom; N = 4; mean ± SEM). F, Graphs showing LFP responses and LFP amplitude (mV; see Supplementary Methods) to whisker barrel stimulation in animals treated with DMSO (control; N = 3; mean ± SEM). G, Graphs showing LFP responses and LFP amplitude (mV; see Supplementary Methods) to whisker barrel stimulation in animals treated with WP1066 (bottom; N = 4; mean ± SEM). For all graphs, cortices of ENU1564 animals are referred to as ipsilateral (I) and contralateral (C).

Close modal

Inhibition of STAT3 in reactive astrocytes improves cerebrovascular function

As expected, (cf. Fig. 1F), DMSO-treated ENU1564 animals, showed a significantly reduced (48%) CBF response to electrical stimulation in the metastasis-bearing cortex compared with the contralateral cortex (Fig. 3D; P < 0.05). In contrast, WP1066-treated ENU1564 animals showed a much smaller and nonsignificant reduction (∼26%) in CBF response compared with the contralateral hemisphere (Fig. 3E). As above, LFP responses to whisker pad stimulation were unaltered in both groups (Fig. 3F and G).

Subsequently, we demonstrated that cerebrovascular responses to hypercapnia (26) were significantly reduced in both ENU1564 (44%) and CNTF-Lv (40%) models, but not in their respective control groups (PBS and LacZ-Lv; Supplementary Fig. S3; P < 0.001 and P < 0.05, respectively). Again, STAT3 inhibition improved the hypercapnia-evoked vascular responses in ENU1564-injected animals, with no significant difference evident between cortices (Supplementary Fig. S3A–S3L).

Together these findings indicate that inhibition of STAT3-mediated astrocyte reactivity can, at least partially, reverse cerebrovascular dysfunction associated with brain metastases.

Reactive astrocytes reduce basal cerebral blood flow

Measurements of basal CBF using ASL-MRI showed a significant reduction compared with the contralateral cortex of animals injected with either ENU1564 cells or CNTF-Lv (Fig. 4AD; P < 0.01). Notably, the area of reactive astrocytes as a percentage of the somatosensory cortex was much larger than the metastatic foci (40% vs. 3%, respectively), and correlated spatially with the region of reduced CBF for both ENU1564 and CNTF-Lv models (Fig. 4E; P < 0.05; Pearson coefficient correlation r2 = 0.33). In contrast, no changes in basal CBF were observed in PBS or LacZ-Lv control groups (Fig. 4F and G).

Figure 4.

Reactive astrocytes reduce basal cerebral blood flow. A, T2-weighted MRI showing site of injection (arrows) of either metastatic ENU1564 cells or CNTF-Lv and quantitative CBF maps obtained from ASL MRI. B, Photomicrographs showing IHC detection of activated astrocytes (GFAP; brown) in the cortical region of reduced basal CBF and in the contralateral cortex of animals injected with either ENU1564 cells (top; *, injection site) or CNTF-Lv (bottom). C, Graph showing basal CBF values in ipsilateral (I) and contralateral (C) cortices of ENU1564-injected animals (N = 7; mean ± SEM; *, P < 0.05). D, Graph showing basal CBF values in ipsilateral (I) and contralateral (C) cortices of CNTF-Lv–injected animals (N = 6; mean ± SEM; **, P < 0.01). E, Graph showing a significant correlation between reduced basal CBF and area of reactive astrocytes, as measured by GFAP staining in both ENU1564 and CNTF-Lv–injected animals. (r2 = 0.33; *, P < 0.05, N = 13). F, Quantitative CBF maps obtained from ASL-MRI in animals injected with either PBS or LacZ-Lv. Arrows, injection site. G, Graphs showing CBF values animals injected with either PBS or LacZ-Lv (N = 4–7; mean ± SEM). Injected cortices are referred as ipsilateral (I) and contralateral (C). H, T2-weighted MRI showing site of injection (arrows) of metastatic ENU1564 cells and quantitative CBF maps obtained from ASL-MRI. I, Graphs showing basal CBF values in DMSO animals (bottom; N = 4; mean ± SEM; paired t test, *, P < 0.05) or with WP1066 (top; N = 4; mean ± SEM).

Figure 4.

Reactive astrocytes reduce basal cerebral blood flow. A, T2-weighted MRI showing site of injection (arrows) of either metastatic ENU1564 cells or CNTF-Lv and quantitative CBF maps obtained from ASL MRI. B, Photomicrographs showing IHC detection of activated astrocytes (GFAP; brown) in the cortical region of reduced basal CBF and in the contralateral cortex of animals injected with either ENU1564 cells (top; *, injection site) or CNTF-Lv (bottom). C, Graph showing basal CBF values in ipsilateral (I) and contralateral (C) cortices of ENU1564-injected animals (N = 7; mean ± SEM; *, P < 0.05). D, Graph showing basal CBF values in ipsilateral (I) and contralateral (C) cortices of CNTF-Lv–injected animals (N = 6; mean ± SEM; **, P < 0.01). E, Graph showing a significant correlation between reduced basal CBF and area of reactive astrocytes, as measured by GFAP staining in both ENU1564 and CNTF-Lv–injected animals. (r2 = 0.33; *, P < 0.05, N = 13). F, Quantitative CBF maps obtained from ASL-MRI in animals injected with either PBS or LacZ-Lv. Arrows, injection site. G, Graphs showing CBF values animals injected with either PBS or LacZ-Lv (N = 4–7; mean ± SEM). Injected cortices are referred as ipsilateral (I) and contralateral (C). H, T2-weighted MRI showing site of injection (arrows) of metastatic ENU1564 cells and quantitative CBF maps obtained from ASL-MRI. I, Graphs showing basal CBF values in DMSO animals (bottom; N = 4; mean ± SEM; paired t test, *, P < 0.05) or with WP1066 (top; N = 4; mean ± SEM).

Close modal

STAT3 inhibition with WP1066 largely reversed the reduction in basal CBF observed in ENU1564 animals (Fig. 4H and I), with a significant difference compared with DMSO-treated animals (P < 0.05). Given that the area of reactive astrocytes showed a much greater reduction after WP1066 treatment (55%) than the metastatic foci (12%), we suggest that reactive astrocytes, rather than the presence of metastases per se, underlie the reduced basal CBF in metastasis-bearing animals.

STAT3-mediated reactive astrocytes regulate vessel diameter and vasoactive pathways

Large vessels (>8.5 μm) showed significant vasoconstriction in the area of reactive astrocytes within the ipsilateral versus contralateral cortex, in both ENU1564 and CNTF-Lv models (Fig. 5A and B; P < 0.05). Moreover, colocalization between cytochrome p450–4A, an endothelium-dependent vasoconstrictor (27, 28), and αSMA, a marker of smooth muscle cells on large blood vessels (29), was significantly greater in ipsilateral versus contralateral cortex for both models (Fig. 5C and D; Supplementary Table SII; P < 0.05). No differences were evident in control animals (Fig. 5C and D; Supplementary Table SII). No changes in cytochrome p450–4A levels were observed in either astrocytes or microglia (Fig. 5E and F).

Figure 5.

Reactive astrocytes are associated with changes in vasoactive pathways. A, Photomicrographs showing immunohistochemical detection of CD31-positive blood vessels (brown) in different parts of the vascular tree in the ipsilateral cortex of a CNTF-Lv animal. Boxes are shown in higher magnification (i and ii), and diameter measurements of typical small (4.42 mm), medium (7.71 mm) and large vessels (19.1 μm) are shown. B, Graph shows diameters of vessels (>8.5 μm) of CNTF-Lv-injected animals. N = 4, total of 50 blood vessels; mean ± SEM; paired t test, *, P < 0.05. C, Immunofluorescence images showing colocalization (arrows) of cytochrome p450-4A-positive cells (green), αSMA-positive cells (red), and nuclei (DAPI, blue) in CNTF-Lv-injected animals. D, Graphs showing percentage of colocalization between cytochrome p450-4A and αSMA staining in ipsilateral (black bar) and contralateral (white bar) cortices of all four experimental groups. E, Graph shows total number of p450/GFAP-positive pixels. F, Graphs shows total number of p450/OX42-positive pixels. G, Immunofluorescence images showing colocalization of endothelial cells (CD31, green), astrocytes (GFAP, red), and nuclei (blue, DAPI) in core (metastasis) and penumbral (reactive astrocytes) regions of the ipsilateral cortex of ENU1564-injected animals. Graph shows diameters of vessels (<8.5 μm) in ipsilateral (i.e., penumbral region; I) and contralateral (C) cortices (as shown in A). N = 4, total of 50 blood vessels; mean ± SEM; paired t test, ***, P < 0.001. H, Immunofluorescence images showing colocalization of endothelial cells (CD31, green), astrocytes (GFAP, red), and nuclei (blue, DAPI) in the ipsilateral cortex of CNTF-Lv-injected animals. Graph shows diameters of vessels (<8.5 μm) in ipsilateral (I) and contralateral (C) cortices (as shown in A). N = 4, total of 50 blood vessels; mean ± SEM; paired t test, ***, P < 0.001. I, Immunofluorescence images showing colocalization of iNOS (green), astrocytes (GFAP, red), and nuclei (blue, DAPI) in both cortices of CNTF-Lv-injected animals. Graph shows percentage colocalization of iNOS and GFAP in ipsilateral (black bar) and contralateral (white bar) cortices of all experimental groups. J, Immunohistochemical images showing detection of nitrotyrosine (NT) in astrocytes (brown) in ipsilateral and contralateral cortices of CNTF-Lv-injected animals. Graph shows area of NT staining. N = 5 animals; four sections per animal; mean ± SEM; paired t test, **, P < 0.01. K, Immunofluorescence images showing colocalization of COX-1 (green), astrocytes (GFAP, red), and nuclei (blue, DAPI) in ipsilateral (black bar) and contralateral (white bar) cortices of CNTF-Lv-injected animals. Graph shows percentage colocalization of COX-1 and GFAP. All quantitative analysis performed using N = 4 animals; four sections per animal, unless stated otherwise; mean ± SEM; paired t test, *, P < 0.05; **, P < 0.01. L, Graphs showing total number of iNOS/GFAP- (top) and COX1/GFAP- (bottom) positive pixels in the ipsilateral cortices of DMSO (control) or WP1066 experimental groups. N = 4 animals; unpaired t test; mean ± SEM; **, P < 0.01. For all graphs, cortices of ENU1564-animals are referred to as ipsilateral (I) and contralateral (C).

Figure 5.

Reactive astrocytes are associated with changes in vasoactive pathways. A, Photomicrographs showing immunohistochemical detection of CD31-positive blood vessels (brown) in different parts of the vascular tree in the ipsilateral cortex of a CNTF-Lv animal. Boxes are shown in higher magnification (i and ii), and diameter measurements of typical small (4.42 mm), medium (7.71 mm) and large vessels (19.1 μm) are shown. B, Graph shows diameters of vessels (>8.5 μm) of CNTF-Lv-injected animals. N = 4, total of 50 blood vessels; mean ± SEM; paired t test, *, P < 0.05. C, Immunofluorescence images showing colocalization (arrows) of cytochrome p450-4A-positive cells (green), αSMA-positive cells (red), and nuclei (DAPI, blue) in CNTF-Lv-injected animals. D, Graphs showing percentage of colocalization between cytochrome p450-4A and αSMA staining in ipsilateral (black bar) and contralateral (white bar) cortices of all four experimental groups. E, Graph shows total number of p450/GFAP-positive pixels. F, Graphs shows total number of p450/OX42-positive pixels. G, Immunofluorescence images showing colocalization of endothelial cells (CD31, green), astrocytes (GFAP, red), and nuclei (blue, DAPI) in core (metastasis) and penumbral (reactive astrocytes) regions of the ipsilateral cortex of ENU1564-injected animals. Graph shows diameters of vessels (<8.5 μm) in ipsilateral (i.e., penumbral region; I) and contralateral (C) cortices (as shown in A). N = 4, total of 50 blood vessels; mean ± SEM; paired t test, ***, P < 0.001. H, Immunofluorescence images showing colocalization of endothelial cells (CD31, green), astrocytes (GFAP, red), and nuclei (blue, DAPI) in the ipsilateral cortex of CNTF-Lv-injected animals. Graph shows diameters of vessels (<8.5 μm) in ipsilateral (I) and contralateral (C) cortices (as shown in A). N = 4, total of 50 blood vessels; mean ± SEM; paired t test, ***, P < 0.001. I, Immunofluorescence images showing colocalization of iNOS (green), astrocytes (GFAP, red), and nuclei (blue, DAPI) in both cortices of CNTF-Lv-injected animals. Graph shows percentage colocalization of iNOS and GFAP in ipsilateral (black bar) and contralateral (white bar) cortices of all experimental groups. J, Immunohistochemical images showing detection of nitrotyrosine (NT) in astrocytes (brown) in ipsilateral and contralateral cortices of CNTF-Lv-injected animals. Graph shows area of NT staining. N = 5 animals; four sections per animal; mean ± SEM; paired t test, **, P < 0.01. K, Immunofluorescence images showing colocalization of COX-1 (green), astrocytes (GFAP, red), and nuclei (blue, DAPI) in ipsilateral (black bar) and contralateral (white bar) cortices of CNTF-Lv-injected animals. Graph shows percentage colocalization of COX-1 and GFAP. All quantitative analysis performed using N = 4 animals; four sections per animal, unless stated otherwise; mean ± SEM; paired t test, *, P < 0.05; **, P < 0.01. L, Graphs showing total number of iNOS/GFAP- (top) and COX1/GFAP- (bottom) positive pixels in the ipsilateral cortices of DMSO (control) or WP1066 experimental groups. N = 4 animals; unpaired t test; mean ± SEM; **, P < 0.01. For all graphs, cortices of ENU1564-animals are referred to as ipsilateral (I) and contralateral (C).

Close modal

In contrast, a significant increase in the diameters of smaller vessels (<8.5 μm diameter) in the region of astrocyte reactivity was evident in both ENU1564 and CNTF-Lv animals (Fig. 5G and H), compared with controls (PBS and LacZ-Lv; Fig. 5G and H; P < 0.001). Colocalization of the vasodilatory molecule iNOS with astrocytes was significantly greater in ENU1564 and CNTF-Lv animals compared with controls (Fig. 5I; P < 0.05 and P < 0.01; respectively), together with increased cortical levels of nitrotyrosine, a marker of nitric oxide (Fig. 5J; ref. 30). Similarly, astrocytic levels of COX-1, a key enzyme in the arachidonic acid vasodilatory pathway (1), were significantly greater in ipsilateral versus contralateral cortex of ENU1564 and CNTF-Lv groups (Fig. 5K; P < 0.05 and P < 0.01, respectively), but not in controls (PBS and LacZ-Lv; Fig. 5K). COX-1 showed greater colocalization with astrocytes than any other cells (Supplementary Table SIII), suggesting they are the dominant cell population driving COX-1–mediated vasodilation, while both astrocytes and endothelial cells showed the highest levels of iNOS colocalization. A significantly higher number of OX42-positive pixels was found in the ENU1564 group compared with PBS, CNTF-Lv or LacZ-Lv–injected groups (Supplementary Table SIV), confirming an inflammatory microglia/macrophages response in presence of brain metastases (31, 32), but no contribution of microglia/macrophages in the CNTF-Lv model (9, 17).

No changes were observed in the levels of COX-2 in astrocytes, iNOS in endothelial cells, or iNOS/COX-1 in microglia in any of the groups (Supplementary Fig. S4A–S4D), suggesting that vasodilation of small vessels reflects activation of the nitric oxide and arachidonic acid pathways primarily in astrocytes. A significant decrease in both iNOS/GFAP and COX1/GFAP colocalization was evident in WP1066- versus DMSO-treated ENU1564 animals (Fig. 5L). However, neither iNOS nor COX-1 was altered in microglia, endothelial cells or smooth muscle cells following WP1066 treatment (Supplementary Fig. S5A–S5D). In addition, expression of iNOS and COX-1 was not altered in microglia (OX42) in the ipsilateral cortices of ENU1564 animals treated with WP1066 (Supplementary Fig. S6A–S6C). These results indicate that STAT3 inhibition is primarily limited to reactive astrocytes within the metastatic microenvironment.

Reactive astrocytes are associated with vasoactive pathways in human brain metastasis

Reactive astrocytes were evident in human brain metastasis biopsies (Oxford Brain Bank: OBB-SH-1088-2013) primarily in the areas surrounding tumor colonies (Fig. 6A). As in the animal models, colocalization between cytochrome p450–4A and αSMA staining was seen on blood vessels associated with brain metastases (Fig. 6B), and both iNOS and COX-1 staining were present in astrocytes (Fig. 6C and D). The percentage of αSMA- and cytochrome p450–4A–positive blood vessels was greater around metastatic foci compared with control brain (Supplementary Table SII). Significantly greater levels of both iNOS and COX-1 per GFAP-positive pixel, and cytochrome p450–4A per αSMA-positive pixel, were found in the area of reactive astrocytes around metastatic foci compared with nontumor tissues (Fig. 6E; P < 0.001 and P < 0.05, respectively).

Figure 6.

Reactive astrocytes are associated with changes in vasoactive pathways in human brain metastasis tissue. A, Photomicrographs showing IHC detection of reactive astrocytes (GFAP; brown) surrounding brain metastases (*) in human tissue. B, Immunofluorescence images showing colocalization (*) of cytochrome p450–4A–positive cells (green), αSMA (red), and nuclei (blue; DAPI). C, Immunofluorescence images showing colocalization of iNOS (green), astrocytes (GFAP, red) and nuclei (blue; DAPI) in the blood vessels surrounding the tumor colonies. Insets i and ii with * indicate iNOS/GFAP colocalization. D, Immunofluorescence images showing colocalization of COX-1 (green), astrocytes (GFAP, red), and nuclei (blue; DAPI). Insets i and ii with * indicate COX-1/iNOS colocalization. E, Graph shows percentage colocalization of iNOS and GFAP, COX-1/GFAP and p450/αSMA in intratumoral (I; black bar) and extratumoral (E; white bar) regions of human tissue (three brains; four sections per brain). Mean ± SEM; unpaired t test, *, P < 0.05, ***, P < 0.001.

Figure 6.

Reactive astrocytes are associated with changes in vasoactive pathways in human brain metastasis tissue. A, Photomicrographs showing IHC detection of reactive astrocytes (GFAP; brown) surrounding brain metastases (*) in human tissue. B, Immunofluorescence images showing colocalization (*) of cytochrome p450–4A–positive cells (green), αSMA (red), and nuclei (blue; DAPI). C, Immunofluorescence images showing colocalization of iNOS (green), astrocytes (GFAP, red) and nuclei (blue; DAPI) in the blood vessels surrounding the tumor colonies. Insets i and ii with * indicate iNOS/GFAP colocalization. D, Immunofluorescence images showing colocalization of COX-1 (green), astrocytes (GFAP, red), and nuclei (blue; DAPI). Insets i and ii with * indicate COX-1/iNOS colocalization. E, Graph shows percentage colocalization of iNOS and GFAP, COX-1/GFAP and p450/αSMA in intratumoral (I; black bar) and extratumoral (E; white bar) regions of human tissue (three brains; four sections per brain). Mean ± SEM; unpaired t test, *, P < 0.05, ***, P < 0.001.

Close modal

In a rat model of brain metastasis, we have demonstrated impairment of cerebrovascular function, disruption of astrocyte-vascular connections and widespread reactive astrocytes. Studies using a model of CNTF-induced astrocyte reactivity, indicated that reactive astrocytes per se can compromise cerebrovascular function, while inhibition of STAT3-mediated astrocyte reactivity partially reversed the cerebrovascular dysfunction associated with brain metastases. These findings suggest that STAT3-mediated astrocyte reactivity in brain metastasis may contribute to cerebrovascular, and hence neurological, dysfunction in patients.

In glioma, tumor cells displace astrocyte end-feet from blood vessels enabling tumor growth in the perivascular space (12, 33). Similarly, we and others have shown that brain metastases grow in this co-optive fashion, particularly during the micrometastatic stages (10, 11, 34). Here, we show that not only are astrocytes displaced from the vasculature mechanically, but that reactive astrocytes distant from the metastatic foci dissociate from the vasculature. Watkins and colleagues (12) hypothesized that displacement of astrocytes from the vasculature by invading glioma cells prevents vasoactive molecules from reaching endothelial cells, thus disrupting neurovascular coupling (12). While displacement-mediated vascular disruption may also occur in brain metastasis, our findings suggest that widespread reactive astrocytes, outside the spatially restricted metastatic foci, contribute significantly to disruption of vascular function. Critically, a similar compromise of cerebrovascular function was observed in the CNTF-Lv model, in which astrocytes were activated to a reactive phenotype for a prolonged period, while neuronal activity was preserved (17). Similarly, CBF responses to hypercapnia were compromised in both models. To date, only two studies have suggested that vasoactive pathways in astrocytes are involved in CBF responses to hypercapnia (35, 36). Our data support this concept and suggest that astrocyte reactivity may compromise autoregulation.

One possible explanation for the reduced CBF responses to both neuronal activation and hypercapnia is that vasodilation across the vascular bed leads to reduced vascular reserve (37, 38). However, although we found vasodilation in the microvascular network, the arteriolar vessels appeared to be constricted and basal blood flow was reduced, in keeping with restricted arteriolar flow. Moreover, we found more cytochrome p450–4A, the enzyme responsible for the biosynthesis of vasoconstrictor 20-HETE (27, 28), on smooth muscle cells at the arterioles. Consequently, we speculate that suppression of CBF responses to stimuli in the presence of reactive astrocytes may reflect either basal upregulation of vasoconstrictory pathways in the arteriolar bed and/or upregulation of enzymes that mediate vasodilation (iNOS and COX-1) downstream of the arterioles and reduced vascular reserve within the microvascular bed. A recent study has suggested that astrocytes regulate CBF at the capillary and arteriole levels through two distinct mechanisms (39), reinforcing the concept that reactive astrocytes exert differential effects on arterioles and capillaries.

Importantly, upregulation of the above vasoactive mediators was mirrored in all human brain metastasis samples studied, typically at the leading edge of the tumor. Recent studies have identified the JAK/STAT3 pathway in reactive astrocytes as a novel target for neurological diseases (40), brain metastasis (15) and glioblastoma (41). Together with significant antitumor effects (15), data suggest that inhibition of STAT3 may also reduce the immunosuppressive environment of brain tumors induced by cross-talk between astrocytes and microglia (15, 42), implicating this inflammatory microenvironment as a potential target for treatment in brain metastasis (43). Given our data showing that STAT3 inhibition restores impaired cerebrovascular function in brain metastasis, we propose that therapy suppressing astrocyte reactivity may also be effective in reducing metastasis-associated neurocognitive effects and in improving the response of current therapies that are limited by tumor-associated hypoxia and abnormal vasculature (44, 45). Notably, reduced blood flow has been linked to the neurological symptoms associated with brain metastases, such as seizures (46–48) and stroke (49), and this could be investigated in ongoing brain tumor clinical trials with STAT3 inhibitors (e.g., NCT01904123).

With regards to limitations of this study, although we have shown that reactive astrocytes are the dominant cell type producing prostaglandin mediated vasodilation via COX-1 activity, NO-mediated vasodilation is not solely driven by reactive astrocytes. Thus, separating the contributions of different stromal cell populations to the production of vasoactive mediators requires further investigation, potentially using genetically encoded calcium indicators, transgenic techniques, or viral constructs, in combination with in vivo recording of hemodynamic response. In addition, only one brain metastasis model was used in this study. Given that ENU model is well established (19, 50–52), rather than using a second brain metastasis model we considered it to be more relevant to use a model of CNTF-induced reactive astrocytes to determine the effect of reactive astrocytes specifically, in the absence of metastases, on neurovascular function. At the same time, the IHC data obtained from human brain metastasis samples lend further support to our experimental findings. Finally, we did not fully suppress astrocyte reactivity in our models via STAT3 inhibition, suggesting that astrocyte activation may be complex and heterogeneous (53). For example, the NFκB signaling pathway has been shown to contribute to the neurotoxicity of a subpopulation of reactive astrocytes induced by activated neuroinflammatory microglia (54) and, thus, may be an important additional pathway of astrocyte activation in brain metastasis that warrants further investigation.

In summary, this study is the first empirical demonstration that STAT3-mediated astrocyte reactivity associated with brain metastasis contributes to acute cerebrovascular dysfunction. STAT3 inhibition may, therefore, limit detrimental neurologic symptoms associated with disease progression and improve outcome for patients with brain metastases.

J.R. Larkin reports a patent for Algorithm for accurate quantification of perfusion using multi-phase arterial spin labeling MRI pending to University of Oxford. N.R. Sibson reports grants from Cancer Research UK during the conduct of the study. No disclosures were reported by the other authors.

M. Sarmiento Soto: Conceptualization, resources, formal analysis, investigation, methodology, writing-original draft, writing-review and editing. J.R. Larkin: Conceptualization, software, formal analysis, investigation. C. Martin: Data curation, software, investigation. A.A. Khrapitchev: Data curation, software, investigation. M. Maczka: Software, formal analysis, methodology. V. Economopoulos: Methodology. H. Scott: Data curation. C. Escartin: Conceptualization, methodology. G. Bonvento: Conceptualization, methodology. S. Serres: Conceptualization, data curation, software, formal analysis, supervision, funding acquisition, investigation, methodology, writing-original draft, writing-review and editing. N.R. Sibson: Conceptualization, supervision, funding acquisition, writing-original draft, project administration, writing-review and editing.

The authors thank James Meakin for assistance with implementation of the pCASL sequence and G. Stoica (Texas A&M University) for the ENU1564 cells.

This work was supported by Cancer Research UK (grant number C5255/A15935 to N.R. Sibson), a FRB's 4-star research booster scheme from the University of Nottingham and Brain Tumour Charity (GN-000537 to S. Serres) and a Marie-Sklodowska Curie Action-IF (H2020-795695 to M. Sarmiento Soto).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Attwell
D
,
Buchan
AM
,
Charpak
S
,
Lauritzen
M
,
Macvicar
BA
,
Newman
EA
. 
Glial and neuronal control of brain blood flow
.
Nature
2010
;
468
:
232
43
.
2.
Howarth
C
. 
The contribution of astrocytes to the regulation of cerebral blood flow
.
Front Neurosci
2014
;
8
:
103
.
3.
Iadecola
C
,
Nedergaard
M
. 
Glial regulation of the cerebral microvasculature
.
Nat Neurosci
2007
;
10
:
1369
76
.
4.
Mathiisen
TM
,
Lehre
KP
,
Danbolt
NC
,
Ottersen
OP
. 
The perivascular astroglial sheath provides a complete covering of the brain microvessels: an electron microscopic 3D reconstruction
.
Glia
2010
;
58
:
1094
103
.
5.
Kacem
K
,
Lacombe
P
,
Seylaz
J
,
Bonvento
G
. 
Structural organization of the perivascular astrocyte endfeet and their relationship with the endothelial glucose transporter: a confocal microscopy study
.
Glia
1998
;
23
:
1
10
.
6.
Pellerin
L
,
Bouzier-Sore
AK
,
Aubert
A
,
Serres
S
,
Merle
M
,
Costalat
R
, et al
Activity-dependent regulation of energy metabolism by astrocytes: an update
.
Glia
2007
;
55
:
1251
62
.
7.
Abbott
NJ
,
Ronnback
L
,
Hansson
E
. 
Astrocyte-endothelial interactions at the blood-brain barrier
.
Nat Rev Neurosci
2006
;
7
:
41
53
.
8.
Sofroniew
MV
,
Vinters
HV
. 
Astrocytes: biology and pathology
.
Acta Neuropathol
2010
;
119
:
7
35
.
9.
Escartin
C
,
Pierre
K
,
Colin
A
,
Brouillet
E
,
Delzescaux
T
,
Guillermier
M
, et al
Activation of astrocytes by CNTF induces metabolic plasticity and increases resistance to metabolic insults
.
J Neurosci
2007
;
27
:
7094
104
.
10.
Carbonell
WS
,
Ansorge
O
,
Sibson
N
,
Muschel
R
. 
The vascular basement membrane as “soil” in brain metastasis
.
PLoS One
2009
;
4
:
e5857
.
11.
Serres
S
,
Soto
MS
,
Hamilton
A
,
McAteer
MA
,
Carbonell
WS
,
Robson
MD
, et al
Molecular MRI enables early and sensitive detection of brain metastases
.
Proc Natl Acad Sci U S A
2012
;
109
:
6674
9
.
12.
Watkins
S
,
Robel
S
,
Kimbrough
IF
,
Robert
SM
,
Ellis-Davies
G
,
Sontheimer
H
. 
Disruption of astrocyte-vascular coupling and the blood-brain barrier by invading glioma cells
.
Nat Commun
2014
;
5
:
4196
.
13.
Kienast
Y
,
von Baumgarten
L
,
Fuhrmann
M
,
Klinkert
WE
,
Goldbrunner
R
,
Herms
J
, et al
Real-time imaging reveals the single steps of brain metastasis formation
.
Nat Med
2010
;
16
:
116
22
.
14.
Hasko
J
,
Fazakas
C
,
Molnar
K
,
Meszaros
A
,
Patai
R
,
Szabo
G
, et al
Response of the neurovascular unit to brain metastatic breast cancer cells
.
Acta Neuropathol Commun
2019
;
7
:
133
.
15.
Priego
N
,
Zhu
L
,
Monteiro
C
,
Mulders
M
,
Wasilewski
D
,
Bindeman
W
, et al
STAT3 labels a subpopulation of reactive astrocytes required for brain metastasis
.
Nat Med
2018
;
24
:
1024
35
.
16.
Lisovoski
F
,
Akli
S
,
Peltekian
E
,
Vigne
E
,
Haase
G
,
Perricaudet
M
, et al
Phenotypic alteration of astrocytes induced by ciliary neurotrophic factor in the intact adult brain, As revealed by adenovirus-mediated gene transfer
.
J Neurosci
1997
;
17
:
7228
36
.
17.
Escartin
C
,
Brouillet
E
,
Gubellini
P
,
Trioulier
Y
,
Jacquard
C
,
Smadja
C
, et al
Ciliary neurotrophic factor activates astrocytes, redistributes their glutamate transporters GLAST and GLT-1 to raft microdomains, and improves glutamate handling in vivo
.
J Neurosci
2006
;
26
:
5978
89
.
18.
Workman
P
,
Aboagye
EO
,
Balkwill
F
,
Balmain
A
,
Bruder
G
,
Chaplin
DJ
, et al
Guidelines for the welfare and use of animals in cancer research
.
Br J Cancer
2010
;
102
:
1555
77
.
19.
Serres
S
,
Martin
CJ
,
Sarmiento Soto
M
,
Bristow
C
,
O'Brien
ER
,
Connell
JJ
, et al
Structural and functional effects of metastases in rat brain determined by multimodal MRI
.
Int J Cancer
2014
;
134
:
885
96
.
20.
Boas
DA
,
Dunn
AK
. 
Laser speckle contrast imaging in biomedical optics
.
J Biomed Opt
2010
;
15
:
011109
.
21.
Ayata
C
,
Dunn
AK
,
Gursoy
OY
,
Huang
Z
,
Boas
DA
,
Moskowitz
MA
. 
Laser speckle flowmetry for the study of cerebrovascular physiology in normal and ischemic mouse cortex
.
J Cereb Blood Flow Metab
2004
;
24
:
744
55
.
22.
Spain
A
,
Howarth
C
,
Khrapitchev
AA
,
Sharp
T
,
Sibson
NR
,
Martin
C
. 
Neurovascular and neuroimaging effects of the hallucinogenic serotonin receptor agonist psilocin in the rat brain
.
Neuropharmacology
2015
;
99
:
210
20
.
23.
Schneider
CA
,
Rasband
WS
,
Eliceiri
KW
. 
NIH Image to ImageJ: 25 years of image analysis
.
Nat Methods
2012
;
9
:
671
5
.
24.
Groves
AR
,
Chappell
MA
,
Woolrich
MW
. 
Combined spatial and non-spatial prior for inference on MRI time-series
.
Neuroimage
2009
;
45
:
795
809
.
25.
Francois-Dainville
E
,
Buchweitz
E
,
Weiss
HR
. 
Effect of hypoxia on percent of arteriolar and capillary beds perfused in the rat brain
.
J Appl Physiol
1986
;
60
:
280
8
.
26.
Xu
HL
,
Koenig
HM
,
Ye
S
,
Feinstein
DL
,
Pelligrino
DA
. 
Influence of the glia limitans on pial arteriolar relaxation in the rat
.
Am J Physiol Heart Circ Physiol
2004
;
287
:
H331
9
.
27.
Guo
AM
,
Janic
B
,
Sheng
J
,
Falck
JR
,
Roman
RJ
,
Edwards
PA
, et al
The cytochrome P450 4A/F-20-hydroxyeicosatetraenoic acid system: a regulator of endothelial precursor cells derived from human umbilical cord blood
.
J Pharmacol Exp Ther
2011
;
338
:
421
9
.
28.
Escalante
B
,
Omata
K
,
Sessa
W
,
Lee
SG
,
Falck
JR
,
Schwartzman
ML
. 
20-hydroxyeicosatetraenoic acid is an endothelium-dependent vasoconstrictor in rabbit arteries
.
Eur J Pharmacol
1993
;
235
:
1
7
.
29.
Rensen
SS
,
Doevendans
PA
,
van Eys
GJ
. 
Regulation and characteristics of vascular smooth muscle cell phenotypic diversity
.
Neth Heart J
2007
;
15
:
100
8
.
30.
Franze
T
,
Weller
MG
,
Niessner
R
,
Poschl
U
. 
Comparison of nitrotyrosine antibodies and development of immunoassays for the detection of nitrated proteins
.
Analyst
2004
;
129
:
589
96
.
31.
Andreou
KE
,
Soto
MS
,
Allen
D
,
Economopoulos
V
,
de Bernardi
A
,
Larkin
JR
, et al
Anti-inflammatory Microglia/Macrophages as a potential therapeutic target in brain metastasis
.
Front Oncol
2017
;
7
:
251
.
32.
O'Brien
ER
,
Kersemans
V
,
Tredwell
M
,
Checa
B
,
Serres
S
,
Soto
MS
, et al
Glial activation in the early stages of brain metastasis: TSPO as a diagnostic biomarker
.
J Nucl Med
2014
;
55
:
275
80
.
33.
Holash
J
,
Maisonpierre
PC
,
Compton
D
,
Boland
P
,
Alexander
CR
,
Zagzag
D
, et al
Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF
.
Science
1999
;
284
:
1994
8
.
34.
Winkler
F
,
Kienast
Y
,
Fuhrmann
M
,
Von Baumgarten
L
,
Burgold
S
,
Mitteregger
G
, et al
Imaging glioma cell invasion in vivo reveals mechanisms of dissemination and peritumoral angiogenesis
.
Glia
2009
;
57
:
1306
15
.
35.
Niwa
K
,
Haensel
C
,
Ross
ME
,
Iadecola
C
. 
Cyclooxygenase-1 participates in selected vasodilator responses of the cerebral circulation
.
Circ Res
2001
;
88
:
600
8
.
36.
Howarth
C
,
Sutherland
B
,
Choi
HB
,
Martin
C
,
Lind
BL
,
Khennouf
L
, et al
A critical role for astrocytes in hypercapnic vasodilation in brain
.
J Neurosci
2017
;
37
:
2403
14
.
37.
Austin
VC
,
Blamire
AM
,
Allers
KA
,
Sharp
T
,
Styles
P
,
Matthews
PM
, et al
Confounding effects of anesthesia on functional activation in rodent brain: a study of halothane and alpha-chloralose anesthesia
.
Neuroimage
2005
;
24
:
92
100
.
38.
Sicard
K
,
Shen
Q
,
Brevard
ME
,
Sullivan
R
,
Ferris
CF
,
King
JA
, et al
Regional cerebral blood flow and BOLD responses in conscious and anesthetized rats under basal and hypercapnic conditions: implications for functional MRI studies
.
J Cereb Blood Flow Metab
2003
;
23
:
472
81
.
39.
Mishra
A
,
Reynolds
JP
,
Chen
Y
,
Gourine
AV
,
Rusakov
DA
,
Attwell
D
. 
Astrocytes mediate neurovascular signaling to capillary pericytes but not to arterioles
.
Nat Neurosci
2016
;
19
:
1619
27
.
40.
Ben Haim
L
,
Carrillo-de Sauvage
MA
,
Ceyzeriat
K
,
Escartin
C
. 
Elusive roles for reactive astrocytes in neurodegenerative diseases
.
Front Cell Neurosci
2015
;
9
:
278
.
41.
Heiland
DH
,
Gaebelein
A
,
Borries
M
,
Worner
J
,
Pompe
N
,
Franco
P
, et al
Microenvironment-derived regulation of HIF signaling drives transcriptional heterogeneity in glioblastoma multiforme
.
Mol Cancer Res
2018
;
16
:
655
68
.
42.
Henrik Heiland
D
,
Ravi
VM
,
Behringer
SP
,
Frenking
JH
,
Wurm
J
,
Joseph
K
, et al
Tumor-associated reactive astrocytes aid the evolution of immunosuppressive environment in glioblastoma
.
Nat Commun
2019
;
10
:
2541
.
43.
Berghoff
AS
,
Preusser
M
. 
The inflammatory microenvironment in brain metastases: potential treatment target?
Chin Clin Oncol
2015
;
4
:
21
.
44.
Berghoff
AS
,
Ilhan-Mutlu
A
,
Dinhof
C
,
Magerle
M
,
Hackl
M
,
Widhalm
G
, et al
Differential role of angiogenesis and tumour cell proliferation in brain metastases according to primary tumour type: analysis of 639 cases
.
Neuropathol Appl Neurobiol
2015
;
41
:
e41
55
.
45.
Berghoff
AS
,
Breckwoldt
MO
,
Riedemann
L
,
Karimian-Jazi
K
,
Loew
S
,
Schlieter
F
, et al
Bevacizumab-based treatment as salvage therapy in patients with recurrent symptomatic brain metastases
.
Neurooncol Adv
2020
;
2
:
vdaa038
.
46.
Weil
RJ
,
Palmieri
DC
,
Bronder
JL
,
Stark
AM
,
Steeg
PS
. 
Breast cancer metastasis to the central nervous system
.
Am J Pathol
2005
;
167
:
913
20
.
47.
Crivellari
D
,
Pagani
O
,
Veronesi
A
,
Lombardi
D
,
Nole
F
,
Thurlimann
B
, et al
High incidence of central nervous system involvement in patients with metastatic or locally advanced breast cancer treated with epirubicin and docetaxel
.
Ann Oncol
2001
;
12
:
353
6
.
48.
Bendell
JC
,
Domchek
SM
,
Burstein
HJ
,
Harris
L
,
Younger
J
,
Kuter
I
, et al
Central nervous system metastases in women who receive trastuzumab-based therapy for metastatic breast carcinoma
.
Cancer
2003
;
97
:
2972
7
.
49.
Dardiotis
E
,
Aloizou
AM
,
Markoula
S
,
Siokas
V
,
Tsarouhas
K
,
Tzanakakis
G
, et al
Cancer-associated stroke: Pathophysiology, detection and management (Review)
.
Int J Oncol
2019
;
54
:
779
96
.
50.
Ray
KJ
,
Simard
MA
,
Larkin
JR
,
Coates
J
,
Kinchesh
P
,
Smart
SC
, et al
Tumor pH and protein concentration contribute to the signal of amide proton transfer magnetic resonance imaging
.
Cancer Res
2019
;
79
:
1343
52
.
51.
Miller
JJ
,
Grist
JT
,
Serres
S
,
Larkin
JR
,
Lau
AZ
,
Ray
K
, et al
(13)C pyruvate transport across the blood-brain barrier in preclinical hyperpolarised MRI
.
Sci Rep
2018
;
8
:
15082
.
52.
Larkin
JR
,
Simard
MA
,
de Bernardi
A
,
Johanssen
VA
,
Perez-Balderas
F
,
Sibson
NR
. 
Improving delineation of true tumor volume with multimodal MRI in a rat model of brain metastasis
.
Int J Radiat Oncol Biol Phys
2020
;
106
:
1028
38
.
53.
Liddelow
SA
,
Barres
BA
. 
Reactive astrocytes: production, function, and therapeutic potential
.
Immunity
2017
;
46
:
957
67
.
54.
Liddelow
SA
,
Guttenplan
KA
,
Clarke
LE
,
Bennett
FC
,
Bohlen
CJ
,
Schirmer
L
, et al
Neurotoxic reactive astrocytes are induced by activated microglia
.
Nature
2017
;
541
:
481
7
.