Introduction: Culturing cerebrovascular smooth muscle cells (CVSMCs) in vitro can provide a model for studying many cerebrovascular diseases. This study describes a convenient and efficient method to obtain mouse CVSMCs by enzyme digestion. Methods: Mouse circle of Willis was isolated, digested, and cultured with platelet-derived growth factor-BB (PDGF-BB) to promote CVSMC growth, and CVSMCs were identified by morphology, immunofluorescence analysis, and flow cytometry. The effect of PDGF-BB on vascular smooth muscle cell (VSMC) proliferation was evaluated by cell counting kit (CCK)-8 assay, morphological observations, Western blotting, and flow cytometry. Results: CVSMCs cultured in a PDGF-BB-free culture medium had a typical peak-to-valley growth pattern after approximately 14 days. Immunofluorescence staining and flow cytometry detected strong positive expression of the cell type-specific markers alpha-smooth muscle actin (α-SMA), smooth muscle myosin heavy chain 11 (SMMHC), smooth muscle protein 22 (SM22), calponin, and desmin. In the CCK-8 assay and Western blotting, cells incubated with PDGF-BB had significantly enhanced proliferation compared to those without PDGF-BB. Conclusion: We obtained highly purified VSMCs from the mouse circle of Willis using simple methods, providing experimental materials for studying the pathogenesis and treatment of neurovascular diseases in vitro. Moreover, the experimental efficiency improved with PDGF-BB, shortening the cell cultivation period.

Cerebrovascular diseases are among the most prevalent diseases worldwide. Mechanistic research has demonstrated that cerebrovascular smooth muscle cells (CVSMCs) play an important role in various cerebrovascular diseases [1]. Pathophysiological alterations of cerebrovascular smooth muscle are associated with the development of several cerebrovascular disorders, such as stroke, cerebral edema, Alzheimer’s disease, and atherosclerosis [2]. Furthermore, CVSMCs are key for maintaining and regulating cerebral perfusion. In addition, gap junctions that connect smooth muscle cells are also involved in the transmission of vascular biochemical signals [3]. Smooth muscle cells are essential in regulating cerebrovascular tone, maintaining vascular integrity, and building brain homeostasis under physiological conditions [4‒6]. For example, cerebral atherosclerosis and vascular dysfunction in persistent hypertension lead to abnormal vascular congestion and obvious self-regulation functional abnormalities [7]. In stroke patients, alpha-smooth muscle actin (α-SMA) expression in vascular smooth muscle and the density of cerebral microvessels decrease, suggesting that the stroke process involves postischemic alterations in the cerebral vascular structure. Numerous studies have shown a cause-and-effect relationship between cerebrovascular inflammation and stroke [8, 9]. Specifically, lacunar strokes are thought to result from cerebral vascular lesions [10]. However, during phenotypic modulation, vascular smooth muscle cells (VSMCs) may cause a series of events that promote vascular inflammatory processes [9]. For instance, cerebrovascular injuries can induce severe vasogenic edema [11, 12], and cerebral herniation can be secondary to fluid-induced brain tissue compression [13]. VSMC hyperactivation can also accelerate vascular aging and atherosclerosis [14]. Furthermore, in patients with diabetes, glycosylation products are significantly upregulated in VSMCs, leading to increased microcirculation leakage in the patient’s brain [15].

In vitro culture of CVSMCs is a promising model for studying cerebrovascular disease mechanisms and selecting relevant therapeutic drugs. However, the cerebral vasculature of mice is more fragile than that of rats or their own aortas, making mouse CVSMC isolation very difficult. Therefore, in vitro studies of knockout mice are difficult owing to the technical challenges of primary cell isolation and cultivation. Moreover, most reports of isolated mouse CVSMCs have been limited to primary cultures, and few have reported passaging cells. Therefore, this study describes a simple and reproducible method to conveniently and efficiently obtain CVSMCs from mice and maintain their activity during the passaging process.

Animals

All experiments were performed using 5- to 6-week-old male C57BL/6J mice purchased from the animal experimental center of the Air Force Medical University, China. All animals with license lot number SCXK (2017-0021) were considered satisfactory for the experiment.

Compliance with Ethical Standards

All animal feeding and experimental procedures followed the guidelines of the Institutional Ethics Review Board of the Air Force Medical University.

Primary Culture

6-week-old healthy male C57BL/6J mice were euthanized using an overdose of 1.5% sodium pentobarbital administered via intraperitoneal injection, and the mouse bodies were immersed in 75% ethanol for 3–5 min to disinfect. The mice were sacrificed by cervical dislocation, and then the brains were quickly removed and placed in ice-cold Dulbecco’s phosphate-buffered saline (DPBS; Gibco™, Thermo Fisher Scientific, Waltham, MA, USA) containing 1% penicillin-streptomycin (Gibco™; #15140163). Cerebral arteries (including the circle of Willis and its branches) were rapidly removed under a microscope (Olympus Corporation, Tokyo, Japan; #SZX7) and kept under sterile conditions (shown in Fig. 1). Isolated vessels were rinsed two to three times with DPBS. The vessels were then transferred into centrifuge tubes with collagenase type II solution (1 mg/mL dissolved in DPBS; Sigma-Aldrich, St. Louis, MO, USA) and mixed thoroughly. Next, the centrifuge tubes with the vessels were incubated in a 37°C shaker (80 times/min) for 15 min, after which the arteries were digested into flocculants. Dulbecco’s modified Eagle medium/F-12 (DMEM/F12; Gibco™; #A4192001) with 10% fetal bovine serum (FBS; Biological Industries, Cromwell, CT, USA; #04-001-1ACS) was added to the centrifuge tubes to terminate the enzymatic reaction, mixed, and then centrifuged (1,500 rpm, 5 min). After discarding the supernatant, 2 mL of DMEM/F12 containing 20% FBS and 1% penicillin-streptomycin was added to the centrifuge tube and mixed thoroughly. The above mixture was transferred to culture dishes (pretreated with 0.1% gelatin) and incubated at 37°C in a 5% carbon dioxide (CO2) incubator. We verified whether the tissues and cells had adhered to the wall 48–72 h later. If we found a large amount of unadhered tissues and cells, the culture medium in the dishes were gently transferred to a centrifuge tube for centrifugation (1,000 rpm, 5 min), and the cell debris at the bottom of the dishes were gently washed twice with DPBS (shown in Fig. 2). After centrifugation, 3 mL of DMEM/F12 containing 20% FBS and 1% penicillin-streptomycin was added to the centrifuge tube to resuspend the tissues, which were then transferred to the original culture dishes to continue cultivation. After another 48 h, the culture was rechecked, and the above steps were repeated if there were still unattached cell tissues. Every 48 h after that, the cell growth was checked, and the medium was changed.

Fig. 1.

The circle of Willis and its branches presented as an image of the base of a mouse brain (left, dark red area) and a schematic diagram (right).

Fig. 1.

The circle of Willis and its branches presented as an image of the base of a mouse brain (left, dark red area) and a schematic diagram (right).

Close modal
Fig. 2.

a Schematic diagram detailing the mouse cerebrovascular smooth muscle cell (VSMCs) isolation and purification steps. Isolation: (1) C57BL/6J mice are sacrificed and the brains collected; (2) the circle of Willis and its branches are isolated; (3) the isolated vessels are digested with collagenase type II; (4) the isolated cells and tissues are placed in an incubator for 3 days; (5) cell debris is removed. Based on the experimental requirements, PDGF-BB can be added to the culture medium as appropriate. Purification: (1) after 10–14 days of culture, the cells are digested with trypsin at RT (observed under a microscope); (2) 20–30 s after adding trypsin, the fibroblasts detach and are discarded; (3) fresh trypsin is added, detaching the VSMCs after approximately 2 min. b Tissue and debris after 3 days of cell culture. Cell debris (red arrows) and unadhered tissues or cells (black arrows) are visible at the bottom of the culture dish. c VSMC growth process (n = 10, isolation/culture every 10 animals; n = 3, three biological replicates). Cells emerge from the tissue edge on the 3rd culture day (×100), cover the bottom of the dish on the 7th day (×100), and enter the typical peak-valley growth pattern by the 14th day (×100).

Fig. 2.

a Schematic diagram detailing the mouse cerebrovascular smooth muscle cell (VSMCs) isolation and purification steps. Isolation: (1) C57BL/6J mice are sacrificed and the brains collected; (2) the circle of Willis and its branches are isolated; (3) the isolated vessels are digested with collagenase type II; (4) the isolated cells and tissues are placed in an incubator for 3 days; (5) cell debris is removed. Based on the experimental requirements, PDGF-BB can be added to the culture medium as appropriate. Purification: (1) after 10–14 days of culture, the cells are digested with trypsin at RT (observed under a microscope); (2) 20–30 s after adding trypsin, the fibroblasts detach and are discarded; (3) fresh trypsin is added, detaching the VSMCs after approximately 2 min. b Tissue and debris after 3 days of cell culture. Cell debris (red arrows) and unadhered tissues or cells (black arrows) are visible at the bottom of the culture dish. c VSMC growth process (n = 10, isolation/culture every 10 animals; n = 3, three biological replicates). Cells emerge from the tissue edge on the 3rd culture day (×100), cover the bottom of the dish on the 7th day (×100), and enter the typical peak-valley growth pattern by the 14th day (×100).

Close modal

Previous studies showed that adding 20 ng/mL platelet-derived growth factor-BB (PDGF-BB; PeproTech; Rocky Hill, NJ, USA; #315-18) promotes VSMC proliferation [16]. Therefore, we also cultured cells with 20 ng/mL of PDGF-BB in the medium.

Cell Passage

The cells were passaged when the cell growth density reached 80–90%. The cells were washed twice with DPBS and digested with 0.25% trypsin (Beyotime Biotechnology, Shanghai, China; #C0208-100 mL). The digestion was terminated immediately when a large part of the cells detached (observed under a microscope), which occurred after approximately 2 min at room temperature (RT). The collected cell suspension was centrifuged at 1,000 rpm for 5 min, and then the supernatant was discarded. The cells in the centrifuge tube were mixed with fresh medium and transferred to two new dishes for further culture.

Cell Purification

The diameter of cerebral vessels in mice is small (0.20 ± 0.05 mm) and difficult to isolate; thus, the intima and fibrous outer layer are difficult to remove. Therefore, during cell passaging, the cells can be purified based on the digestion times of the fibroblasts and CVSMCs [17]. Fibroblasts detach from the wall before CVSMCs. Therefore, we microscopically observed the cells during the digestion step of passaging and discarded the cells digested after the first 20–30 s, which were mainly fibroblasts. Then, fresh trypsin was added to complete the passaging process.

There are more CVSMCs than fibroblasts and endothelial cells. Therefore, CVSMCs are proliferatively superior during culturing, but this can inhibit the proliferation of other cells. Moreover, CVSMC purification gradually improves as the number of cells increases. PDGF-BB stimulates VSMC proliferation; therefore, we added PDGF-BB to the cerebral artery CVSMC cultivation since it may enhance smooth muscle cell (SMC) proliferation (shown in Fig. 2) [18].

Morphological Observations

The cells’ shape, size, and growth pattern were observed under a microscope (Olympus Corporation; #CKX53).

Immunofluorescence

The purified VSMCs (second passage after purification) were uniformly plated in laser confocal Petri dishes during the logarithmic growth phase. After 1 day, the medium was discarded. After fixation with 4% paraformaldehyde (diluted in PBS; Beyotime; #P0099-500 mL) for 20 min, the stimulated VSMCs were permeabilized with 0.1% Triton X-100 (Beyotime; #ST797-100 mL) in phosphate-buffered saline (PBS; Beyotime; #C0221A) for 10 min. Cells were blocked with 5% bovine serum albumin (#9048-46-8; Sigma-Aldrich) for 1 h at RT. Anti-α-SMA (Abcam; #ab5694), Anti-smooth muscle myosin heavy chain 11 (SMMHC, ProteinTech; #60222-1-Ig), and Anti-VE cadherin (Abcam; #ab205336) antibodies were added and incubated at 4°C overnight. On the second day, the cells were rewarmed for 30 min at RT and then washed 3 times (5 min each) with PBS. Goat anti-rabbit (ProteinTech; #SA00003-2) or goat anti-mouse (ProteinTech; #SA00013-1) secondary antibodies were added in a dark environment, and the cells were incubated at RT for 1 h. The cells were washed 3 times (5 min each) with PBS, and the nuclei were stained with 4′,6-Diamidino-2-phenylindole (Beyotime; #C1006) following the manufacturer’s instructions. The results were assessed by confocal microscopy (Zeiss, Oberkochen, Germany; #LSM 900).

Flow Cytometry

When the second passage (P2) VSMCs were between 80 and 90% confluent, they were digested, counted, and then inoculated in 6-well plates at 5 × 106 cells/ml and incubated in a 5% CO2 incubator for 24 h. The first set of cells was digested with 0.25% trypsin (without ethylenediaminetetraacetic acid; Beyotime; #C0205) and collected. We used the Annexin V-FITC/Pi Cell Apoptosis Detection Kit (following the manufacturer’s instructions) and flow cytometry (BD FACSAria ™ III; Becton Dickinson, Franklin Lakes, NJ, USA) to analyze VSMC apoptosis (Servicebio, Wuhan, China; #G1511-50T).

The P2 primary CVSMCs were collected and washed twice with PBS, centrifuged at 1,000 rpm for 5 min, then resuspended in PBS. Anti-α-SMA (ProteinTech; #14395-1-AP), anti-smooth muscle protein 22 (SM22; ProteinTech; #60213-1-Ig), anti-calponin 1 (Bioss Antibodies Inc., Woburn, MA, USA; #bs-0095R), and anti-desmin (ProteinTech; #CL488-16520) antibodies were added and processed following the manufacturer’s instructions. Finally, the cells were examined using flow cytometry. Each experiment was repeated 3 times.

Cell Counting Kit-8 Assay

One thousand well-grown VSMCs were inoculated per well in 96-well plates and incubated at 37°C with 5% CO2. The experimental group was incubated with 20 ng/mL PDGF-BB per well; the control group was incubated without PDGF-BB. At the same time, the same volume of PBS was used as the blank control group. All groups were subdivided based on the culture time into 1-, 2-, 3-, 4-, 5-, 6-, and 7-day groups. Each time group had six replicates. Two hours before the maximum incubation time (e.g., at hour 22 of 24), 10 μL of cell counting kit-8 solution (Dojindo, Kumamoto, Japan; #CK04) was added to each well, and the absorbance was measured at 450 nm using a multifunctional enzyme assay detector (BioTek, Winooski, VA, USA; #Synergy H1). In order to normalize the data, the final calculated data were obtained by subtracting the absorbance values of the blank control group from the absorbance values of the experimental group and the control group, respectively. Then, the cell proliferation rate was calculated and plotted using GraphPad Prism 8 (GraphPad Software, San Diego, CA, USA, www.graphpad.com).

Western Blotting

Two passage of primary CVSMCs were collected for protein extraction, each sample was separated by electrophoresis, and then the proteins were transferred to polyvinylidene fluoride membranes (Millipore, Burlington, MA, USA; #IPVH00010). The membranes were blocked for 1 h in tris-buffered saline with Tween (TBST) containing 50 g/L skim milk (pH 8.0), 10 mmol/L Tris-HCl, 150 mmol/L NaCl, and 0.2% Tween-20. Then, various primary antibodies were added and incubated at 4°C overnight. The next day, the membranes were washed 3 times (10 min each) with TBST, and the secondary antibodies were added for 2 h at RT. After, the membranes were washed 3 times (10 min each) with TBST, and an enhanced chemiluminescence reagent (Beyotime; #P0018AM) was added to the membrane for visualization on X-ray film. The optical density of each target protein band was assessed using Fusion FX. EDGE (Vilber Bio Imaging, Paris, France, www.vilber.com) and normalized to the density of the glyceraldehyde 3-phosphate dehydrogenase (GAPDH) band in the same sample. The following primary antibodies were used: anti-α-SMA (ProteinTech; #14395-1-AP), anti-SM22 (ProteinTech; #60213-1-Ig), anti-osteopontin (OPN; Abcam, Cambridge, UK; #ab283656), anti-proliferating cell nuclear antigen (PCNA; ProteinTech; #60097-1-Ig), anti-cyclin D1 (ProteinTech; #60186-1-Ig), anti-p27 (ProteinTech; #67355-1-Ig), mouse anti-GAPDH (ProteinTech; #60004-1-Ig), and rabbit anti-GAPDH (ProteinTech; #10494-1-AP).

Statistical Analyses

Every ten animals were used to isolate/culture primary CVSMCs, and all experiments were repeated 3 times. All data were normally distributed and presented as means ± standard errors. Between-group differences were assessed by one-way analysis of variance and t tests. A p value of <0.05 indicated statistical significance.

Cell Culture

On the first day, only a few pieces of cerebrovascular fragments and cells were attached to the culture dish. After 3 days, a few cells had released from the tissue’s edge. After 7 days, the cells grew in parallel in the culture dish in several shapes and sizes, such as fusiform and ribbon. The cytoplasm and nuclei of the cells also varied. At this stage, the cells tended to be arranged in a net-like array. A typical peak-to-valley growth pattern was observed after approximately 14 days (shown in Fig. 2).

Cell Identification

The second passage cells were cultured and immunofluorescently stained with VSMC-specific marker proteins (α-SMA and SMMHC) and endothelial cell-specific marker (VE-cadherin) proteins. In addition, the nuclei were counterstained with 4′, 6-Diamidino-2-phenylindole, and the images were merged.

Finally, the cells were identified by flow cytometry, and 97.4 ± 1.8% of VSMCs stained positively for α-SMA and SM22, 97.7 ± 2.0% of VSMCs stained positively for calponin and desmin. These are representative results to show the characterization of the VSMC culture and its purity (shown in Figure 3). It is worth noting that although the results in Figure 3 were obtained from P2 CVSMCs, the fluorescence results of P4 CVSMCs were almost the same as those of P2 (shown in online suppl. Data 3; for all online suppl. material, see https://doi.org/10.1159/000532033). This means that the cell phenotype of early passages (P2–P4) are stable.

Fig. 3.

Vascular smooth muscle cell (VSMC) identification by immunofluorescence (P2, n = 3). a The expression and location of alpha-smooth muscle actin (α-SMA; red), smooth muscle myosin heavy chain 11 (SMMHC; red), and VE-cadherin (red); the nuclei are counterstained with 4′, 6-Diamidino-2-phenylindole (DAPI; blue). Scale bar: 100 μm or 50 μm. b The positive expression rates of α-SMA, SM22, calponin, and desmin (n = 3).

Fig. 3.

Vascular smooth muscle cell (VSMC) identification by immunofluorescence (P2, n = 3). a The expression and location of alpha-smooth muscle actin (α-SMA; red), smooth muscle myosin heavy chain 11 (SMMHC; red), and VE-cadherin (red); the nuclei are counterstained with 4′, 6-Diamidino-2-phenylindole (DAPI; blue). Scale bar: 100 μm or 50 μm. b The positive expression rates of α-SMA, SM22, calponin, and desmin (n = 3).

Close modal

PDGF-BB Induces VSMC Proliferation

PDGF-BB promotes VSMC proliferation; thus, we examined how adding PDGF-BB during cell culture affected VSMC dedifferentiation and the cell culture process. PDGF-BB increased VSMC proliferation and decreased the time required to reach the logarithmic growth phase. Also, OPN, α-SMA, and SM22 expression changed, indicating that PDGF-BB induced a phenotypic alteration of VSMCs from differentiation to dedifferentiation. Moreover, cell proliferation markers, such as PCNA, cyclin D1, and p27, are associated with VSMC proliferation and migration. We found markedly upregulated PCNA and cyclin D1 protein levels and decreased p27 protein expression in VSMCs incubated with PDGF-BB (shown in Fig. 4).

Fig. 4.

Platelet-derived growth factor-BB (PDGF-BB) induces vascular smooth muscle cells (VSMCs) to dedifferentiate, proliferate, and migrate. a The VSMC proliferation rate (cell counting kit-8 assay) was significantly higher in the 20 ng/mL PDGF-BB culture group than in the group without PDGF-BB from the third day. *p < 0.05, **p < 0.01, ***p < 0.001; n = 3 per group. b VSMC growth with and without adding 20 ng/mL of PDGF-BB (observed microscopically). PDGF-BB accelerated the cell proliferation rate; the peak-to-valley growth pattern was observed after 1–10 days. c Representative images of osteopontin (OPN), alpha-smooth muscle actin (α-SMA), smooth muscle protein 22 (SM22), proliferating cell nuclear antigen (PCNA), cyclin D1, and p27 protein expression, and the relative protein expression of OPN, α-SMA, PCNA, cyclin D1, and p27 were quantified (presented as means ± standard errors). *p < 0.05, **p < 0.01; n = 3 per group.

Fig. 4.

Platelet-derived growth factor-BB (PDGF-BB) induces vascular smooth muscle cells (VSMCs) to dedifferentiate, proliferate, and migrate. a The VSMC proliferation rate (cell counting kit-8 assay) was significantly higher in the 20 ng/mL PDGF-BB culture group than in the group without PDGF-BB from the third day. *p < 0.05, **p < 0.01, ***p < 0.001; n = 3 per group. b VSMC growth with and without adding 20 ng/mL of PDGF-BB (observed microscopically). PDGF-BB accelerated the cell proliferation rate; the peak-to-valley growth pattern was observed after 1–10 days. c Representative images of osteopontin (OPN), alpha-smooth muscle actin (α-SMA), smooth muscle protein 22 (SM22), proliferating cell nuclear antigen (PCNA), cyclin D1, and p27 protein expression, and the relative protein expression of OPN, α-SMA, PCNA, cyclin D1, and p27 were quantified (presented as means ± standard errors). *p < 0.05, **p < 0.01; n = 3 per group.

Close modal

Effect of PDGF-BB on Apoptosis

The VSMC apoptosis rate per group was detected by flow cytometry. The apoptosis rate of cells cultivated with and without PDGF-BB was 4.0 ± 0.2% and 3.7 ± 0.4%, respectively (shown in Fig. 5); the apoptosis rate did not differ between the groups (p > 0.05).

Fig. 5.

VSMC apoptosis and activity comparisons between cells with and without PDGF-BB treatment. The apoptosis rates did not differ between the groups (n = 3, p = 0.275).

Fig. 5.

VSMC apoptosis and activity comparisons between cells with and without PDGF-BB treatment. The apoptosis rates did not differ between the groups (n = 3, p = 0.275).

Close modal

This study details the CVSMC isolation, primary cell cultivation, purification, and identification from the mouse circle of Willis. Previous reports have described CVSMC isolation, but they were limited to primary culture. Moreover, the reagents they used for cell isolation were complex and difficult to prepare. We compared the new method in this paper with several reported CVSMC isolation and culture methods. Obviously, our method is simpler and cheaper while ensuring cell purity, and the experimental steps in this article are described in detail (shown in Table 1). Therefore, to more conveniently and efficiently isolate and culture mouse CVSMCs, we improved the enzyme digestion time and conditions based on the traditional enzyme digestion method and isolated CVSMCs from the circle of Willis using only a single enzyme. This method is simple, reproducible, and can maintain cell activity during the passaging process. In addition, we increased the cell growth rate by adding PDGF-BB, a VSMC stimulator, to the culture environment, increasing the primary cell culture efficiency.

Table 1.

Comparing our protocol with other studies

SpeciesVesselsMain materialsPurified or notCultivation period
Mice (6 weeks) The circle of Willis and its branches DPBS; 1% penicillin-streptomycin; collagenase type II; DMEM/F12; FBS Yes 10–14 days 
Rats (3 weeks) [19The middle cerebral artery Tyrode’s solution (including papain and dithiothreitol); collagenase; trypsin inhibitor; elastase; Dulbecco’s modified Eagle’s medium; FBS; 1% penicillin-streptomycin 
Rats [20The circle of Willis and its branches PBS; DMEM/F12; NaHCO3; l-ascorbic acid; bovine serum albumin; FBS; epidermal growth factor; fibroblast growth factor; heparin; insulin; penicillin; streptomycin; amphotericin B No 10–20 days 
Dogs (10–12 months) [21Cerebral basilar arteries DMEM/F12; streptomycin; l-glutamine; FBS Yes 
Guinea pigs (200–500 mg) [22Meninges and attached vessels Hanks’ balanced salt solution; 25 mm N-2-hydroxyethylpiperazine-N′-2-ethanesulfonic acid; trypsin; bovine deoxyribonuclease I; calf serum; bovine serum albumin; Neomycin; penicillin; streptomycin; FBS; DPBS; soybean trypsin inhibitor; elastase type III; collagenase type I No 
SpeciesVesselsMain materialsPurified or notCultivation period
Mice (6 weeks) The circle of Willis and its branches DPBS; 1% penicillin-streptomycin; collagenase type II; DMEM/F12; FBS Yes 10–14 days 
Rats (3 weeks) [19The middle cerebral artery Tyrode’s solution (including papain and dithiothreitol); collagenase; trypsin inhibitor; elastase; Dulbecco’s modified Eagle’s medium; FBS; 1% penicillin-streptomycin 
Rats [20The circle of Willis and its branches PBS; DMEM/F12; NaHCO3; l-ascorbic acid; bovine serum albumin; FBS; epidermal growth factor; fibroblast growth factor; heparin; insulin; penicillin; streptomycin; amphotericin B No 10–20 days 
Dogs (10–12 months) [21Cerebral basilar arteries DMEM/F12; streptomycin; l-glutamine; FBS Yes 
Guinea pigs (200–500 mg) [22Meninges and attached vessels Hanks’ balanced salt solution; 25 mm N-2-hydroxyethylpiperazine-N′-2-ethanesulfonic acid; trypsin; bovine deoxyribonuclease I; calf serum; bovine serum albumin; Neomycin; penicillin; streptomycin; FBS; DPBS; soybean trypsin inhibitor; elastase type III; collagenase type I No 

In our laboratory studies, SMCs were isolated from the explants’ edge after approximately 3 days and reached confluence after approximately 14 days. Furthermore, we obtained highly pure VSMCs during the second-generation purification process and identified a typical peak-to-valley growth pattern. Moreover, flow cytometry analyses demonstrated that >95% of cell-specific markers were positively expressed. Immunofluorescence results also confirmed that specific marker proteins were positively expressed. We also found that the phenotype of CVSMCs in the early passages (P2–P4) were stable and could be used for experiments, but the number of passage times and the stability of cell phenotype might be related to the conditions of different laboratories.

We attempted to shorten the cell culture cycle by adding 20 ng/mL of PDGF-BB to the cell culture medium, which greatly enhanced the cell growth rate. Cells cultivated with PDGF-BB reached confluence after 7–10 days and displayed a typical peak-to-valley growth pattern. Compared with the control group, contractile protein expression, such as α-SMA and SM22, decreased in VSMCs after adding PDGF-BB, but OPN expression (a synthetic protein) significantly increased [23]. In VSMCs, PCNA and cyclin D1 are pro-proliferation genes, whereas p27 has an anti-proliferation role [24‒26]. In this study, PCNA and cyclin D1 expression significantly increased in cells incubated with PDGF-BB, whereas p27 expression markedly decreased. These results suggest that PDGF-BB promoted cell proliferation and migration by altering the cytoarchitecture of the VSMCs. Notably, increased PDGF-BB expression indicates tumor angiogenesis in many tumors [27]. In addition, PDGF-BB is also a primary mediator for wound healing and tissue repair [28]. Therefore, if future studies incubate cells with PDGF-BB, its effect on cell structure should be considered.

Minimizing contamination with brain parenchyma and quickly isolating vessels during mouse cerebrovascular isolation is crucial to ensure cell viability. The efficiency of the vessel isolation primarily determines the number of CVSMCs; if it takes too long, fewer primary cells will be obtained in the subsequent cultivation. Therefore, we recommended no more than 10 min between sacrificing the mouse and isolating the blood vessels.

Finally, experimental conditions likely differ among laboratories. Therefore, the optimal enzymatic digestion time should be further explored based on various laboratory conditions. The digestion time is determined based on the degree to which the blood vessels are digested. In this study, blood vessel digestion was observed every 5 min, and the digestion was terminated when the blood vessels were flocculent or ruptured.

We obtained highly purified SMCs from the circle of Willis of mice and improved the experimental efficiency by including PDGF-BB, which shortened the cell cultivation period. These methods may provide ideal experimental material for studying underlying pathophysiological changes in neurovascular system diseases in vitro.

All animal feeding and experimental procedures followed the guidelines of the Institutional Ethics Review Board of the Air Force Medical University.

The authors have no conflicts of interest to declare.

This study was supported by the Shaanxi Provincial Key R & D Plan General Project (2021SF-146, 2022SF-246, 2021SF-253).

Junhui Xue, Wei Chang, Fengzhou Liu, and Yajuan Li were responsible for the conception and design of the research. Wei Chang, Yajuan Li, Peiran Zhang, and Shuai Qu performed the experiments. Wei Chang, Fengzhou Liu, Yajuan Li, Kehai Zang, and Shuai Qu analyzed the data and prepared the figures. Wei Chang and Jingyu Zhao drafted the manuscript. All authors listed contributed to the manuscript and approved the final version.

Additional Information

Wei Chang and Yajuan Li contributed equally to this work and should be considered co-first authors.

All data generated or analyzed during this study are included in this article and its supplementary material files. Further inquiries can be directed to the corresponding author.

1.
Zlokovic
BV
.
Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders
.
Nat Rev Neurosci
.
2011
;
12
(
12
):
723
38
.
2.
Frösen
J
,
Joutel
A
.
Smooth muscle cells of intracranial vessels: from development to disease
.
Cardiovasc Res
.
2018
;
114
(
4
):
501
12
.
3.
Poittevin
M
,
Lozeron
P
,
Hilal
R
,
Levy
BI
,
Merkulova-Rainon
T
,
Kubis
N
.
Smooth muscle cell phenotypic switching in stroke
.
Transl Stroke Res
.
2014
;
5
(
3
):
377
84
.
4.
Belin de Chantemèle
EJ
,
Retailleau
K
,
Pinaud
F
,
Vessières
E
,
Bocquet
A
,
Guihot
AL
.
Notch3 is a major regulator of vascular tone in cerebral and tail resistance arteries
.
Arterioscler Thromb Vasc Biol
.
2008
;
28
(
12
):
2216
24
.
5.
Ellis
PD
,
Chen
Q
,
Barker
PJ
,
Metcalfe
JC
,
Kemp
PR
.
Nov gene encodes adhesion factor for vascular smooth muscle cells and is dynamically regulated in response to vascular injury
.
Arterioscler Thromb Vasc Biol
.
2000
;
20
(
8
):
1912
9
.
6.
Welsh
DG
,
Tran
CHT
,
Hald
BO
,
Sancho
M
.
The conducted vasomotor response: function, biophysical basis, and pharmacological control
.
Annu Rev Pharmacol Toxicol
.
2018
;
58
:
391
410
.
7.
van Sloten
TT
,
Sedaghat
S
,
Carnethon
MR
,
Launer
LJ
,
Stehouwer
CDA
.
Cerebral microvascular complications of type 2 diabetes: stroke, cognitive dysfunction, and depression
.
Lancet Diabetes Endocrinol
.
2020
;
8
(
4
):
325
36
.
8.
Hollander
MC
,
Latour
LL
,
Yang
D
,
Ishii
H
,
Xiao
Z
,
Min
Y
.
Attenuation of myeloid-specific TGFβ signaling induces inflammatory cerebrovascular disease and stroke
.
Circ Res
.
2017
;
121
(
12
):
1360
9
.
9.
Low
A
,
Mak
E
,
Rowe
JB
,
Markus
HS
,
O’Brien
JT
.
Inflammation and cerebral small vessel disease: a systematic review
.
Ageing Res Rev
.
2019
;
53
:
100916
.
10.
Caplan
LR
.
Lacunar infarction and small vessel disease: pathology and pathophysiology
.
J Stroke
.
2015
;
17
(
1
):
2
6
.
11.
Mastorakos
P
,
Mihelson
N
,
Luby
M
,
Burks
SR
,
Johnson
K
,
Hsia
AW
.
Temporally distinct myeloid cell responses mediate damage and repair after cerebrovascular injury
.
Nat Neurosci
.
2021
;
24
(
2
):
245
58
.
12.
Marcolini
E
,
Stretz
C
,
DeWitt
KM
.
Intracranial hemorrhage and intracranial hypertension
.
Emerg Med Clin North Am
.
2019
;
37
(
3
):
529
44
.
13.
Liberale
L
,
Montecucco
F
,
Tardif
JC
,
Libby
P
,
Camici
GG
.
Inflamm-ageing: the role of inflammation in age-dependent cardiovascular disease
.
Eur Heart J
.
2020
;
41
(
31
):
2974
82
.
14.
Cui
X
,
Chopp
M
,
Zacharek
A
,
Ye
X
,
Roberts
C
,
Chen
J
.
Angiopoietin/Tie2 pathway mediates type 2 diabetes induced vascular damage after cerebral stroke
.
Neurobiol Dis
.
2011
;
43
(
1
):
285
92
.
15.
Lu
QB
,
Wan
MY
,
Wang
PY
,
Zhang
CX
,
Xu
DY
,
Liao
X
.
Chicoric acid prevents PDGF-BB-induced VSMC dedifferentiation, proliferation and migration by suppressing ROS/NFκB/mTOR/P70S6K signaling cascade
.
Redox Biol
.
2018
;
14
:
656
68
.
16.
Biela
SA
,
Su
Y
,
Spatz
JP
,
Kemkemer
R
.
Different sensitivity of human endothelial cells, smooth muscle cells and fibroblasts to topography in the nano-micro range
.
Acta Biomater
.
2009
;
5
(
7
):
2460
6
.
17.
Higashi
Y
,
Mummidi
S
,
Sukhanov
S
,
Yoshida
T
,
Noda
M
,
Delafontaine
P
.
Minocycline inhibits PDGF-BB-induced human aortic smooth muscle cell proliferation and migration by reversing miR-221- and −222-mediated RECK suppression
.
Cell Signal
.
2019
;
57
:
10
20
.
18.
Pan
S
,
Lin
H
,
Luo
H
,
Gao
F
,
Meng
L
,
Zhou
C
.
Folic acid inhibits dedifferentiation of PDGF-BB-induced vascular smooth muscle cells by suppressing mTOR/p70s6K signaling
.
Am J Transl Res
.
2017
;
9
(
3
):
1307
16
.
19.
Guo
Y
,
Wang
S
,
Liu
Y
,
Fan
L
,
Booz
GW
,
Roman
RJ
.
Accelerated cerebral vascular injury in diabetes is associated with vascular smooth muscle cell dysfunction
.
Geroscience
.
2020
;
42
(
2
):
547
61
.
20.
Mariana
M
,
Roque
C
,
Baltazar
G
,
Cairrao
E
.
In vitro model for ischemic stroke: functional analysis of vascular smooth muscle cells
.
Cell Mol Neurobiol
.
2022
;
42
(
7
):
2289
304
.
21.
Su
J
,
Li
J
,
Li
W
,
Altura
BT
,
Altura
BM
.
Cocaine induces apoptosis in cerebral vascular muscle cells: potential roles in strokes and brain damage
.
Eur J Pharmacol
.
2003
482
1–3
61
6
.
22.
Seidel
MF
,
Simard
JM
,
Hunter
SF
,
Campbell
GA
.
Isolation of arteriolar microvessels and culture of smooth muscle cells from cerebral cortex of Guinea pig
.
Cell Tissue Res
.
1991
;
265
(
3
):
579
87
.
23.
Xie
C
,
Guo
Y
,
Zhu
T
,
Zhang
J
,
Ma
PX
,
Chen
YE
.
Yap1 protein regulates vascular smooth muscle cell phenotypic switch by interaction with myocardin
.
J Biol Chem
.
2012
;
287
(
18
):
14598
605
.
24.
Han
JH
,
Lee
SG
,
Jung
SH
,
Lee
JJ
,
Park
HS
,
Kim
YH
.
Sesamin inhibits PDGF-mediated proliferation of vascular smooth muscle cells by upregulating p21 and p27
.
J Agric Food Chem
.
2015
;
63
(
33
):
7317
25
.
25.
Liao
XH
,
Wang
N
,
Zhao
DW
,
Zheng
DL
,
Zheng
L
,
Xing
WJ
.
STAT3 protein regulates vascular smooth muscle cell phenotypic switch by interaction with myocardin
.
J Biol Chem
.
2015
;
290
(
32
):
19641
52
.
26.
Xue
Y
,
Lim
S
,
Yang
Y
,
Wang
Z
,
Jensen
LD
,
Hedlund
EM
.
PDGF-BB modulates hematopoiesis and tumor angiogenesis by inducing erythropoietin production in stromal cells
.
Nat Med
.
2011
;
18
(
1
):
100
10
.
27.
Jian
K
,
Yang
C
,
Li
T
,
Wu
X
,
Shen
J
,
Wei
J
.
PDGF-BB-derived supramolecular hydrogel for promoting skin wound healing
.
J Nanobiotechnology
.
2022
;
20
(
1
):
201
.
28.
Evrova
O
,
Bürgisser
GM
,
Ebnöther
C
,
Adathala
A
,
Calcagni
M
,
Bachmann
E
.
Elastic and surgeon friendly electrospun tubes delivering PDGF-BB positively impact tendon rupture healing in a rabbit Achilles tendon model
.
Biomaterials
.
2020
;
232
:
119722
.