Tumor-associated macrophages (TAMs) drive the protumorigenic responses and facilitate tumor progression via matrix remodeling, angiogenesis, and immunosuppression by interacting with extracellular matrix proteins via integrins. However, the expression dynamics of integrin and its correlation with TAM functional programming in the tumors remain unexplored. In this study, we examined surface integrins’ role in TAM recruitment and phenotypic programming in a 4T1-induced murine breast tumor model. Our findings show that integrin α5β1 is upregulated in CD11b+Ly6Chi monocytes in the bone marrow and blood by day 10 after tumor induction. Subsequent analysis revealed elevated integrin α5β1 expression on tumor-infiltrating monocytes (Ly6ChiMHC class II [MHCII]low) and M1 TAMs (F4/80+Ly6ClowMHCIIhi), whereas integrin αvβ3 was predominantly expressed on M2 TAMs (F4/80+Ly6ClowMHCIIlow), correlating with higher CD206 and MERTK expression. Gene profiling of cells sorted from murine tumors showed that CD11b+Ly6GF4/80+α5+ TAMs had elevated inflammatory genes (IL-6, TNF-α, and STAT1/2), whereas CD11b+Ly6GF4/80+αv+ TAMs exhibited a protumorigenic phenotype (IL-10, Arg1, TGF-β, and STAT3/6). In vitro studies demonstrated that blocking integrin α5 and αv during macrophage differentiation from human peripheral blood monocytes reduced cell spreading and expression of CD206 and CD163 in the presence of specific matrix proteins, fibronectin, and vitronectin. Furthermore, RNA sequencing data analysis (GEO dataset: GSE195857) from bone marrow–derived monocytes and TAMs in 4T1 mammary tumors revealed differential integrin α5 and αv expression and their association with FAK and SRC kinase. In line with this, FAK inhibition during TAM polarization reduced SRC, STAT1, and STAT6 phosphorylation. In conclusion, these findings underscore the crucial role of integrins in TAM recruitment, polarization, and reprogramming in tumors.

Tumor-associated macrophages (TAMs) are a prominent component of the tumor microenvironment (TME), accounting for up to 50% of tumor mass (1). These heterogeneous groups of macrophages are functionally and phenotypically diverse and account for the immunosuppressive nature of the microenvironment, with increased TAM counts associated with aggravation of tumor growth, undesirable prognosis, and therapy resistance (1, 2). Signals from the tumor and its microenvironment shape immune cell behavior in the tumor’s stroma. The TME recruits myeloid cells, such as monocytes, granulocytes, and macrophages, which often exhibit an immunosuppressive, proangiogenic, and profibrotic characteristic (3–5).

Several tumor-derived chemoattractants, such as chemokines, ILs, and cytokines, are essential in recruiting myeloid cells into tumors (6, 7). Following recruitment, monocytes differentiate into macrophages and switch their functional phenotypes between classical inflammatory (M1) to an immunosuppressive phenotype (M2) by sensing their environment (8, 9). TAMs are shown to resemble the M2 phenotype, and they are delivered to facilitate tumor progression via matrix remodeling, promoting angiogenesis and contributing to immunosuppression in the tumor environment (10, 11). TAMs are protumorigenic macrophages that produce immunosuppressive cytokines, chemokines, and growth factors such as arginase, IL-10, and TGF-β, inhibiting antitumor immunity and encouraging tumor progression (12, 13).

Integrins are transmembrane heterodimeric glycoproteins that mediate cell migration in coordination with molecules such as GTPases and chemokine receptors (14). Signals from the binding of extracellular matrices (ECMs) with integrins also contribute to various cellular processes, such as adhesion, migration, differentiation, proliferation, and immunological functions (15, 16). Myeloid cells interact with specific ligands within the extracellular matrices of the TME to localize and navigate within the tumor site (17, 18). Integrins can influence the activation and function of myeloid cells in the TME and modulate the balance of proinflammatory and anti-inflammatory responses, affecting the tumor’s overall immune landscape. Our previous study has also demonstrated that integrin α4 is associated with monocyte mobilization from bone marrow to the site of inflammation under systemic inflammatory conditions and that integrins α5 and αv could mediate polarization into an M1 and M2 phenotype (19). Thus, understanding the pivotal role of integrins in infiltrating myeloid cells into tumors is critical for developing immunomodulatory cancer therapeutic strategies that target the integrins or their downstream signaling pathways.

In this study, using M-CSF or MDA-MB-231 tumor supernatant (TS)–derived in vitro–generated TAMs from human peripheral blood monocytes and INF-γ– and LPS–derived M1 polarization or IL-4– and IL-13–derived M2 polarization or 4T1 TS-derived polarization in RAW 264.7 cells and a 4T1-induced murine breast tumor model, we investigated the role of surface integrin in tissue-specific recruitment and functional reprogramming of TAMs during tumor progression. Our study showed that integrin α5β1 drives the recruitment of monocytes from bone marrow and blood to peritoneum and tumor and correlated with the M1 phenotype. In contrast, TAMs expressing high levels of CD206 and MERTK (MER proto-oncogene, tyrosine kinase) exhibited increased αvβ3 expression and contributed to M2-type polarization. Our data derived from gene expression studies in sorted TAMs expressing specific integrin, functional blocking studies, and bioinformatics data analysis demonstrated an essential role of integrins α5β1 and αvβ3 in TAM reprogramming in mammary tumors. Our data show that adhesion receptors such as integrins play an essential role in the recruitment, polarization, and reprogramming of TAMs from inflammatory to immunosuppressive phenotypes in the tumor microenvironment and could be potential targets for TAM-targeting cancer therapeutics.

Human breast adenocarcinoma (MDA-MB-231, MCF-7), pancreas ductal carcinoma (PANC-1, MIA PaCa-2), mouse mammary carcinoma (4T1), and mouse macrophage RAW 264.7 cell lines were obtained from the National Centre for Cell Science (Pune, India). MDA-MB-231 and MIA PaCa-2 cells were cultured in DMEM (HiMedia, Kennett Square, PA) with 10% FBS, whereas MCF-7, PANC-1, 4T1, and RAW 264.7 cells were cultured in RPMI 1640 (Life Technologies) with 10% FBS and 1% antibiotic/antimycotic solution, all at 37°C with 5% CO2. Conditioned medium (TS) was collected by incubating cells in serum-free DMEM or RPMI 1640 for 24 h after reaching 90% confluency. Supernatants were collected and centrifuged at 12,000 × g for 10 min, and aliquots were stored at −80°C for further use.

All human blood monocyte experiments were conducted with blood drawn from healthy donors following protocols endorsed by the Institute Human Ethics Committee, Indian Institute of Technology (Roorkee, India). PBMCs were isolated using Histopaque 1077 (Sigma-Aldrich, St. Louis, MO), followed by careful isolation of monocytes using the plastic adherence method. Briefly, 5 × 105 PBMCs were seeded for 1 h in 400 µl of RPMI 1640 devoid of FBS in a 48-well flat-bottom plate. To differentiate monocytes into macrophages, they were cultured for 7 d in RPMI 1640 medium supplemented with 5% FBS and either TSs or M-CSF (Life Technologies, Thermo Fisher Scientific, Waltham, MA) as described by Chakraborty et al. (20). Cells were harvested at various intervals (days 1, 3, 5, and 7) and prepared for flow cytometry and quantitative PCR (qPCR). In a separate experiment, before adding the TSs, cells were pretreated with 10 µg/ml anti-integrins α5 and αv and 3 μM focal adhesion kinase (FAK) inhibitor (PF-573288) (Sigma-Aldrich, St. Louis, MO) and cultured for days 3 and 7 after seeding.

In additional experiments, monocytes were isolated using the MojoSort human pan monocyte isolation kit (BioLegend, San Diego, CA) and cultured on plates coated with ECM proteins (fibronectin, fibrinogen, vitronectin, and vimentin) (5 μg/ml) (Merck & Co., Rahway, NJ). The plates were then blocked using 2% BSA. Monocytes were cultured for 7 d in RPMI 1640 medium with 5% FBS in the presence of either M-CSF or MDA-MB-231 TS. On day 7, cells were collected and processed for gene analysis. In a different experiment, only fibronectin and vitronectin (5 μg/ml) were coated, and before adding cells to the wells, monocytes were incubated with anti-integrins α5β1 and αvβ3 and the cells were then incubated in RPMI 1640 medium in the presence of MDA-MB-231 TS. After 7 d of culture, flow cytometry was performed.

RAW 264.7 cells were polarized as described previously with modifications (21, 22). Briefly, RAW 264.7 cells were cultured in the presence or absence of FAK inhibitor (PF-573288, 3 μM) followed by stimulation with IFN-γ (20 ng/ml) combined with LPS (20 ng/ml) (Escherichia coli O26:B6; Sigma-Aldrich, St. Louis, MO) or IL-4 (20 ng/ml) combined with IL-13 (20 ng/ml) or culture supernatant of a 4T1 cell line and cultured for 24 and 48 h. The recombinant Abs were purchased from PeproTech (Rocky Hill, NJ). The cells were collected and flow cytometry was performed.

Female BALB/c mice (10–12 wk old) were obtained from the Indian Institute of Science Education and Research (Mohali, India), with approval from the Institute Animal Ethics Committee at the Indian Institute of Technology (Roorkee, India). Tumor induction involved a s.c. injection of 1 × 106 4T1 cells in 100 μl of sterile 1× PBS near the fourth nipple. Tumor growth was assessed periodically by measuring size with vernier calipers and calculating volume using the following equation: V = (L × W2) × 0.50, where V indicates volume, L indicates length, and W indicates the perpendicular width. Mice were sacrificed on days 3, 7, 10, 15, 23, and 30, and various samples, including blood, bone marrow, peritoneal lavage, and lungs, were collected. Tumor samples were obtained on days 10, 15, 23, and 30 for subsequent analysis.

On days 3 and 7, in vitro–generated macrophages derived from human peripheral blood monocytes were detached using ice-cold PBS, followed by surface marker staining using anti-human CD11b-FITC/allophycocyanin, CD14-PerCP, MHC class II (MHCII)–FITC, CD206-PE, CD86-PE/Cy7, and CD163-allophycocyanin/Cy7 Abs. The differentiated RAW 264.7 cells were collected after 24 and 48 h and stained using purified anti-mouse FcR (CD16/CD32), CD11b-BV510 (BD Biosciences, San Diego, CA), F4/80-AF488/PerCP, MHCII-PerCP, CD206(MMR)-BV605, MERTK-allophycocyanin, CD49e-PE/Cy7, and CD51-PE. After surface staining, the cells were immediately fixed and permeabilized using an intracellular fixation and permeabilization buffer set (Thermo Fisher Scientific, Waltham, MA), and intracellular staining was done using phospho–SRC (proto-oncogene tyrosine-protein kinase c-Src)-AF488 (Thermo Fisher Scientific, Waltham, MA), anti-STAT1 phospho-PE, and anti-STAT6 phospho-PE/Cy7.

For the in vivo study, blood, bone marrow, peritoneal lavage, lungs, and tumors were isolated on the above-mentioned days. To prepare single-cell suspensions of lung and tumor, minced lung and tumor were enzymatically digested using collagenase IV (Sigma-Aldrich, St. Louis, MO). Then, the mixture was mechanically disrupted by passing it over a 70-μm cell strainer. Contaminated erythrocytes were eliminated using RBC lysis buffer (BioLegend, San Diego, CA). For surface marker staining, purified anti-mouse FcR (CD16/CD32), CD11b–BV510, Ly6G-BUV395 (BD Biosciences, San Diego, CA), Ly6C-allophycocyanin/Cy7, F4/80-AF488/PerCP, MHCII-PerCP, CD206(MMR)-BV605, CD64-BV786 (BD Biosciences, San Diego, CA), CCR2-FITC, CXCR4-allophycocyanin, MERTK-allophycocyanin, CD49e-PE/Cy7, and CD51-PE Abs were used.

In vitro and in vivo samples were gated using BD Horizon fixable viability stain 450 (FVS450; BD Biosciences, San Jose, CA). All flow cytometry Abs were purchased from BioLegend (San Diego, CA) unless otherwise noted. All samples were fixed with 1% paraformaldehyde (HiMedia, Kennett Square, PA) and collected on a FACSAria Fusion flow cytometer and FACSLyric flow cytometer (BD Biosciences, San Jose, CA).

A single-cell tumor suspension was stained with Abs in a 500-μl vol for 30 min on ice. Live cells were gated using BD Horizon FVS450. Surface marker staining included Abs against purified anti-mouse FcR (CD16/CD32), CD11b-BV510, Ly6G-BUV395, Ly6C-allophycocyanin/Cy7, F4/80-AF488, CD49e-PE/Cy7, and CD51-PE. After washing, cells were filtered through 70- and 40-μm cell strainers without fixation. Cell sorting was performed using the BD FACSAria Fusion sorter. Tumor tissue macrophages were gated based on integrins α5 and αv on FVS450CD11b+Ly6GF4/80+ cells, excluding cell doublets. Sorted cells (α5+, αv+, and α5+αv+) were collected into 5-ml FACS tubes with 2 ml of complete RPMI 1640 medium without FBS. Ly6Chi monocytes from bone marrow served as the control group. The postsorting purity of macrophages was determined, and gene expression was assessed using qPCR.

The total RNA from differentiated macrophages/TAMs generated in vitro and tumor tissues were extracted using TRIzol reagent. The total RNA of cells sorted from tumor tissues was extracted using an RNeasy kit (Qiagen, Germantown, MD). RNA was reverse transcribed using a high-capacity cDNA reverse transcription kit (Applied Biosystems, Foster City, CA). Real-time PCR was carried out according to the manufacturer’s instructions using PowerUp SYBR Green master mix (Applied Biosystems, Foster City, CA). A StepOnePlus real-time PCR system (Applied Biosystems, Forster City, CA) was used for the quantitative real-time PCR analysis. Samples were normalized to the endogenous control (human GAPDH or mouse HPRT), and relative gene expression was measured using the 2−ΔΔCt method. The primers used for the study are listed in Supplemental Table I.

Peripheral blood samples from naive and tumor-induced mice on days 3, 7, 10, 15, 23, and 30 were collected via retro-orbital bleeding. Serum was obtained by allowing blood to clot for 15 min at room temperature, followed by centrifugation at 1800 × g for 5 min at 4°C. IL-6 and IL-10 levels in serum samples were measured using a sandwich ELISA protocol with Abs from PeproTech (Rockey Hill, NJ). The color reaction was initiated with a tetramethylbenzidine liquid substrate system (BioLegend, San Diego, CA) and stopped with 2 N H2SO4 (HiMedia, Kennett Square, PA) before measuring absorbance at 450 nm using a BioTek Epoch 2 plate reader (BioTek, Winooski, VT).

An RNA sequencing dataset (GSE195857) was downloaded from the GEO database. This dataset contains the transcriptome of TAMs, mammary gland macrophages (MGMs) from healthy mammary fat pads, and bone marrow–derived monocytes from healthy and tumor-bearing mice (BMDM-Ts/BMDM-Hs). Quantification of transcript expression was done by the Salmon algorithm (23), followed by quality control. The data were further processed on Bioconductor using the DESeq2 package (24) with test = Wald, fitType = parametric, and a p value cutoff of 0.05 to identify the differentially expressed genes (DEGs). Functional enrichment analysis on DEGs was performed using the goseq package (25). The genes obtained from the top pathway after functional enrichment were mapped to a protein–protein interaction network using the STRING v12.0 database (http://string-db.org/) (26).

Statistical analysis was performed using a one- or two-way ANOVA with a Bonferroni correction. The Holm–Sidak method was used to analyze tumor sizes with various t tests. The values are presented as mean ± SEM, with p values ≤0.05 considered statistically significant across all datasets. Data were analyzed using GraphPad Prism 9.5.1 for Windows (GraphPad Software, La Jolla, CA). All flow cytometry data were analyzed using FlowJo v10.8.1 (Tree Star, Ashland, OR).

To investigate the distribution pattern of monocytes in the TME, flow cytometric analysis was performed on different days (days 3–30) postinduction to determine their recruitment from the bone marrow to blood, tumor, peritoneum, and lungs after induction of 4T1-induced breast tumor. Cells were isolated from the tissues and stained with different myeloid cell surface markers (CD11b, Ly6G, and Ly6C) to differentiate monocytes and neutrophils. CD11b+Ly6GLy6Chi cells were gated to indicate monocytes in various tissues (Fig. 1A). In the bone marrow, the frequency of monocytes increased from day 3 to day 15 after tumor induction followed by gradually decreasing as compared with naive animals (Fig. 1A, first row). Conversely, Ly6Chi monocytes increased in blood after day 10, indicating migration from bone marrow to blood after day 7 (Fig. 1A, second row). The frequencies of Ly6Chi cells in the peritoneum and lung increased steadily from day 3 to 30. However, there was a reduction in Ly6Chi cell frequency in tumor tissues (Fig. 1A).

FIGURE 1.

Distribution of monocytes and macrophages at different time points after tumor induction. (A) FACS plots showing the distribution of monocytes (CD11b+Ly6GLy6Chi) from the parent population (FVS450CD11b+) in the bone marrow, blood, tumor, peritoneum, and lungs of naive and tumor-induced mice at days 3, 7, 10, 15, 23, and 30. (B) The tumor growth index was recorded after the tumor was palpated. The bar diagram shows the tumor volume on days 10, 15, 23, and 30. (C) FACS plots showing the gating strategies used to differentiate monocytes (CD11b+Ly6GLy6Chi and CD11b+Ly6GLy6Clow) and macrophages (CD11b+Ly6GF4/80+) in the tumor, peritoneum, and lungs. (DF) Line diagrams showing the number of Ly6Chi and Ly6Clow and F4/80+ cells in the (D) tumor, (E) peritoneum, and (F) lungs. All values are presented as mean ± SEM (n = 4–6 per group). *p ≤ 0.05, **p ≤ 0.01, ****p ≤ 0.0001.

FIGURE 1.

Distribution of monocytes and macrophages at different time points after tumor induction. (A) FACS plots showing the distribution of monocytes (CD11b+Ly6GLy6Chi) from the parent population (FVS450CD11b+) in the bone marrow, blood, tumor, peritoneum, and lungs of naive and tumor-induced mice at days 3, 7, 10, 15, 23, and 30. (B) The tumor growth index was recorded after the tumor was palpated. The bar diagram shows the tumor volume on days 10, 15, 23, and 30. (C) FACS plots showing the gating strategies used to differentiate monocytes (CD11b+Ly6GLy6Chi and CD11b+Ly6GLy6Clow) and macrophages (CD11b+Ly6GF4/80+) in the tumor, peritoneum, and lungs. (DF) Line diagrams showing the number of Ly6Chi and Ly6Clow and F4/80+ cells in the (D) tumor, (E) peritoneum, and (F) lungs. All values are presented as mean ± SEM (n = 4–6 per group). *p ≤ 0.05, **p ≤ 0.01, ****p ≤ 0.0001.

Close modal

After injection of 4T1 cancer cells in the mice, the growth of mammary tumors was measured on days 10, 15, 23, and 30, showing a significant increase in volume from day 10 to 30 (Fig. 1B). Monocyte distribution in the tumor, peritoneum (the nearest site to tumor tissue), and lungs (a distant site of metastasis) was studied during the course of tumor progression. The study revealed two Ly6C monocyte populations gated as CD11b+Ly6GLy6Chi and CD11b+Ly6GLy6Clow and CD11b+Ly6GF4/80+ macrophages in these tissues (Fig. 1C). As shown in Fig. 1D, in the tumor, there was a decrease in the number of Ly6Chi monocytes and more Ly6Clow monocytes after day 10, alongside a significant increase in the number of F4/80+ macrophages, suggesting differentiation of Ly6Chi monocytes into macrophages (Fig. 1D). Interestingly, peritoneum displayed no substantial change in Ly6Clow monocyte numbers but showed a significant increase in the number of Ly6Chi monocytes and a huge increase in F4/80+ macrophages after day 10, indicating monocyte differentiation (Fig. 1E). In the lungs, there was no substantial change in monocyte/macrophage numbers initially (days 0–7); however, from day 10 on, there was a rise in Ly6Chi and Ly6Clow monocytes, alongside a significant increase in F4/80+ macrophages (Fig. 1F). This sudden increase in the macrophage population in the lungs suggests that monocytes/macrophages have migrated from their precursor sites and infiltrated the lungs in large numbers after tumor induction.

CCR2, a G protein–coupled receptor, is a critical functional receptor for CCL2 that primarily directs the chemotactic responses of monocytes to the site of inflammation (27). Similarly, the CXCR4/CXCL12 axis has been correlated with breast tumor growth, metastasis, and invasion upon binding with the ligands, triggering signaling that orchestrates the retention of cells in the bone marrow. Several studies have correlated high levels of CXCR4 expression in cancers with poor prognosis and resistance to chemotherapy (28, 29). Our analysis revealed the CCR2 and CXCR4 expression in monocytes across bone marrow, blood, tumor, peritoneum, and lungs. In bone marrow and blood, CCR2 expression increased from day 3 to 30 in Ly6Chi monocytes compared with naive animals (Fig. 2A). However, CXCR4 expression decreased in bone marrow from day 3 to 10. It increased in blood, which suggests the migration of monocytes from bone marrow to the blood (Fig. 2B). The histogram overlay demonstrates the significant changes in CCR2 and CXCR4 levels in the bone marrow and blood of naive and day 7 post-4T1 injection mice, indicating early migration activity. This is essential for capturing the immune response in its early stages and the mobilization of monocytes. These findings underscore the regulatory roles of CCR2 and CXCR4 in monocyte migration from bone marrow and blood during tumorigenesis.

FIGURE 2.

Expression kinetics of chemokines and their receptors in different tissues after 4T1 injection. (A and B) Line diagrams represent the MFI and histogram (day 7) of (A) CCR2 and (B) CXCR4 in monocytes (CD11b+Ly6GLy6Chi) in blood and bone marrow at different days (days 3, 7, 10, 15, 23, and 30) after 4T1 injection. (CH) Line diagrams represent the MFI and histogram (day 15) of CCR2 and CXCR4 in Ly6Chi, Ly6Clow, and F4/80+ cells in (C and D) tumor, (E and F) peritoneum, and (G and H) lungs at above-mentioned days postinduction of the tumor (n = 4–6 per group). (I and J) Line diagrams represent the expression kinetics (mRNA fold change) of (I) chemokines (Ccl2, Cxcl12, Ccl3, Ccl5) and (J) chemokine receptors (Ccr2, Cxcr4) in tumor tissues at the days as mentioned above. All mRNA fold changes were calculated with respect to naive mice (mammary gland tissues). Samples were collected from three to four mice per group. All values are presented as mean ± SEM. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

FIGURE 2.

Expression kinetics of chemokines and their receptors in different tissues after 4T1 injection. (A and B) Line diagrams represent the MFI and histogram (day 7) of (A) CCR2 and (B) CXCR4 in monocytes (CD11b+Ly6GLy6Chi) in blood and bone marrow at different days (days 3, 7, 10, 15, 23, and 30) after 4T1 injection. (CH) Line diagrams represent the MFI and histogram (day 15) of CCR2 and CXCR4 in Ly6Chi, Ly6Clow, and F4/80+ cells in (C and D) tumor, (E and F) peritoneum, and (G and H) lungs at above-mentioned days postinduction of the tumor (n = 4–6 per group). (I and J) Line diagrams represent the expression kinetics (mRNA fold change) of (I) chemokines (Ccl2, Cxcl12, Ccl3, Ccl5) and (J) chemokine receptors (Ccr2, Cxcr4) in tumor tissues at the days as mentioned above. All mRNA fold changes were calculated with respect to naive mice (mammary gland tissues). Samples were collected from three to four mice per group. All values are presented as mean ± SEM. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

Close modal

We investigated myeloid cell trafficking from bone marrow to tumor, peritoneum, and lungs, analyzing CCR2 and CXCR4 expression in Ly6Chi/low and F4/80+ cells. In the tumor, Ly6Chi monocytes exhibited higher CCR2 and CXCR4 expression compared with Ly6Clow and F4/80+ cells, whereas CXCR4 increased in F4/80+ macrophages in the tumor after day 15 (Fig. 2C, 2D). Contrary to the monocytes, F4/80+ macrophages showed higher CCR2 and CXCR4 expression in the peritoneum than did Ly6Chi and Ly6Clow monocytes, which were upregulated from day 3 to 30 (Fig. 2E, 2F). In the lungs, CCR2 and CXCR4 expression in Ly6Chi/low, F4/80+ cells resembled naive animals initially (days 0–7) but increased after day 10 of tumor induction, suggesting myeloid cell migration to the lungs after day 10 (Fig. 2G, 2H). The histogram overlay shows the difference between the Ly6Chi/low and F4/80+ populations on day 15, as well as a significant increase in CCR2 and CXCR4 expression levels in the tumor, peritoneum, and lungs.

Chemokines such as CCL2 and CXCL12 participate in the recruitment of monocytes into tumors, with CCR2 and CXCR4 serving as their respective receptors (30). MIP-1α or CCL3 and CCL5 also recruit and activate immune cells, promoting inflammation of the tumor and the progression of the disease (31, 32). The transcriptional expression of these chemokines (Ccl2, Cxcl12, Ccl3, and Ccl5) and receptors (Ccr2 and Cxcr4) in breast tumors was examined from day 0 to day 30. Ccl2 expression significantly peaked from day 0 to day 10 compared with Cxcl12, Ccl3, and Ccl5 (Fig. 2I). Receptors Ccr2 and Cxcr4 showed upregulated expression toward the early time point on day 3, followed by a significant decrease as the tumor progressed (Fig. 2J). The gene expression of CCL2/CCR2 and CXCL12/CXCR4 chemokines decreased significantly as the tumor grew, suggesting mRNA translating into proteins, which correlates with our previous finding on increased levels of CCR2 and CXCR4 in the tumor after day 10.

Integrin receptors mediate the migration of immune cells and are also necessary for immune cell phagocytosis, migration, and cellular adhesion (33). When analyzing Ly6Chi monocytes across bone marrow, blood, tumor, and lungs after tumor induction, we found that among the different integrins (α2, α3, α4, α5, α6, αv) analyzed, the expression of integrin α5β1 was upregulated in the bone marrow and blood as the tumor progressed. Moreover, in the tumor and lungs, αvβ3 was upregulated in tumor-infiltrating monocytes and macrophages (data not shown). Further analysis of integrin α5β1 and αvβ3 expression patterns in bone marrow and blood samples in a series of time points collected on days 3, 7, 10, 15, 23, and 30 after tumor induction revealed upregulated α5β1 expression in Ly6Chi monocytes (Fig. 3A, 3B). The histogram overlay shows that in the bone marrow and blood, the expression of integrin α5β1 significantly increased after day 15 after tumor induction as compared with the naive mice, and there was a significant decrease in the expression of integrin αvβ3 in the bone marrow.

FIGURE 3.

Expression kinetics of integrins α5β1 and αvβ3 in myeloid cell populations after induction of tumor. (A and B) Line diagrams represent the MFI and histogram (day 15) of integrins α5β1 and αvβ3 in monocytes (CD11b+Ly6GLy6Chi) in the (A) bone marrow and (B) blood at days 3, 7, 10, 15, 23, and 30 after inductions of the tumor. (CH) Line diagrams showing the MFI and histogram (day 23) of integrins α5β1 and αvβ3 in Ly6Chi, Ly6Clow, and F4/80+ cells in the (C and D) tumor, (E and F) peritoneum, and (G and H) lungs at above-mentioned days. All values are presented as mean ± SEM (n = 4–6 per group). (IL) Bar diagrams represent the MFI of (I) CD206, (J) CD163, (K) CD86, and (L) MHCII of M-CSF and various TS (MDA-MB-231, MCF-7, PANC-1, and MIA PaCa2)-polarized macrophages at day 7 after seeding. (MP) Line diagrams represent integrins α5 and αv expression kinetics in different TS-polarized macrophages such as (M) MDA-MB-231, (N) MCF-7, (O) PANC-1, and (P) MIA PaCa2 at days 1, 3, 5, and 7 after seeding. Data represent three independent experiments performed in duplicates; n = 6. (Q and R) Bar diagrams represents the MFI of (Q) integrin α5 and (R) integrin αv in RAW 264.7 cells culturing with either media or IFN-γ combined with LPS or IL-4 combined with IL-13 or 4T1 TS for 24 and 48 h. Data represent three independent experiments performed in duplicates; n = 6. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

FIGURE 3.

Expression kinetics of integrins α5β1 and αvβ3 in myeloid cell populations after induction of tumor. (A and B) Line diagrams represent the MFI and histogram (day 15) of integrins α5β1 and αvβ3 in monocytes (CD11b+Ly6GLy6Chi) in the (A) bone marrow and (B) blood at days 3, 7, 10, 15, 23, and 30 after inductions of the tumor. (CH) Line diagrams showing the MFI and histogram (day 23) of integrins α5β1 and αvβ3 in Ly6Chi, Ly6Clow, and F4/80+ cells in the (C and D) tumor, (E and F) peritoneum, and (G and H) lungs at above-mentioned days. All values are presented as mean ± SEM (n = 4–6 per group). (IL) Bar diagrams represent the MFI of (I) CD206, (J) CD163, (K) CD86, and (L) MHCII of M-CSF and various TS (MDA-MB-231, MCF-7, PANC-1, and MIA PaCa2)-polarized macrophages at day 7 after seeding. (MP) Line diagrams represent integrins α5 and αv expression kinetics in different TS-polarized macrophages such as (M) MDA-MB-231, (N) MCF-7, (O) PANC-1, and (P) MIA PaCa2 at days 1, 3, 5, and 7 after seeding. Data represent three independent experiments performed in duplicates; n = 6. (Q and R) Bar diagrams represents the MFI of (Q) integrin α5 and (R) integrin αv in RAW 264.7 cells culturing with either media or IFN-γ combined with LPS or IL-4 combined with IL-13 or 4T1 TS for 24 and 48 h. Data represent three independent experiments performed in duplicates; n = 6. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

Close modal

In our study, we observed differential expression of integrins α5β1 and αvβ3 on monocyte and macrophage subsets in the tumor, peritoneum, and lungs. In tumors, α5β1 expression was higher in Ly6Chi and Ly6Clow cells compared with F4/80+ macrophages, and integrin αvβ3 was upregulated in all three cell populations after day 15 with a more robust upregulation in F4/80+ macrophages (Fig. 3C, 3D). Peritoneal Ly6Chi and Ly6Clow monocytes showed similar integrin expression patterns, with significant upregulation of both integrins in F4/80+ macrophages (Fig. 3E, 3F). In the lungs, both integrins α5β1 and αvβ3 were upregulated in F4/80+ macrophages after day 10 compared with Ly6Chi/low monocytes (Fig. 3G, 3H). The histogram overlay shows that there was a distinct difference in the expression of integrins α5β1 and αvβ3 between these populations, that is, Ly6Chi, Ly6Clow, and F4/80+ cells, during day 23 postinjection as compared with the naive cells. Our study concludes that in the later stage of tumor development, αvβ3 integrin expression is higher in F4/80+ macrophages compared with tumor-infiltrating monocytes in the tumor. In contrast, F4/80+ macrophages also expressed higher α5β1 and αvβ3 in both the peritoneum and lungs.

Integrin profiling of TAMs differentiated from human peripheral blood monocytes using culture supernatants from breast cancer (MDA-MB-231, MCF-7) and pancreatic cancer cell lines (PANC-1, MIA PaCa-2) revealed that MDA-MB-231 TS induced an M2 phenotype with higher CD206 and CD163 and lower MHCII and CD86 expression levels compared with other TS- and M-CSF–treated cells (Fig. 3I–L). Integrin expression profiles in TAMs from different TSs were examined during several days (days 1, 3, 5, and 7 after seeding), showing upregulation of α5 and αv integrins in MDA-MB-231 TAMs compared with control cells cultured with media alone (just media) (Fig. 3M). In contrast, TAMs from MCF-7 TS exhibited initial downregulation and late upregulation of α5 and αv integrins compared with control (Fig. 3N), whereas there was a downregulation of integrins α5 and αv in PANC-1 and MIA PaCa-2 TS-differentiated macrophages (Fig. 3O, 3P). The results show that MDA-MB-231 TS-treated macrophages have higher levels of integrins α5 and αv compared with other culture supernatants, which were used for further in vitro studies. RAW 264.7 cells stimulated with IFN-γ/LPS for M1 macrophages, IL-4/IL-13 for M2 macrophages, or 4T1 TS also showed increased α5 and αv expression compared with controls (media only) (Fig. 3Q, 3R). These findings highlight that MDA-MB-231 TS and 4T1 TS induce significant upregulation of integrins α5 and αv in human peripheral blood monocyte-derived macrophages and RAW 264.7 cells, respectively, suggesting that these integrins are key markers for macrophage polarization in breast cancer.

Previous studies on TAM phenotypic markers in the 4T1-induced tumor model demonstrated a progressive shift from Ly6Chi to Ly6Clow populations, which were further categorized into M1 and M2 TAMs based on MHCII expression (34). According to this study, F4/80+ cells were gated on CD11b+Ly6G populations (Fig. 4A), yielding six groups: Ly6ChiMHCIIlow (gate 1), Ly6CintMHCIIlow (gate 2), Ly6ClowMHCIIlow (M2 TAMs; gate 3), Ly6ChiMHCIIhi (gate 4), Ly6CintMHCIIhi (gate 5), and Ly6ClowMHCIIhi (M1 TAMs; gate 6) as shown in Fig. 4B. The frequency and cell number of M2 TAMs significantly increased from day 10 to day 30 after tumor induction, whereas Ly6ChiMHCIIlow cells were decreased after day 10 (Fig. 4C). The number of Ly6CintMHCIIlow cells increased after day 10 and then decreased subsequently. This suggests the polarization of Ly6Chi cells into M2 TAMs via an intermediate population of the Ly6CintMHCIIlow phenotype. Additionally, the number of M1 TAMs decreased after day 10, possibly indicating repolarization to M2 TAMs later (Fig. 4C). The phenotype of these subsets was also verified by analyzing the surface marker expression levels of CD206, MERTK, and CD64. M2 TAMs displayed higher CD206 and MERTK expression levels compared with M1 TAMs and Ly6Chi cells, whereas Ly6ChiMHCIIlow (gate 1), Ly6CintMHCIIlow (gate 2), Ly6ChiMHCIIhi (gate 4), and Ly6CintMHCIIhi (gate 5) populations exhibited higher CD64 expression compared with both M1 and M2 TAMs (Fig. 4D–F).

FIGURE 4.

M1 and M2 TAMs in tumor expressed differential α5β1 and αvβ3 expression. (A) Flow cytometry plots represent the gating strategy used to study F4/80+ cells in tumors. (B) Flow cytometry plots represent the gating strategy and frequency of different subsets of tumor-infiltrated (CD11b+Ly6GF4/80+) TAMs at days 10, 15, 23, and 30 after tumor induction. (C) Line diagram represents the cell number of Ly6ChiMHCIIlow (gate 1), Ly6CintMHCIIlow (gate 2), Ly6ClowMHCIIlow (gate 3), Ly6ChiMHCIIhi (gate 4), Ly6CintMHCIIhi (gate 5), and Ly6ClowMHCIIhi (gate 6) cells in F4/80+ cells at different time points. (DF) Line diagrams represent the MFI of (D) CD206, (E) MERTK, and (F) CD64 in different subsets of F4/80+ cells at different time points. (G and H) Histogram overlays of integrins α5 and αv of different subsets of F4/80+ cells at days 10, 15, 23, and 30. (I and J) Line diagrams represent the expression pattern and MFI of α5 and αv in different F4/80+ TAM subsets after tumor induction. All values are presented as mean ± SEM (n = 4–6 per group). *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

FIGURE 4.

M1 and M2 TAMs in tumor expressed differential α5β1 and αvβ3 expression. (A) Flow cytometry plots represent the gating strategy used to study F4/80+ cells in tumors. (B) Flow cytometry plots represent the gating strategy and frequency of different subsets of tumor-infiltrated (CD11b+Ly6GF4/80+) TAMs at days 10, 15, 23, and 30 after tumor induction. (C) Line diagram represents the cell number of Ly6ChiMHCIIlow (gate 1), Ly6CintMHCIIlow (gate 2), Ly6ClowMHCIIlow (gate 3), Ly6ChiMHCIIhi (gate 4), Ly6CintMHCIIhi (gate 5), and Ly6ClowMHCIIhi (gate 6) cells in F4/80+ cells at different time points. (DF) Line diagrams represent the MFI of (D) CD206, (E) MERTK, and (F) CD64 in different subsets of F4/80+ cells at different time points. (G and H) Histogram overlays of integrins α5 and αv of different subsets of F4/80+ cells at days 10, 15, 23, and 30. (I and J) Line diagrams represent the expression pattern and MFI of α5 and αv in different F4/80+ TAM subsets after tumor induction. All values are presented as mean ± SEM (n = 4–6 per group). *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

Close modal

Because integrins α5 and αv were differentially expressed in the F4/80+ cells in the tumor, the expression pattern and kinetics of these two integrins were examined at various time points on M1 and M2 TAMs and intermediate populations. As shown in Fig. 4Gand 4H, the shift of the histograms and the increase in the mean fluorescence intensity (MFI) values suggest that M1 TAMs, M2 TAMs, and Ly6CintMHCIIlow populations showed an upregulated expression of integrin α5 after day 10. In contrast, Ly6ChiMHCIIhi (gate 4) and Ly6CintMHCIIhi (gate 5) populations showed elevated expression of integrin α5 on days 10 and 15, then decreased significantly, but there were no significant changes in the Ly6ChiMHCIIlow cells. Similarly, integrin αv expression significantly increased in the M2 TAM population compared with Ly6Chi cells and M1 TAMs (Fig. 4I, 4J). The Ly6ChiMHCIIhi and Ly6CintMHCIIhi populations also showed elevated expression of αv on day 10 and reduced expression after day 15 (Fig. 4I, 4J). This suggests that M2 TAMs expressed significantly higher levels of integrins α5 and αv and gradually increased in expression from day 10 to day 30 after tumor induction as compared with other subsets, whereas M1 TAMs expressed significantly higher integrin α5 as compared with different subsets.

In the TME, macrophages undergo a phenotypic shift from proinflammatory (M1) to anti-inflammatory (M2), facilitating tumor growth. CD206, a mannose receptor, and MERTK signaling contribute significantly to establishing an immunosuppressive milieu crucial for tumor progression (35, 36). Therefore, CD206hi/low and MERTKhi/low cells in M1 and M2 TAMs in the tumor and lungs at days 15 and 30 were analyzed, and the expression of integrins α5 and αv in CD206 and MERTK expressed cells were checked. Distinct populations were gated based on the expression of CD206 and MERTK on Ly6ClowMHCIIlow (M1) and Ly6ClowMHCIIhi (M2) TAMs (Fig. 5A, 5B, 5I, 5J). In the tumor and lungs, M2 TAMs expressed significantly higher CD206 and MERTK than M1 TAMs on days 15 and 30, which correlates with the cell number (Fig. 5C, 5D, 5K, 5L) and frequency of the cells (Fig. 5A, 5B, 5I, 5J). The cell number of the CD206hi cells in the M2 TAMs was higher than CD206low cells on both day 15 and day 30, and there was a significant increase in the number of CD206hi and MERTKhi cells in M2 TAMs compared with M1 TAMs in both tumors and lungs (Fig. 5C, 5D, 5K, 5L).

FIGURE 5.

TAM subsets (M1 and M2) in tumor and lungs showed differential integrin α5 and αv expression in CD206hi/low and MERTKhi/low cells. (A and B) Dot plots showing frequency and distribution of M1 TAMs (Ly6ClowMHCIIhigh) and M2 TAMs (Ly6ClowMHCIIlow) in CD11b+Ly6GF4/80+ cells in the tumors at days 15 and 30 concerning their (A) CD206 and (B) MERTK expression. (C and D) Bar diagrams represent the cell number of (C) CD206hi/low and (D) MERTKhi/low in M1 and M2 TAMs in tumor at days 15 and 30. (EH) Line diagrams represent the MFI of (E) integrin α5 and (F) integrin αv in M1 and M2 TAMs in CD206hi/low cells and (G) integrin α5 and (H) integrin αv in M1 and M2 TAMs in MERTKhi/low in tumor at days 15 and 30. (IL) Dot plots showing frequency and distribution of M1 TAMs and M2 TAMs in the lung of tumor-bearing mice at days 15 and 30 with respect to their (I) CD206 and (J) MERTK expression. Bar diagrams represent the cell number of (K) CD206hi/low and (L) MERTKhi/low in M1 and M2 TAMs in lungs at days 15 and 30. (MP) Line diagrams represent the MFI of (M) integrin α5 and (N) integrin αv in M1 and M2 TAMs in CD206hi/low cells and the MFI of (O) integrin α5 and (P) integrin αv in M1 and M2 TAMs in MERTKhi/low cells in lungs at days 15 and 30. All values are presented as mean ± SEM (n = 4–6 per group). *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

FIGURE 5.

TAM subsets (M1 and M2) in tumor and lungs showed differential integrin α5 and αv expression in CD206hi/low and MERTKhi/low cells. (A and B) Dot plots showing frequency and distribution of M1 TAMs (Ly6ClowMHCIIhigh) and M2 TAMs (Ly6ClowMHCIIlow) in CD11b+Ly6GF4/80+ cells in the tumors at days 15 and 30 concerning their (A) CD206 and (B) MERTK expression. (C and D) Bar diagrams represent the cell number of (C) CD206hi/low and (D) MERTKhi/low in M1 and M2 TAMs in tumor at days 15 and 30. (EH) Line diagrams represent the MFI of (E) integrin α5 and (F) integrin αv in M1 and M2 TAMs in CD206hi/low cells and (G) integrin α5 and (H) integrin αv in M1 and M2 TAMs in MERTKhi/low in tumor at days 15 and 30. (IL) Dot plots showing frequency and distribution of M1 TAMs and M2 TAMs in the lung of tumor-bearing mice at days 15 and 30 with respect to their (I) CD206 and (J) MERTK expression. Bar diagrams represent the cell number of (K) CD206hi/low and (L) MERTKhi/low in M1 and M2 TAMs in lungs at days 15 and 30. (MP) Line diagrams represent the MFI of (M) integrin α5 and (N) integrin αv in M1 and M2 TAMs in CD206hi/low cells and the MFI of (O) integrin α5 and (P) integrin αv in M1 and M2 TAMs in MERTKhi/low cells in lungs at days 15 and 30. All values are presented as mean ± SEM (n = 4–6 per group). *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

Close modal

In the previous results, as the prominent expression of integrins α5 and αv in the different TAM subsets was observed, we hypothesized that these integrins might have been involved in the reprogramming of TAMs. Therefore, we analyzed the expression of integrins α5 and αv in CD206- and MERTK-expressing cells. As shown in Fig. 5E–H, integrin α5 and αv expression levels in the CD206hi/low and MERTKhi/low cells were notably elevated in M2 TAMs compared with M1 TAMs at both day 15 and 30, suggesting that these adhesion receptors might be involved in TAM reprogramming. Specifically, CD206hi and MERTKhi cells within M2 TAMs showed increased expression of α5 and αv compared with CD206low and MERTKlow, respectively, on days 15 and 30. However, no significant difference in α5 expression was observed in the M1 TAMs.

Similarly, in the lungs, M2 TAMs expressing CD206hi have higher integrin α5 expression than do CD206low cells, and there were no significant differences in CD206hi and CD206low in M1 TAMs (Fig. 5M), whereas integrin αv in CD206hi cells expressed M1 and M2 TAMs was significantly upregulated as compared with the CD206low cells both at day 15 and 30 (Fig. 5N). M2 TAMs expressing MERTKhi have higher α5 and αv expression than do MERTKlow cells (Fig. 5O, 5P). Also, the expression levels of integrins α5 and αv in both CD206hi/low and MERTKhi/low cells were significantly increased in M2 TAMs as compared with M1 TAMs (Fig. 5M–P). With CD206 and MERTK being essential markers of TAM polarization, these findings suggest the possibility of the involvement of integrin αv as well as integrin α5 in the reprogramming of M2 TAMs.

In subsequent experiments, we investigated the functional role of integrins α5 and αv in TAM reprogramming at the transcriptional level. Macrophage subsets expressing these integrins were sorted from day 15 tumor tissues based on FVS450CD11b+Ly6GF4/80+ expression. Three macrophage populations were identified based on integrin expression: α5+, αv+, and α5+αv+ (Fig. 6A). The frequency and purity of different sorted populations are shown in Fig. 6B. Interestingly, α5+ macrophages showed enhanced levels of inflammatory cytokines such as Tnfa and Il6 as compared with αv+ and α5+αv+ cells transcriptionally (Fig. 6C, 6D). However, αv+ macrophages showed an elevated level of M2 markers Cd163 and Cd206 at the transcriptional level compared with other macrophage populations (α5+ and α5+αv+) (Fig. 6E, 6F). Similarly, αv+ macrophages also showed increased levels of protumorigenic genes such as Il10, Arg1, Tgfb, and metastasis and angiogenic marker Mmp9 transcriptionally (Fig. 6G–J).

FIGURE 6.

Transcriptional expression profile of integrins expressing CD11b+F4/80+ macrophages in the tumor. (A) Flow cytometry plots represent the gating strategy used to sort α5+, αv+, and α5+αv+ cells from FVS450CD11b+Ly6GF4/80+ macrophages in tumors at day 15 postinjection. (B) Flow cytometry plots represent the frequency and purity of sorted cells (α5+, αv+, and α5+αv+). (CO) Bar diagrams represents the expression kinetics of (C) Tnfa, (D) Il6, (E) Cd206, (F) Cd163, (G) Il10, (H) Arg1, (I) Tgfb, (J) Mmp9, (K) Stat1, (L) Stat2, (M) Stat3, (N) Stat6, and (O) Pparg of sorted cells (Ly6Chi, α5+, αv+, and α5+αv+ cells). Fold changes were calculated with respect to the Ly6Chi monocytes. Isolated cells were pooled from four mice into two sets (n = 4). (P) Line diagram showing the serum concentration (pg/ml) of IL-6 and IL-10 in the blood of naive and 4T1-induced tumor-bearing mice at different time points using sandwich ELISA. Plasma was collected from four animals performed in technical duplicates; n = 8. All values are presented as mean ± SEM. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

FIGURE 6.

Transcriptional expression profile of integrins expressing CD11b+F4/80+ macrophages in the tumor. (A) Flow cytometry plots represent the gating strategy used to sort α5+, αv+, and α5+αv+ cells from FVS450CD11b+Ly6GF4/80+ macrophages in tumors at day 15 postinjection. (B) Flow cytometry plots represent the frequency and purity of sorted cells (α5+, αv+, and α5+αv+). (CO) Bar diagrams represents the expression kinetics of (C) Tnfa, (D) Il6, (E) Cd206, (F) Cd163, (G) Il10, (H) Arg1, (I) Tgfb, (J) Mmp9, (K) Stat1, (L) Stat2, (M) Stat3, (N) Stat6, and (O) Pparg of sorted cells (Ly6Chi, α5+, αv+, and α5+αv+ cells). Fold changes were calculated with respect to the Ly6Chi monocytes. Isolated cells were pooled from four mice into two sets (n = 4). (P) Line diagram showing the serum concentration (pg/ml) of IL-6 and IL-10 in the blood of naive and 4T1-induced tumor-bearing mice at different time points using sandwich ELISA. Plasma was collected from four animals performed in technical duplicates; n = 8. All values are presented as mean ± SEM. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

Close modal

M1 macrophage polarization is driven by STAT1 and STAT2, which enhance inflammatory cytokine production and immune responses against tumors (11). Conversely, STAT3 and STAT6 influence M2 macrophage polarization, fostering an immunosuppressive environment that promotes tumor growth and metastasis (37, 38). Our results showed that there was a significant increase in Stat1 and Stat2 levels in the α5+ macrophages, whereas αv+ macrophages have significantly increased Stat3 and Stat6 expression in transcript level as compared with control Ly6Chi cells (Fig. 6K–N). Additionally, integrins αvβ5 and β3 depended on peroxisome proliferator-activated receptor-γ (PPARγ) signaling to regulate M2 polarization in breast tumor, further supporting a tumor-promoting microenvironment (39, 40). The Pparg gene expression was higher in αv+ macrophages as compared with α5+ macrophages (Fig. 6O). This interaction between α5 and αv integrins and tumor extracellular matrix modulates these signaling pathways, influencing TAM polarization and tumor progression. Our study indicates that αv+ integrins may play a prominent role in the differentiation and polarization of M2 TAMs and help in tumor progression.

Inflammatory macrophages generate mediators such as IL-6 and IL-10, fostering a mutagenic environment. As tumors evolve, macrophages transition into M2-like TAMs, further promoting tumor growth while dampening immune responses (41). In our study, IL-6 and IL-10 levels were significantly higher in serum from tumor-bearing mice from day 3 to day 30 compared with controls (Fig. 6P). Elevated IL-6 suggests enhanced cancer cell survival, whereas increased IL-10 indicates a regulatory effect on the immune response, potentially suppressing antitumor reactions.

To check the function of α5 and αv integrins in macrophage polarization, human peripheral blood monocytes were cultured in MDA-MB-231 TS in the presence or absence of integrin α5 and αv blocking Abs. By day 7, M-CSF– and TS-treated monocytes differentiated into macrophages, displaying elongated morphology compared with controls, which was notably reduced upon integrin α5 and αv blockade (Fig. 7A). Quantitative analysis confirmed that integrin blocking significantly decreased cell spreading (Fig. 7B) and elongation (Fig. 7C), highlighting the importance of α5 and αv in TAM differentiation. Furthermore, gene expression analysis revealed that blocking integrins α5 and αv led to reduced expression of M2 markers (Cd206, Cd163) and protumorigenic genes (Arg1, Vegf) in the differentiated TAMs compared with TS control and M-CSF on day 7 (Fig. 7D–G). This indicates that integrins α5 and αv are crucial for promoting M2-type polarization and the expression of genes associated with tumor progression. Additionally, there was no significant impact on the expression of M1 markers (Cd80, Inos2) with integrin blocking, suggesting the role of α5 and αv integrin specificity toward M2 polarization (Fig. 7H, 7I).

FIGURE 7.

Blocking of integrins α5 and αv inhibits polarization of monocyte-derived macrophages and RAW 264.7 cells in vitro. (A) Images of differentiated macrophages at day 7 after treatment with RPMI 1640 medium, M-CSF (25 ng/ml), MDA-MB-231 TS, and isotype control, α5 and αv blocking Abs taken at ×40 objective. Scale bars, 50 μm. (B and C) Quantification of (B) cell area (μm2) and (C) cell axis length (μm) of differentiated macrophages at day 7 analyzed using ImageJ software. n = 50 cells/group counted from four separate fields of microscopic images taken at ×40 objective. (DI) Bar diagrams represents the expression kinetics (mRNA fold change) of (D) Cd206, (E) Cd163, (F) Arg1, (G) Vegf, (H) Cd80, and (I) Inos2 in macrophages polarized in M-CSF or MDA-MB-231 TS and with anti-α5 or anti-αv Abs. (JM) Bar diagrams represents the MFI of (J) MHCII, (K) CD86, (L) CD206, and (M) CD163 in human peripheral blood monocyte-derived macrophages differentiated using M-CSF or MDA-MB-231 TS with or without FAK inhibition (PF-573228) on days 3 and 7 after seeding. (NP) Bar diagrams represents the MFI of (N) MHCII, (O) CD206, and (P) MERTK in RAW 264.7 cells treated with either media or IFN-γ and LPS or IL-4 and IL-13 or 4T1 TS with or without FAK inhibition (PF-573228) for 24 and 48 h. Data represent three independent experiments performed in duplicates; n = 6. All values are represented as mean ± SEM. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

FIGURE 7.

Blocking of integrins α5 and αv inhibits polarization of monocyte-derived macrophages and RAW 264.7 cells in vitro. (A) Images of differentiated macrophages at day 7 after treatment with RPMI 1640 medium, M-CSF (25 ng/ml), MDA-MB-231 TS, and isotype control, α5 and αv blocking Abs taken at ×40 objective. Scale bars, 50 μm. (B and C) Quantification of (B) cell area (μm2) and (C) cell axis length (μm) of differentiated macrophages at day 7 analyzed using ImageJ software. n = 50 cells/group counted from four separate fields of microscopic images taken at ×40 objective. (DI) Bar diagrams represents the expression kinetics (mRNA fold change) of (D) Cd206, (E) Cd163, (F) Arg1, (G) Vegf, (H) Cd80, and (I) Inos2 in macrophages polarized in M-CSF or MDA-MB-231 TS and with anti-α5 or anti-αv Abs. (JM) Bar diagrams represents the MFI of (J) MHCII, (K) CD86, (L) CD206, and (M) CD163 in human peripheral blood monocyte-derived macrophages differentiated using M-CSF or MDA-MB-231 TS with or without FAK inhibition (PF-573228) on days 3 and 7 after seeding. (NP) Bar diagrams represents the MFI of (N) MHCII, (O) CD206, and (P) MERTK in RAW 264.7 cells treated with either media or IFN-γ and LPS or IL-4 and IL-13 or 4T1 TS with or without FAK inhibition (PF-573228) for 24 and 48 h. Data represent three independent experiments performed in duplicates; n = 6. All values are represented as mean ± SEM. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

Close modal

FAK is a cytoplasmic tyrosine kinase crucial for intracellular signaling by integrins, influencing adhesion, migration, survival, growth, and differentiation (42, 43). To study the impact of FAK inhibition on macrophage polarization, human peripheral blood monocytes were treated with the FAK inhibitor PF-573228 and cultured with M-CSF and MDA-MB-231 TS for 7 d. Results showed a significant decrease in both M1 (MHC II, CD86) and M2 (CD206, CD163) markers in M-CSF– and TS-induced macrophages after FAK inhibition compared with cells without FAK inhibition (Fig. 7J–M). Similarly, in RAW 264.7 cells, IFN-γ and LPS stimulation induced M1 polarization evidenced by increased expression of MHCII (Fig. 7N), whereas increased expression of CD206 and MERTK was observed in IL-4– and IL-13–stimulated cells (Fig. 7O, 7P). FAK inhibition significantly reduced MHCII expression, impairing M1 polarization, and also reduced CD206 and MERTK expression, impacting M2 polarization. Additionally, treatment with 4T1-conditioned medium effectively polarized RAW 264.7 cells into M2 macrophages, as indicated by elevated CD206 and MERTK and lowered MHC II levels. However, the presence of the FAK inhibitor notably decreased these markers, demonstrating a disruption in M2 macrophage differentiation (Fig. 7O, 7P). These findings highlight FAK’s role in macrophage polarization and the effects of its inhibition.

During tumorigenesis, tumor-infiltrating myeloid cells can support cancer cell survival, proliferation, and migration, partly by influencing ECM remodeling (44). To that note, the expression levels of ECM proteins such as fibronectin, vitronectin, fibrinogen, collagen IV, and vimentin were checked in tumor tissues (days 10, 15, 23, and day 30) and mammary gland tissues (tissues from the site of injection on days 3 and 7) and compared them with mammary gland tissue from naive mice as a control. Results showed increased fibronectin, vitronectin, and collagen IV expression in tumors compared with naive control, fibrinogen, and vimentin as the tumor progressed (Fig. 8A). We further explored the impact of ECM proteins on TAM polarization using M-CSF and MDA-MB-231 TS. Fibronectin and vitronectin enhanced the expression of integrins α and αv in TAMs, indicating their regulatory role in TAM polarization through integrins (Fig. 7B, 7C). Blocking integrins α5 and αv in monocytes cultured with MDA-MB-231 TS and ECM proteins reduced M2 markers (CD206 and CD163) but did not significantly affect M1 markers (MHCII and CD86), except for integrin αv, which lowered CD86 and MHCII in vitronectin-coated wells (Fig. 8D–G). This suggests that fibronectin and vitronectin, through integrins, primarily influence M2 TAM polarization.

FIGURE 8.

Blocking integrins α5 and αv in the presence of ECM proteins inhibits differentiation and polarization of monocytes into TAMs. (A) Line diagram represents the expression kinetics (mRNA fold change) of ECM proteins (fibronectin, vitronectin, fibrinogen, collagen-iv, and vimentin) in tumor tissues at different days (days 3, 7, 10, 15, 23, and 30) after induction of the tumor. All mRNA fold changes were calculated with respect to naive mice (mammary gland tissues). Samples were collected from three to four mice per group. (B and C) Bar diagrams represents the expression kinetics (mRNA fold change) of integrins α5 and αv in M-CSF and MDA-MB-231 TS-polarized macrophages on the surface of different ECM proteins (fibronectin, fibrinogen, vitronectin, and vimentin) at day 7 after seeding. (DG) Bar graphs represents the MFI of (D) CD206, (E) CD163, (F) CD86, and (G) MHCII in macrophages polarized in MDA-MB-231 TS with anti-α5 or anti-αv Abs on the surface of fibronectin and vitronectin proteins at day 7 after seeding. All data represent three independent experiments performed in duplicates; n = 6. All mRNA fold changes were calculated with respect to control cells cultured in only media. All values are represented as mean ± SEM. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

FIGURE 8.

Blocking integrins α5 and αv in the presence of ECM proteins inhibits differentiation and polarization of monocytes into TAMs. (A) Line diagram represents the expression kinetics (mRNA fold change) of ECM proteins (fibronectin, vitronectin, fibrinogen, collagen-iv, and vimentin) in tumor tissues at different days (days 3, 7, 10, 15, 23, and 30) after induction of the tumor. All mRNA fold changes were calculated with respect to naive mice (mammary gland tissues). Samples were collected from three to four mice per group. (B and C) Bar diagrams represents the expression kinetics (mRNA fold change) of integrins α5 and αv in M-CSF and MDA-MB-231 TS-polarized macrophages on the surface of different ECM proteins (fibronectin, fibrinogen, vitronectin, and vimentin) at day 7 after seeding. (DG) Bar graphs represents the MFI of (D) CD206, (E) CD163, (F) CD86, and (G) MHCII in macrophages polarized in MDA-MB-231 TS with anti-α5 or anti-αv Abs on the surface of fibronectin and vitronectin proteins at day 7 after seeding. All data represent three independent experiments performed in duplicates; n = 6. All mRNA fold changes were calculated with respect to control cells cultured in only media. All values are represented as mean ± SEM. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

Close modal

The involvement of integrins α5 and αv has emerged as a focal point of investigation due to their known roles in cell adhesion, migration, and TAM reprogramming. RNA sequencing analysis from the 4T1 murine breast tumor model (GEO dataset: GSE195857) compared TAMs to MGMs and BMDM-Hs to BMDM-Ts. This revealed 4148 DEGs for TAMs versus MGMs and 401 for BMDM-Ts versus BMDM-Hs, with significant pathways including the “immune system” (Reactome ID: R-MMU-168256). Notable integrins such as Itgam (CD11b) and Itgax (CD11c) were significantly upregulated in both macrophages and monocytes (Fig. 9A, 9B). Additionally, downregulation of integrin α5 suggests reduced proinflammatory pathways, whereas upregulation of integrin αv points to a shift toward a protumorigenic and immunosuppressive phenotype.

FIGURE 9.

Differential enriched gene analysis and protein–protein interaction network analysis of RNA sequencing data from the GEO dataset (GSE195857). (A and B) Log2 fold changes of differentially expressed integrins are shown in the categories (A) bone marrow–derived monocytes from healthy and tumor-bearing mice (BMDM-Hs versus BMDM-Ts) and (B) mammary gland macrophages from healthy mice versus tumor-associated macrophages from tumor-bearing mice (MGMs versus TAMs), with p value ≤0.05. Protein–protein interaction (PPI) analyses were conducted using the STRING analysis tool. (C and D) The STRING PPI network compares the interactions of integrins (C) α5 (Itga5) and (D) αv (Itgav) with the DEGs of the top functional pathways of BMDM-Hs versus BMDM-Ts categories with an interaction score of 0.400 (medium confidence) that are shown in the network. (E and F) The STRING PPI network compares the interaction of integrins (E) α5 (Itga5) and (F) αv (Itgav) with the DEGs of the top functional pathways in TAMs versus MGMs categories. Only connected nodes and interactions with high confidence (0.7) are shown in the network. The line thickness indicates the strength of data support for each interaction. (G–I) Bar diagrams represents the MFI of phosphorylated (G) SRC, (H) STAT1, and (I) STAT6 in human BMDMs differentiated using M-CSF or MDA-MB-231 TS with or without FAK inhibition (PF-573228) on days 3 and 7 after seeding. DMSO-treated cells were taken as negative solvent control. Data represent three independent experiments performed in duplicates; n = 6. (JL) Bar diagrams represents the MFI of phosphorylated (J) SRC, (K) STAT1, and (L) STAT6 in RAW 264.7 cells treated with either media or IFN-γ combined with LPS or IL-4 combined with IL-13 or 4T1 TS with or without FAK inhibition (PF-573228) after 24 and 48 h. Data represent three independent experiments performed in duplicates; n = 6. All values are represented as mean ± SEM. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

FIGURE 9.

Differential enriched gene analysis and protein–protein interaction network analysis of RNA sequencing data from the GEO dataset (GSE195857). (A and B) Log2 fold changes of differentially expressed integrins are shown in the categories (A) bone marrow–derived monocytes from healthy and tumor-bearing mice (BMDM-Hs versus BMDM-Ts) and (B) mammary gland macrophages from healthy mice versus tumor-associated macrophages from tumor-bearing mice (MGMs versus TAMs), with p value ≤0.05. Protein–protein interaction (PPI) analyses were conducted using the STRING analysis tool. (C and D) The STRING PPI network compares the interactions of integrins (C) α5 (Itga5) and (D) αv (Itgav) with the DEGs of the top functional pathways of BMDM-Hs versus BMDM-Ts categories with an interaction score of 0.400 (medium confidence) that are shown in the network. (E and F) The STRING PPI network compares the interaction of integrins (E) α5 (Itga5) and (F) αv (Itgav) with the DEGs of the top functional pathways in TAMs versus MGMs categories. Only connected nodes and interactions with high confidence (0.7) are shown in the network. The line thickness indicates the strength of data support for each interaction. (G–I) Bar diagrams represents the MFI of phosphorylated (G) SRC, (H) STAT1, and (I) STAT6 in human BMDMs differentiated using M-CSF or MDA-MB-231 TS with or without FAK inhibition (PF-573228) on days 3 and 7 after seeding. DMSO-treated cells were taken as negative solvent control. Data represent three independent experiments performed in duplicates; n = 6. (JL) Bar diagrams represents the MFI of phosphorylated (J) SRC, (K) STAT1, and (L) STAT6 in RAW 264.7 cells treated with either media or IFN-γ combined with LPS or IL-4 combined with IL-13 or 4T1 TS with or without FAK inhibition (PF-573228) after 24 and 48 h. Data represent three independent experiments performed in duplicates; n = 6. All values are represented as mean ± SEM. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001.

Close modal

Furthermore, the correlation of DE genes in the immune system pathway with the integrins α5 and αv was checked with the STRING database. In the BMDM-Ts versus BMDM-Hs comparison, integrins α5 and αv were found to strongly interact with VCAM-1 and NCAM-1 (neural cell adhesion molecule-1), which further associate with MMP9 and STAT3 (Fig. 9C, 9D). VCAM-1 and NCAM-1 facilitate cell adhesion and motility, crucial for inflammatory responses. MMP-9 promotes ECM remodeling, aiding cell invasion and metastasis. Activation of STAT3 in monocytes fosters their differentiation into immunosuppressive TAMs, which also facilitate the secretion of various protumorigenic factors, including cytokines (Il-10ra, Il-13ra1, and Il-4ra), promoting tumor progression and immune evasion. In the TAMs versus MGMs category (Fig. 9E, 9F), many DEGs such as FAK, also known as Ptk2 (protein tyrosine kinase 2) proto-oncogene protein tyrosine kinases (Src, Crk, Lyn), vasodilator-stimulated phosphoprotein (Vasp), and several integrins subunits (Itgal, Itgb5, Itga4, Itgb7) show strong association with both α5 and αv integrins. FAK and SRC kinases regulate cell adhesion, migration, and survival, whereas VASP enhances motility and invasion by modulating actin dynamics, and integrin subunits mediate cell–ECM interactions. This network highlights the central roles of α5 and αv integrins in cancer metastasis, emphasizing their potential as therapeutic targets in cancer treatment.

To explore how FAK inhibition affects macrophage polarization signaling, human peripheral blood monocytes were treated with the FAK inhibitor PF-573228. This treatment significantly reduced the phosphorylation of SRC, STAT1, and STAT6 in macrophages differentiated with M-CSF and MDA-MB-231 TS compared with untreated controls (Fig. 9G–I). In RAW 264.7 cells, FAK inhibition led to decreased expression of integrins αv and α5 in both M1 macrophages (induced by IFN-γ and LPS) and M2 macrophages (induced by IL-4 and IL-13), as well as in cells treated with 4T1 TS (data not shown). Further analysis showed that FAK inhibition reduced SRC, STAT1, and STAT6 phosphorylation in RAW 264.7 cells under various treatments (Fig. 9J–L), highlighting the importance of FAK-mediated integrin signaling in macrophage polarization. The reduced integrin expression impairs the activation of key signaling pathways necessary for macrophage polarization.

The balance between the M1 and M2 polarization states of TAM can influence the fate of tumors (6, 45). Integrins play a crucial role in orchestrating the infiltration of myeloid cells into the TME and are known to influence the behavior of TAMs and their communication with cancer cells (46, 47). Despite the abundance of literature on inflammatory molecules involved in tumorigenesis, the series of events that occur before the appearance of palpable tumors, driving immune cell recruitment and polarization of TAMs in the dynamic TME, have still remained unknown. Thus, our study performed an in-depth analysis of the molecular landscape governing monocyte and macrophage trafficking from the bone marrow to the systemic circulation and tumor tissue, including recruitment and polarization of TAMs in possible metastatic visceral organs, lungs, and a site close to the tumor development, that is, peritoneum. Combined with reprogramming and polarization, immune cell recruitment mechanisms will provide essential insights into the TME and potential targets for therapeutic approaches.

A previous comprehensive study on the macrophage phenotypes in breast and lung carcinoma suggested that Ly6Chi subsets were the most prominent population, serving as a precursor for all distinct TAM subsets. According to that study, these inflammatory monocytes differentiate rapidly into TAMs (34). Using the 4T1 murine model of breast tumors, our timeline study indicated an increase in the frequencies of Ly6Chi monocytes in the bone marrow, blood, and peritoneum from day 3 to day 30 after tumor induction. Their frequencies declined in the tumor tissues from day 15 to day 30 after differentiation into TAMs, where the Ly6Chi cells changed into Ly6Clow cells and polarized into different subpopulations of M1 (Ly6ClowMHCIIhi) and M2 (Ly6ClowMHCIIlow) TAMs. Our data suggest that the CCL2/CCR2 signaling axis facilitates monocyte infiltration from bone marrow to tumor tissues.

Integrins facilitate myeloid cell adhesion to the ECM and endothelial cells, aiding TAM recruitment within the tumor stroma and enhancing infiltration into the tumor parenchyma (48, 49). Our results show that integrin α5β1 was highly expressed on the Ly6Chi monocytes within the bone marrow/blood during early tumor progression. These cells migrated to the tumors and surrounding tissues, peaking at day 15 before their decrease in bone marrow. This suggests that integrin α5 mediates the early events of monocyte recruitment to tumors. These monocytes further differentiated into F480+ macrophages and expressed higher levels of integrin αvβ3. We found that both integrins α5β1 and αvβ3 increased in their expression with the progression of the tumors, thereby underscoring their function in monocyte differentiation and TAM polarization. We further observed a gradual increase in M2-type TAMs as tumors advanced, with Ly6Chi monocytes continuously infiltrating into the tumors. In line with this observation, previous studies have shown that the Ly6ClowMHCIIlow subset of TAMs was the primary population associated with tumor growth, invasion, and metastasis in mammary adenocarcinoma (50).

Furthermore, using multicolor flow cytometry studies, we identified six TAM subpopulations based on Ly6C and MHCII expression, each showing unique integrin α5 and αv patterns. Integrin α5 was upregulated from day 10 to 30 after tumor induction in the M1, M2, and intermediate populations (Ly6ChiMHCIIhi and Ly6CintMHCIIhi). It was highly expressed in the M1 TAMs when compared with the M2 TAMs. In contrast to M1 TAMs, integrin αv was highly expressed in M2 and upregulated integrin α5 at a later time point. In contrast, in the intermediate populations, the expression of integrins α5 and αv increased until day 15 but decreased thereafter. These findings suggest that integrins α5 and αv both may be involved in M2 TAM polarization in the tumor microenvironment. Our findings are consistent with a prior study by Shu et al. (39) that illustrates how integrin β3 via PPARγ is essential for the polarization of M2 TAMs in a 4T1-induced breast tumor model and that blockade of integrin β3 suppressed macrophage M2 polarization. However, in this study, the authors analyzed the entire F4/80-expressing cells and not the cellular subsets. A more detailed analysis of M1 and M2 TAMs, based on the surface expression of anti-inflammatory markers, such as CD206 and MERTK, in tumors and lungs, revealed that the expression of integrin αv was higher in both CD206hi and MERTKhi M2 TAMs compared with M1 TAMs in both tumor and lung tissues.

In our studies, we have further validated the role of integrins in TAM polarization by performing detailed pro- and antitumorigenic gene profiling in TAMs expressing specific integrins that were sorted from 4T1-induced murine breast tumors. Our data showed that CD11b+F4/80+αv+ TAMs showed higher M2 markers (Cd206, Cd163) and protumorigenic genes (Tgfb, Arg1), whereas CD11b+F4/80+α5+ cells demonstrated an increase in inflammatory cytokine genes (Tnfa, Il6). This correlated with higher expression of stat1 and stat2 in α5+ cells and Stat3, Stat6, and Pparg signaling molecules in αv+ cells at transcript levels.

In our functional in vitro assays, blocking of integrins αv and α5 disrupted TAM differentiation and polarization, as shown from the lower expression levels of M2 polarization markers such as CD206 and CD163, which again supported the role of integrin αv and α5 in different stages of TAM polarization. In support of these findings, we had previously demonstrated that a C-terminal fragment of adhesion protein Fibulin7 (Fbln7-C) binds to monocyte integrin α5β1 and negatively regulates monocyte and macrophage migration, differentiation, and polarization via promoting STAT and ERK1/2, signaling. Administration of Fbln7-C also reduced the growth of 4T1-induced murine tumors and delayed the programming of TAMs in murine tumors (20). Similarly, in murine polymicrobial sepsis and LPS-induced endotoxemia models, integrin α5 and integrin αv demonstrated a robust correlation with the inflammatory and anti-inflammatory phenotype of macrophages, respectively (19). In this study, while F4/80+CD206hiαvhi macrophages showed higher expression of TGFBR1, the F4/80+α5hi macrophages displayed increased p-SRC expression levels at 12 h after induction (19).

Interaction of surface integrins with ECM proteins present in the TME can play an essential role in cell adhesion and migration, proliferation, survival, and metastasis (16, 51). Integrins recognize multiple matrix ligands, such as fibronectin, vitronectin, collagen, and laminin, as well as cell surface receptors, such as ICAM-1 or VCAM-1 (51, 52). In our study, the expression of fibronectin and vitronectin in tumor tissues was upregulated at the transcript level and blocked integrin–ECM interactions, especially with fibronectin and vitronectin, which inhibited M2 TAM polarization. This may imply that interactions of integrins with the specific ECM proteins in the TME can play an essential role in TAM polarization and reprogramming.

The bioinformatics analysis of RNA sequencing data of BMDCs and TAMs acquired from a GEO dataset (GSE195857) was performed. Protein–protein interaction analysis via STRING revealed that SRC family kinase and FAK are key signaling molecules associated with integrins α5 and αv. Studies have shown that SRC and FAK can regulate TAM adhesion, migration, and polarization through downstream PI3K/AKT, Ras/MAPK, or STAT3 pathways (49, 53). FAK phosphorylates several downstream signaling molecules, which influence cellular responses such as migration, survival, and differentiation (54). Our results from the experiments with FAK blocking demonstrate that inhibition of FAK can significantly disrupt the polarization macrophages under tumor microenvironment-like conditions. This was confirmed in TAMs generated in vitro using RAW 264.7 cell lines and human PBMC-derived macrophages. A critical role of FAK in macrophage polarization in the above models was demonstrated by decreased MHCII expression in M1 macrophages and reduced CD206, CD163, and MERTK in M2 macrophages and an impaired expression of p-SRC, STAT1, and STAT6 under FAK blocking condition. Targeting these pathways could modulate TAM behavior and immune response in the TME.

In summary, our data demonstrate the role of integrins α5 and αv in regulating macrophage infiltration, differentiation, and polarization in breast cancer models. Understanding these mechanisms could aid in comprehending immunoediting, cancer progression, and developing TAM-based therapies.

The authors have no financial conflicts of interest.

We gratefully acknowledge Dr. Prerna Sharma’s valuable contributions in critically reviewing the manuscript.

This work was supported by Department of Biotechnology, Ministry of Science and Technology, India Grant BT/010/IYBA/2017/04 (to P.P.S.); CSIR Human Resource Development Group Grant 09/143(0919)/2018-EMR-I; a CSIR-GOI fellowship to N.D.; a DBT-GOI fellowship to S.K.R.; a UGC-GOI fellowship to S.P.D.; and an MHRD-GOI fellowship to P.K. and D.S.

The online version of this article contains supplemental material.

BMDM

bone marrow–derived monocyte

BMDM-H

BMDM from healthy mouse

BMDM-T

BMDM from tumor-bearing mouse

DEG

differentially expressed gene

ECM

extracellular matrix

FAK

focal adhesion kinase

FVS450

fixable viability stain 450

MERTK

MER proto-oncogene, tyrosine kinase

MFI

mean fluorescence intensity

MGM

mammary gland macrophage

MHCII

MHC class II

qPCR

quantitative PCR

SRC

proto-oncogene tyrosine-protein kinase c-Src

TAM

tumor-associated macrophage

TME

tumor microenvironment

TS

tumor supernatant

1
Solinas
,
G.
,
G.
Germano
,
A.
Mantovani
,
P.
Allavena
.
2009
.
Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation
.
J. Leukoc. Biol.
86
:
1065
1073
.
2
Ngambenjawong
,
C.
,
H. H.
Gustafson
,
S. H.
Pun
.
2017
.
Progress in tumor-associated macrophage (TAM)-targeted therapeutics
.
Adv. Drug Deliv. Rev.
114
:
206
221
.
3
Albini
,
A.
,
A.
Bruno
,
D. M.
Noonan
,
L.
Mortara
.
2018
.
Contribution to tumor angiogenesis from innate immune cells within the tumor microenvironment: implications for immunotherapy
.
Front. Immunol.
9
:
527
.
4
Kim
,
J.
,
J.-S.
Bae
.
2016
.
Tumor-associated macrophages and neutrophils in tumor microenvironment
.
Mediators Inflamm.
2016
:
6058147
.
5
Hao
,
N.-B.
,
M.-H.
,
Y.-H.
Fan
,
Y.-L.
Cao
,
Z.-R.
Zhang
,
S.-M.
Yang
.
2012
.
Macrophages in tumor microenvironments and the progression of tumors
.
Clin. Dev. Immunol.
2012
:
948098
.
6
Wang
,
J.
,
D.
Li
,
H.
Cang
,
B.
Guo
.
2019
.
Crosstalk between cancer and immune cells: role of tumor‐associated macrophages in the tumor microenvironment
.
Cancer Med.
8
:
4709
4721
.
7
Mortezaee
,
K.
,
J.
Majidpoor
.
2022
.
Roles for macrophage-polarizing interleukins in cancer immunity and immunotherapy
.
Cell. Oncol. (Dordr.)
45
:
333
353
.
8
Shapouri-Moghaddam
,
A.
,
S.
Mohammadian
,
H.
Vazini
,
M.
Taghadosi
,
S.-A.
Esmaeili
,
F.
Mardani
,
B.
Seifi
,
A.
Mohammadi
,
J. T.
Afshari
,
A.
Sahebkar
, et al
.
2018
.
Macrophage plasticity, polarization, and function in health and disease
.
J. Cell. Physiol.
233
:
6425
6440
.
9
Chen
,
S.
,
A. F. U. H.
Saeed
,
Q.
Liu
,
Q.
Jiang
,
H.
Xu
,
G. G.
Xiao
,
L.
Rao
,
Y.
Duo
.
2023
.
Macrophages in immunoregulation and therapeutics
.
Signal Transduct. Target. Ther.
8
:
207
.
10
Xu
,
Y.
,
X.
Wang
,
L.
Liu
,
J.
Wang
,
J.
Wu
,
C.
Sun
.
2022
.
Role of macrophages in tumor progression and therapy
.
Int. J. Oncol.
60
:
57
.
11
Gao
,
J.
,
Y.
Liang
,
L.
Wang
.
2022
.
Shaping polarization of tumor-associated macrophages in cancer immunotherapy
.
Front. Immunol.
13
:
888713
.
12
Mantovani
,
A.
,
F.
Marchesi
,
S.
Jaillon
,
C.
Garlanda
,
P.
Allavena
.
2021
.
Tumor-associated myeloid cells: diversity and therapeutic targeting
.
Cell. Mol. Immunol.
18
:
566
578
.
13
Ham
,
S.
,
L. G.
Lima
,
E.
Lek
,
A.
Möller
.
2020
.
The impact of the cancer microenvironment on macrophage phenotypes
.
Front. Immunol.
11
:
1308
.
14
Conway
,
J. R.
,
G.
Jacquemet
.
2019
.
Cell matrix adhesion in cell migration
.
Essays Biochem.
63
:
535
551
.
15
Karamanos
,
N. K.
,
A. D.
Theocharis
,
Z.
Piperigkou
,
D.
Manou
,
A.
Passi
,
S. S.
Skandalis
,
D. H.
Vynios
,
V.
Orian-Rousseau
,
S.
Ricard-Blum
,
C. E. H.
Schmelzer
, et al
.
2021
.
A guide to the composition and functions of the extracellular matrix
.
FEBS J.
288
:
6850
6912
.
16
Yousefi
,
H.
,
M.
Vatanmakanian
,
M.
Mahdiannasser
,
L.
Mashouri
,
N. V.
Alahari
,
M. R.
Monjezi
,
S.
Ilbeigi
,
S. K.
Alahari
.
2021
.
Understanding the role of integrins in breast cancer invasion, metastasis, angiogenesis, and drug resistance
.
Oncogene
40
:
1043
1063
.
17
Paulynn Chin
,
S
,
D. J.
Louise
,
H. A.
Frances
.
2013
. Integrins in prostate cancer invasion and metastasis. In
Advances in Prostate Cancer.
H.
Gerhard
, ed.
IntechOpen
,
Rijeka, Croatia
. Available at: http://dx.doi.org/10.5772/53482.
18
Liu
,
F.
,
Q.
Wu
,
Z.
Dong
,
K.
Liu
.
2023
.
Integrins in cancer: emerging mechanisms and therapeutic opportunities
.
Pharmacol. Ther.
247
:
108458
.
19
Dash
,
S. P.
,
P.
Chakraborty
,
P. P.
Sarangi
.
2021
.
Inflammatory monocytes and subsets of macrophages with distinct surface phenotype correlate with specific integrin expression profile during murine sepsis
.
J. Immunol.
207
:
2841
2855
.
20
Chakraborty
,
P.
,
S. P.
Dash
,
N.
Dalpati
,
P.
Kumar
,
D.
Jain
,
P. P.
Sarangi
.
2021
.
A C-terminal fragment of adhesion protein fibulin‐7 inhibits growth of murine breast tumor by regulating macrophage reprogramming
.
FEBS J.
288
:
803
817
.
21
Murray
,
P. J.
,
J. E.
Allen
,
S. K.
Biswas
,
E. A.
Fisher
,
D. W.
Gilroy
,
S.
Goerdt
,
S.
Gordon
,
J. A.
Hamilton
,
L. B.
Ivashkiv
,
T.
Lawrence
, et al
.
2014
.
Macrophage activation and polarization: nomenclature and experimental guidelines
.
Immunity
41
:
14
20
.
22
Pireaux
,
V.
,
A.
Sauvage
,
B.
Bihin
,
M.
Van Steenbrugge
,
A.
Rousseau
,
P.
Van Antwerpen
,
K.
Zouaoui Boudjeltia
,
M.
Raes
.
2016
.
Myeloperoxidase‐oxidized LDLs enhance an anti‐inflammatory M2 and antioxidant phenotype in murine macrophages
.
Mediators Inflamm.
2016
:
8249476
.
23
Patro
,
R.
,
G.
Duggal
,
M. I.
Love
,
R. A.
Irizarry
,
C.
Kingsford
.
2017
.
Salmon provides fast and bias-aware quantification of transcript expression
.
Nat. Methods
14
:
417
419
.
24
Love
,
M.
,
S.
Anders
,
W.
Huber
.
2014
.
Differential analysis of count data—the DESeq2 package
.
Genome Biol.
15
:
10
:
1186
.
25
Young
,
M. D.
,
M. J.
Wakefield
,
G. K.
Smyth
,
A.
Oshlack
.
2010
.
Gene ontology analysis for RNA-seq: accounting for selection bias
.
Genome. Biol.
11
:
R14
.
26
Szklarczyk
,
D.
,
R.
Kirsch
,
M.
Koutrouli
,
K.
Nastou
,
F.
Mehryary
,
R.
Hachilif
,
A. L.
Gable
,
T.
Fang
,
N. T.
Doncheva
,
S.
Pyysalo
, et al
.
2023
.
The STRING database in 2023: protein–protein association networks and functional enrichment analyses for any sequenced genome of interest
.
Nucleic Acids Res.
51
:
D638
D646
.
27
Ingersoll
,
M. A.
,
A. M.
Platt
,
S.
Potteaux
,
G. J.
Randolph
.
2011
.
Monocyte trafficking in acute and chronic inflammation
.
Trends Immunol.
32
:
470
477
.
28
Zhang
,
Z.
,
C.
Ni
,
W.
Chen
,
P.
Wu
,
Z.
Wang
,
J.
Yin
,
J.
Huang
,
F.
Qiu
.
2014
.
Expression of CXCR4 and breast cancer prognosis: a systematic review and meta-analysis
.
BMC Cancer
14
:
49
.
29
Yang
,
P.
,
Y.
Hu
,
Q.
Zhou
.
2020
.
The CXCL12-CXCR4 signaling axis plays a key role in cancer metastasis and is a potential target for developing novel therapeutics against metastatic cancer
.
Curr. Med. Chem.
27
:
5543
5561
.
30
Midavaine
,
É.
,
J.
Côté
,
P.
Sarret
.
2021
.
The multifaceted roles of the chemokines CCL2 and CXCL12 in osteophilic metastatic cancers
.
Cancer Metastasis Rev.
40
:
427
445
.
31
Ntanasis-Stathopoulos
,
I.
,
D.
Fotiou
,
E.
Terpos
.
2020
.
CCL3 signaling in the tumor microenvironment
.
Adv. Exp. Med. Biol.
1231
:
13
21
.
32
Zeng
,
Z.
,
T.
Lan
,
Y.
Wei
,
X.
Wei
.
2022
.
CCL5/CCR5 axis in human diseases and related treatments
.
Genes. Dis.
9
:
12
27
.
33
Nourshargh
,
S.
,
R.
Alon
.
2014
.
Leukocyte migration into inflamed tissues
.
Immunity
41
:
694
707
.
34
Movahedi
,
K.
,
D.
Laoui
,
C.
Gysemans
,
M.
Baeten
,
G.
Stangé
,
J.
Van den Bossche
,
M.
Mack
,
D.
Pipeleers
,
P.
In’t Veld
,
P.
De Baetselier
, et al
.
2010
.
Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes
.
Cancer Res.
70
:
5728
5739
.
35
Jaynes
,
J. M.
,
R.
Sable
,
M.
Ronzetti
,
W.
Bautista
,
Z.
Knotts
,
A.
Abisoye-Ogunniyan
,
D.
Li
,
R.
Calvo
,
M.
Dashnyam
,
A.
Singh
, et al
.
2020
.
Mannose receptor (CD206) activation in tumor-associated macrophages enhances adaptive and innate antitumor immune responses
.
Sci. Transl. Med.
12
: eaax6337.
36
Huelse
,
J. M.
,
D. M.
Fridlyand
,
S.
Earp
,
D.
DeRyckere
,
D. K.
Graham
.
2020
.
MERTK in cancer therapy: targeting the receptor tyrosine kinase in tumor cells and the immune system
.
Pharmacol. Ther.
213
:
107577
.
37
Xia
,
T.
,
M.
Zhang
,
W.
Lei
,
R.
Yang
,
S.
Fu
,
Z.
Fan
,
Y.
Yang
,
T.
Zhang
.
2023
.
Advances in the role of STAT3 in macrophage polarization
.
Front. Immunol.
14
:
1160719
.
38
Tugal
,
D.
,
X.
Liao
,
M. K.
Jain
.
2013
.
Transcriptional control of macrophage polarization
.
Arterioscler. Thromb. Vasc. Biol.
33
:
1135
1144
.
39
Shu
,
Y.
,
M.
Qin
,
Y.
Song
,
Q.
Tang
,
Y.
Huang
,
P.
Shen
,
Y.
Lu
.
2020
.
M2 polarization of tumor‐associated macrophages is dependent on integrin β3 via peroxisome proliferator‐activated receptor‐γ up‐regulation in breast cancer
.
Immunology
160
:
345
356
.
40
Yao
,
Q.
,
J.
Liu
,
Z.
Zhang
,
F.
Li
,
C.
Zhang
,
B.
Lai
,
L.
Xiao
,
N.
Wang
.
2018
.
Peroxisome proliferator–activated receptor γ (PPARγ) induces the gene expression of integrin αVβ5 to promote macrophage M2 polarization
.
J. Biol. Chem.
293
:
16572
16582
.
41
Wang
,
H.
,
M. M. H.
Yung
,
H. Y. S.
Ngan
,
K. K. L.
Chan
,
D. W.
Chan
.
2021
.
The impact of the tumor microenvironment on macrophage polarization in cancer metastatic progression
.
Int. J. Mol. Sci.
22
:
6560
.
42
Mierke
,
C. T.
2013
.
The role of focal adhesion kinase in the regulation of cellular mechanical properties
.
Phys. Biol.
10
:
065005
.
43
Tai
,
Y.-L.
,
L.-C.
Chen
,
T.-L.
Shen
.
2015
.
Emerging roles of focal adhesion kinase in cancer
.
Biomed. Res. Int.
2015
:
690690
.
44
Domagala
,
M.
,
C.
Laplagne
,
E.
Leveque
,
C.
Laurent
,
J.-J.
Fournié
,
E.
Espinosa
,
M.
Poupot
.
2021
.
Cancer cells resistance shaping by tumor infiltrating myeloid cells
.
Cancers (Basel)
13
:
165
.
45
Mehta
,
A. K.
,
S.
Kadel
,
M. G.
Townsend
,
M.
Oliwa
,
J. L.
Guerriero
.
2021
.
Macrophage biology and mechanisms of immune suppression in breast cancer
.
Front. Immunol.
12
:
643771
.
46
Harjunpää
,
H.
,
M.
Llort Asens
,
C.
Guenther
,
S. C.
Fagerholm
.
2019
.
Cell adhesion molecules and their roles and regulation in the immune and tumor microenvironment
.
Front. Immunol.
10
:
1078
.
47
Soe
,
Z. Y.
,
E. J.
Park
,
M.
Shimaoka
.
2021
.
Integrin regulation in immunological and cancerous cells and exosomes
.
Int. J. Mol. Sci.
22
:
2193
.
48
Eble
,
J. A.
,
S.
Niland
.
2019
.
The extracellular matrix in tumor progression and metastasis
.
Clin. Exp. Metastasis
36
:
171
198
.
49
Lorusso
,
G.
,
C.
Rüegg
,
F.
Kuonen
.
2020
.
Targeting the extra-cellular matrix—tumor cell crosstalk for anti-cancer therapy: emerging alternatives to integrin inhibitors
.
Front. Oncol.
10
:
1231
.
50
Georgoudaki
,
A.-M.
,
K. E.
Prokopec
,
V. F.
Boura
,
E.
Hellqvist
,
S.
Sohn
,
J.
Östling
,
R.
Dahan
,
R. A.
Harris
,
M.
Rantalainen
,
D.
Klevebring
, et al
.
2016
.
Reprogramming tumor-associated macrophages by antibody targeting inhibits cancer progression and metastasis
.
Cell Rep.
15
:
2000
2011
.
51
Su
,
C.-Y.
,
J.-Q.
Li
,
L.-L.
Zhang
,
H.
Wang
,
F.-H.
Wang
,
Y.-W.
Tao
,
Y.-Q.
Wang
,
Q.-R.
Guo
,
J.-J.
Li
,
Y.
Liu
, et al
.
2020
.
The biological functions and clinical applications of integrins in cancers
.
Front. Pharmacol.
11
:
579068
.
52
Morshed
,
A.
,
A. B.
Abbas
,
J.
Hu
,
H.
Xu
.
2019
.
Shedding new light on the role of ανβ3 and α5β1 integrins in rheumatoid arthritis
.
Molecules
24
:
1537
.
53
Cuesta
,
C.
,
C.
Arévalo-Alameda
,
E.
Castellano
.
2021
.
The importance of being PI3K in the RAS signaling network
.
Genes (Basel)
12
:
1094
.
54
Zhao
,
X.
,
J.-L.
Guan
.
2011
.
Focal adhesion kinase and its signaling pathways in cell migration and angiogenesis
.
Adv. Drug Deliv. Rev.
63
:
610
615
.

Supplementary data