Abstract Carcinomas are associated with metastasis to specific organs while sparing others. Breast cancer presents with lung metastasis but rarely kidney metastasis. Using this difference as an example, we queried the mechanism(s) behind the proclivity for organ specific metastasis. We employed spontaneous and implant models of metastatic mammary carcinoma coupled with inflammatory tissue fibrosis, single cell sequencing analyses, and functional studies to unravel the causal determinants of organ-specific metastasis. Here we show that lung metastasis is driven by Angiopoietin-2 (Ang2) mediated suppression of lung specific endothelial tight junction protein Claudin 5, which is augmented by the inflammatory fibrotic microenvironment and prevented by anti-Ang2 blocking antibodies; while kidney metastasis is prevented by non-Ang2 responsive Claudin 2 and Claudin 10. Suppression of Claudin 2/10 was sufficient to induce emergence of kidney metastasis. This study illustrates the influence of organ specific vascular heterogeneity in determining organotropic metastasis, independent of cancer cell intrinsic mechanisms. Introduction Metastasis is the primary cause of morbidity and mortality in breast cancer, and accounts for about 90% of breast cancer-related deaths ^[62]1. Clinically, breast cancer metastasizes to different secondary sites, including bone, lung, liver, and brain, but very rarely to organs such as the kidney ^[63]2, despite the fact that 20% of total blood volume is associated with the renal parenchyma at any given time. The preferential spread of metastases to specific organs (organotropic metastasis) has been hypothesized to depend on interactions between ‘seed’ (cancer cells and their intrinsic properties) and ‘soil’ (or the host microenvironment) ^[64]3. Cancer cells may acquire novel genetic and/or epigenetic features to overcome microenvironmental pressures, with selection of clones with distinct fitness to different microenvironments at distant metastatic site(s). The parenchymal turnover rate, immune surveillance, metabolic fitness, angiogenic potential, biomechanical properties, and other features of the metastatic sites may contribute to the survival and adaptation of cancer cells, but may also present with restrictive inherent features that limit successful formation of metastatic disease. Organotropic metastasis is likely regulated by multiple factors, including organ-specific metastatic niche formation involving tumor cell-secreted factors and exosomes ^[65]4–[66]8. The vascular and perivascular niches may also play a role establishing metastasis and needs mechanistic elucidation ^[67]9–[68]13. The rate limiting function of specific parenchymal remodeling events in organotropic metastasis remains unknown. Given vascular remodeling is critical for hematological dissemination of cancer cells for metastasis to occur, we aimed to ascertain the mechanistic underpinning of organotropic metastasis, with a specific focus on the permissive and restrictive properties to blood vessels in this process. Studies suggest that tissue alterations associated with organ injury and fibrosis may promote metastatic outgrowth^[69]14–[70]17. While vascular remodeling is a hallmark of tissue repair in cancer and non-cancer pathologies, the functional and mechanistic significance of organ specific vascular heterogeneity in metastasis is yet to be defined. Results Lung fibrosis forms a pro-metastatic niche for lung metastasis Fibrosis is considered a precursor for future emergence of cancer and potentially serves as a metastatic niche for secondary spread of cancer ^[71]14,[72]18,[73]19. To evaluate the impact of the pro-metastatic milieu in the recruitment of lung metastasis, we induced lung fibrosis in 4T1 mammary tumor bearing mice to generate an inflammatory tissue microenvironment. Emergence of bleomycin-induced lung fibrosis was associated with prototypical parenchymal damage and remodeling that includes immune infiltration, deposition of extracellular matrix proteins such as collagens, accumulation of smooth muscle αactin expressing (αSMA^+) myofibroblasts, and transcriptional upregulation of fibronectin (Fn1) and type I collagen (Col1a1) ([74]Figure 1A-[75]B, [76]Extended Data Figure 1A-[77]D). Lung fibrosis was induced in 4T1 orthotopic tumor bearing mice when total tumor volume reached approximately 500 mm^3 ([78]Figure 1C). Induction of lung fibrosis had no significant impact on the growth kinetics of the primary mammary tumors ([79]Figure 1C), but significantly enhanced lung metastasis and surface lung nodules when compared to non-fibrotic lungs of tumor bearing mice ([80]Figure 1D-[81]E). An increase in GFP^+ cancer cells was noted in the fibrotic lungs compared to the non-fibrotic control lungs ([82]Extended Data Figure 1E), in agreement with previous studies ^[83]20. Similar results were observed in the spontaneous murine MMTV-PyMT metastatic mammary carcinoma model ([84]Figure 1F-[85]H). Lung fibrosis in this model, initiated with intratracheal bleomycin administration when the tumor burden reached approximately 500 mm^3, also failed to impact the growth kinetics of primary mammary tumors but significantly enhanced lung metastatic disease ([86]Figure 1F-[87]H). Despite enhanced lung metastasis observed in both models of mammary carcinoma with lung fibrosis, the metastasis to the liver, evaluated by histology and immunolabeling for GFP^+ cancer cells, was unchanged ([88]Extended Data Figure 1F-[89]H). Cancer cells were not detected in the heart and bowel ([90]Extended Data Figure 1I-[91]J). Collectively, these results provide evidence for the specific pro-metastatic potential of an inflammatory lung fibrotic microenvironment. Figure 1. Fibrosis enhances tropic metastasis with no impact on non-tropic sites. Figure 1. [92]Open in a new tab (A-B) Representative MTS of lung (A) and kidney (B) fibrosis. Scale bar, 100 μm. (C) Orthotopic 4T1 tumor volume over time +/− lung fibrosis, n=6 mice per group. (D) Representative lung H&E of 4T1 orthotopic tumor bearing mice +/− lung fibrosis. Scale bar, 25 μm. (E) Quantification of metastatic area and surface lung nodules in 4T1 orthotopic tumor bearing mice +/− lung fibrosis, n=6 mice per group. (F) MMTV-PyMT tumor volume over time +/− lung fibrosis, n=5 to 7 mice per group. (G) Representative lung H&E of MMTV-PyMT tumor bearing mice +/− lung fibrosis. Scale bar, 25 μm. (H) Quantification of metastatic area and surface lung nodules in MMTV-PyMT tumor bearing mice +/− lung fibrosis, n=5 to 7 mice per group. (I) Orthotopic 4T1 tumor volume over time +/− kidney fibrosis, n=6 mice per group. (J) Representative lung H&E of 4T1 orthotopic tumor bearing mice +/− kidney fibrosis. Scale bar, 25 μm. (K) Quantification of metastatic area and surface lung nodules in 4T1 orthotopic tumor bearing mice +/− kidney fibrosis, n=6 mice per group. (L) MMTV-PyMT tumor volume over time +/− kidney fibrosis, n=6 mice per group. (M) Representative lung H&E of MMTV-PyMT tumor bearing mice +/− kidney fibrosis. Scale bar, 25 μm. (N) Quantification of metastatic area and surface lung nodules in MMTV-PyMT tumor bearing mice +/− kidney fibrosis, n=5 to 6 mice per group. (O) Representative lung H&E of the lung of mice following 4T1 i.v. injection +/− lung fibrosis and +/− kidney fibrosis. Scale bar, 25 μm. (P) Quantification of lung colonization following 4T1 i.v. injection +/− lung fibrosis or kidney fibrosis, n=6 mice per group. Data are presented as mean values +/− SEM. ctrl: control, LF: lung fibrosis, KF: kidney fibrosis. For B, E, H, K, N, P: unpaired two-tailed t test with Welch’s correction applied for unequal variances (determined by F-test); for C, I: two-way ANOVA with Sidak’s multiple comparisons test; for F, L: multiple unpaired t tests of tumor measurements. P values are listed, ns: not significant. Kidney fibrosis does not promote kidney metastasis but enhances lung metastasis Breast cancer is rarely associated with metastasis to the kidney in humans or in mouse models of disease ^[93]21,[94]22. Based on the observation of increased lung metastasis induced by inflammatory lung fibrotic environment (vide supra), we decided to test the contribution of an inflammatory and pro-tumorigenic kidney fibrotic microenvironment on potential emergence of metastasis to the kidney. We induced fibrosis in the kidneys of 4T1 tumor bearing mice. Unilateral ureter obstruction (UUO) induced inflammatory kidney fibrosis with a prototypical parenchymal remodeling of fibrosis, similar to the changes observed in lung fibrosis ([95]Figure 1A-[96]B, [97]Extended Data Figure 1A-[98]D). Kidney fibrosis was induced in 4T1 orthotopic tumor bearing mice when the total tumor volume reached approximately 500 mm^3 ([99]Figure 1I). Kidney fibrosis had no significant impact on the growth kinetics of the primary tumor but significantly increased lung metastasis and surface lung metastatic nodules in two independently performed studies ([100]Figure 1J-[101]K, [102]Extended Data Figure 1K-[103]M). These histological findings were corroborated by immunolabeling for GFP^+ cancer cells, which were increased in frequencies in the lung of mice with kidney fibrosis compared to sham controls ([104]Extended Data Figure 1N-[105]O). Similar results were observed in the MMTV-PyMT transgenic mouse model with inflammatory kidney fibrosis ([106]Figure 1L-[107]N). Liver metastasis was unchanged with inflammatory kidney fibrosis ([108]Extended Data Figure 1P), and cancer cells were not detected in any other organs surveyed ([109]Extended Data Figure 1Q-[110]R). Despite inflammatory tissue remodeling (lung and kidney fibrosis) increasing metastasis to the lungs, neither 4T1 orthotopic or MMTV-PyMT models revealed metastasis to the parenchyma of kidneys ([111]Extended Data Figure 2A-[112]G). In MMTV-PyMT mice, PyMT^+ cancer cells were not detected in the parenchyma of kidneys ([113]Extended Data Figure 2G). The inflammatory fibrosis status in all experiments (4T1 orthotopic, 4T1 i.v., and or MMTV-PyMT models) were confirmed histologically ([114]Extended Data Figure 2H-[115]J). Moreover, in both the 4T1 orthotopic and 4T1 i.v. models, circulating cancer cells can be identified within the blood vessels of the kidney ([116]Extended Data Figure 3A). However, cancer cells were not detected in the parenchyma of perfused kidneys; but they were detected in kidneys without perfusion, confined within the vasculature, unable to exit the blood vessels. This was ascertained by flow analysis ([117]Extended Data Figure 3B-[118]C, [119]Supplementary Figure 1A-[120]B) and QPCR analysis ([121]Extended Data Figure 3D-[122]E). Taken together, these results establish that unlike inflammatory lung fibrosis, inflammatory kidney fibrosis does not induce or facilitate metastases to the kidney. Instead, we observe that inflammatory kidney fibrosis enhances metastasis to the lung. The impact of organ fibrosis on colonization of 4T1 cells administered intravenously via the orbital plexus was further validated using intracardiac injections, and in both cases metastatic colonization in the kidney was not observed ([123]Extended Data Figure 3F-[124]I). The use of intracardiac rouse of administration of 4T1 cancer cells eliminated the potential for first pass interaction of cancer cells with the lung capillary bed as a rate limiting factor for the lack of colonization in the renal parenchyma. Fibrosis from cutaneous wound injury does not impact metastasis We next examined whether inflammatory cutaneous dorsal wound injury and healing impacts metastasis. 4T1 metastatic mammary tumor bearing mice with cutaneous wounds did not display measurable differences in primary tumor growth kinetics or incidence of lung metastasis ([125]Extended Data Figure 3J-[126]L). Similarly, MMTV-PyMT mice with cutaneous wounds also did not reveal differences in primary tumor growth kinetics or incidence of lung metastasis ([127]Extended Data Figure 3M-[128]O). Wound closure rate was unchanged between tumor-bearing and non-tumor-bearing mice ([129]Extended Data Figure 3P). Taken together, these results indicate that the fibrotic remodeling associated with the cutaneous wound healing response has no impact on lung metastasis in this setting and does not induce metastasis to other organs. Cancer cells form tumor in the kidney parenchyma with lung metastasis Mice with either inflammatory lung fibrosis, kidney fibrosis, or cutaneous wounds were injected with 4T1 cancer cells intravenously to bypass the influence of primary tumor in metastatic colonization. In this setting, we observed increased colonization of cancer cells in fibrotic lungs when compared to control lungs ([130]Figure 1O-[131]P). Increased lung colonization was also observed in mice with kidney fibrosis compared to controls ([132]Figure 1O-[133]P). Lung colonization was not impacted by cutaneous wounds ([134]Extended Data Figure 3Q-[135]R). To define whether lack of metastasis or colonization of cancer cells in the kidneys was reflective of intrinsic, cell-autonomous properties of cancer cells, we tested their ability to establish tumors when implanted directly into the kidney parenchyma. The direct injection of 4T1 cancer cells into the kidney parenchyma resulted in tumor formation in the kidney, and inflammatory fibrosis further enhanced the ability to form mammary carcinoma tumors in the kidney ([136]Figure 2A-[137]F). Figure 2. 4T1 cells form tumors when injected under the renal capsule. Figure 2. [138]Open in a new tab (A) Experimental design for the evaluation of tumor formation in mice injected with 4T1 cells under the renal capsule +/− kidney fibrosis induced immediately post injection. (B) Representative kidney H&E of mice with 4T1 cells injected under the renal capsule and +/− kidney fibrosis. Scale bar, 100 μm. (C-D) Kidney weight (C) and quantification of primary tumor area (D) in kidneys of the indicated groups, n=7 mice per group. (E-F) Representative kidney H&E of mice with 4T1 cells injected under the renal capsule together without (E) or with kidney fibrosis (F). Scale bar, 20 μm. (G-H) Representative images for GFP immunolabeling (G) and quantification of percent of the GFP positive area (H) in the indicated groups, n=4 mice per group. Scale bar, 50 μm. (I) Representative lung H&E images of mice with 4T1 cells injected under the renal capsule +/− kidney fibrosis. Scale bar, 25 μm. (J-K) Quantification of metastatic area/whole lung (J) and surface lung nodules (K) in the indicated groups, n=7 mice per group. (L-M) Representative H&E images of kidney in mice with 4T1 i.v. administration and with lung (L) or kidney fibrosis (M). Scale bar, 25 μm. (N) Number of mice with microscopic kidney tumors in the indicated groups, n=6 mice per group. Data are presented as mean values +/− SEM. For D, H, J, K: unpaired two-tailed t test, with Welch’s correction applied for unequal variances (determined by F-test). For C: one-way ANOVA with Tukey multiple comparisons test. P values are listed, ns: not significant. Evaluation of kidneys for GFP^+ cancer cells further confirmed the increased tumor burden in fibrotic kidneys compared to non-fibrotic control kidneys ([139]Figure 2G-[140]H). These experiments demonstrate that inflammatory kidney fibrosis has the capacity to enhance local tumor growth, similar to inflammatory lung fibrosis. Mammary tumors induced in the kidney resulted in lung metastasis, despite not originating from the mammary fat pads ([141]Figure 2I-[142]K). It also suggests that the permeability of blood vessels associated with the primary tumor in the kidney may be different when compared to the endogenous normal kidney vasculature. Colonization studies with intravenous administration of cancer cells revealed no metastasis to the kidney in mice with either inflammatory lung fibrosis or inflammatory kidney fibrosis ([143]Figure 2L-[144]N), unlike the increase in lung colonization associated with inflammatory lung fibrosis ([145]Figure 1O-[146]P). Taken together, these results suggest that inflammatory lung fibrosis and kidney fibrosis exhibit equal potential to aid local tumor growth mediated by pro-tumor growth mediators and that circulating factors from the inflammatory fibrotic kidney, but not healing cutaneous wounds, might play a role in the lung tissue to increase metastasis. Lung or kidney fibrosis induces lung specific vascular remodeling To explore the underlying mechanism(s) associated with the enhanced lung metastasis induced by lung and kidney fibrosis, we evaluated global transcriptomic changes in healthy lungs (HL), healthy kidneys (HK), fibrotic lungs (LF), fibrotic kidney (KF) and lungs from mice with kidney fibrosis (LoKF). Fibrotic remodeling in both lung and kidneys showed similar patterns of transcriptional changes associated with extracellular matrix remodeling, fibroblast proliferation, fibronectin binding, collagen signaling, and metabolism ([147]Extended Data Figure 4A-[148]B, [149]Supplementary Table 1). Analyses of differentially expressed genes (DEGs) revealed an increased expression of 2830 genes and decreased expression of 3235 genes in fibrotic lung when compared to the healthy lung ([150]Figure 3A-[151]B). Increased expression of 1173 genes and decreased expression of 999 genes in lungs from mice with kidney fibrosis was observed when compared to healthy lung ([152]Figure 3C-[153]D). The gene ontology enrichment of the comparative analyses revealed vascular ([154]Figure 3B, [155]D; [156]Supplementary Tables 2-[157]3) and immune ([158]Extended Data Figure 4C-[159]D; [160]Supplementary Tables 2-[161]3) remodeling in lungs with fibrosis and lungs from mice with kidney fibrosis. Overlapping upregulated or downregulated genes between LF vs HL and lungs from mice with kidney fibrosis vs HL confirmed significant vascular remodeling in both LF and LoKF compared to the HL ([162]Extended Data Figure 4E-[163]F, [164]Supplementary Table 4). Inflammatory and vascular remodeling cytokines ar also upregulated in both LF and KF ([165]Extended data Figure 4G), and collectively, these data suggest a role for a potential regulator of lung vascular remodeling that is induced by kidney fibrosis. Figure 3. Ang2 mediates fibrosis-induced lung metastasis. [166]Figure 3. [167]Open in a new tab (A, C) Volcano plots depicting the number of differentially regulated genes in healthy lung vs. lung fibrosis (A) and healthy lung vs. lung from mice with kidney fibrosis (C). (B, D) Gene ontology (GO) enrichment ratio and false discovery rate (FDR) values for deregulated genes implicated in vascular remodeling in lung with fibrosis (B) or lung of mice with kidney fibrosis (D). The numbers of deregulated genes out of all the genes in the pathways are indicated in parenthesis with a star denoting common pathways in B and D. (E) Heatmap of relative mean levels of the listed cytokines in the serum of mice in the indicated group. Healthy, n=2; wound, n=2; lung fibrosis, n=3; kidney fibrosis, n=3 mice. (F) ELISA quantification of circulating Ang2 levels in the serum of mice in the indicated groups. Healthy, n=5; wound, n=3; 4T1 orthotopic tumor bearing mice (T), n=4; control for lung fibrosis (ctrl LF), n=6; lung fibrosis, n=6; control for kidney fibrosis (ctrl KF), n=6; kidney fibrosis (KF), n=5 mice. (G-H) Representative images and respective quantification of dextran extravasation (green) and CD31 immunolabeling (red) in the lung (G) and kidney (H) after rAng2 or PBS control treatment (individual images in [168]Supplementary Figure 2). Scale bar, 20 μm. Quantification of FITC-Dextran area per CD31 positive area, n=4 to 5 mice per group. (I-J) Representative lung H&E of mice with PBS or rAng2 treatment and with (J) and without (I) i.v. injection of 4T1 tumor cells. Scale bar, 200 μm. Quantification of pleural edema and thickness in the lungs, n=5 to 6 mice per group (I), and quantification of lung colonization and surface lung nodules (J) in the indicated groups, n=5 to 6 mice per group. Data are presented as mean values +/− SEM. Unpaired two-tailed t test, with Welch’s correction applied for unequal variances (determined by F-test). P values are listed, ns: not significant. Flow cytometry was conducted to further evaluate the immune microenvironment of the lung in response to lung fibrosis (LF) and kidney fibrosis (KF). The overall number of CD45^+ leukocytes was not changed upon LF and KF, but the percent of CD3^+ T cells were significant reduced. A significant increase in CD11b^+ myeloid cells and CD11b^+Ly6G^+Ly6C^- neutrophils were observed and similar immune cell alterations were also observed in the context of renal fibrosis, suggesting that neutrophils may be involved in the increased lung metastasis. However, effector T cells (CD4^+FoxP3^-), cytotoxic T cells (CD8^+), gamma delta (γδ^+) T cells, regulatory T cells (CD4^+FoxP3+), dendritic cells (CD11c^+) and B cells (CD19^+) were specifically altered in the lung of mice with LF but not KF. No change on CD11b^+ Ly6C^+ Ly6G^- and natural killer (NK1.1^+) cells ([169]Extended Data Figure 5A, [170]Supplementary Figure 1C-[171]D). We similarly evaluated the immune infiltrate of the kidney in response to LF and KF and found that a significant upregulation of CD45^+ cells, regulatory T cells (CD4^+FoxP3+), natural killer (NK1.1^+) cells, and B cells (CD19^+), CD11b^+ myeloid cells and CD11b^+ Ly6C^+ Ly6G^- cells was observed in the kidneys of mice with KF but not LF. A significant down-regulation of CD3^+ T cells and effector T cells (CD4^+FoxP3^-) were found in the kidneys of mice with KF but not LF. No change could be found on cytotoxic T cells (CD8^+), gamma delta (γδ^+) T cells, dendritic cells (CD11c^+) and CD11b^+Ly6G^+Ly6C^- neutrophils in the kidney of mice with KF ([172]Extended Data Figure 5B, [173]Supplementary Figure 1C-[174]D). The analyses further support that neutrophils may play collaborative role in aiding lung metastasis but by themselves they do not determine kidney metastasis. Taken together, these findings suggest that the immune microenvironment in fibrotic lungs could promote the proliferation of mammary carcinoma cells in lung, but it fails to facilitate the growth of cancer cells in the kidney, despite similar changes in immune cell composition. To evaluate the cytokines/chemokines secreted from the fibrotic kidney that may enhance vascular remodeling and favor metastasis, we performed targeted cytokine array using the serum from healthy mice, mice with cutaneous wounds (wound), mice with lung fibrosis (LF), and mice with kidney fibrosis (KF). Several cytokines were uniquely upregulated in KF and LF compared to healthy mice and mice with cutaneous wounds, suggesting their involvement in the vascular remodeling of the lungs. Induced inflammatory cytokines LIX/Cxcl5 and Reg3G, neutrophil products lipocalin-2/NGAL, myeloperoxidase, endothelial cell regulator angiopoietin-2 (Ang2), resistin, and osteopontin were identified as candidate effectors ([175]Figure 3E). To ascertain the cytokines specifically upregulated in LF and KF compared to healthy and cutaneous wound carrying mice, the data was normalized and ranked. Amongst the cytokines implicated in vascular remodeling, Ang2 was the most differentially upregulated in kidney fibrosis compared to healthy kidney and lung ([176]Figure 3E, [177]Extended Data Figure 4G). Ang2 is associated with perivascular remodeling in the primary tumor and its impact on endothelial cell signaling plays a rate limiting role in metastatic dissemination ^[178]13,[179]23–[180]29. While our prior studies demonstrate the importance of vascular remodeling by Ang2 at the primary tumor site ^[181]13,[182]25, the impact of vascular heterogeneity and permeability in determining organotropic metastasis is unknown. Validation studies using quantitative ELISA showed an increase in the levels of Ang2 in the serum of mice with kidney fibrosis when compared to control mice, with the levels of Ang2 being similar to those found in mice with 4T1 mammary carcinoma (T, tumor bearing mice, [183]Figure 3F). These results suggest that Ang2 is upregulated in the serum of mice with lung metastasis associated with kidney fibrosis, whereas its impact may be locally driven in the context of lung fibrosis. Interestingly, circulating Ang2 has also been reported as being increased in patients with breast cancer where elevated levels are associated with reduced survival, higher histological grade, more advanced clinical stage, and presence of lymph node metastasis^[184]30. Moreover, breast cancer patients with three or more metastatic sites demonstrate increased circulating Ang2 compared to patients with less metastatic sites^[185]31. Taken together, the results implicate Ang2 as a potential mediator of both kidney fibrosis and lung fibrosis-induced enhancement of lung metastasis. Ang2 compromises lung vascular permeability in fibrosis-induced lung metastasis We next evaluated whether the observed increase in circulating Ang2 plays a role in vascular permeability at the metastatic site to promote colonization of lungs by cancer cells. We treated healthy mice with recombinant mouse Ang2 protein (rAng2) and assessed the lungs, kidneys, and livers from these mice. rAng2 was administered intraperitoneally before intravenous infusion of FITC-conjugated dextran. Enhanced extravasation of FITC-dextran was observed in the lungs after rAng2 administration compared to controls, indicating that rAng2 increases vascular permeability in the lungs ([186]Figure 3G, [187]Supplementary Figure 2). In contrast, no increase in vascular permeability was noted in the kidney and liver following rAng2 administration ([188]Figure 3H, [189]Extended Data Figure 5C, [190]Supplementary Figure 2). The lung parenchyma of mice administered rAng2 showed significant edema and increased pleural thickness compared to controls ([191]Figure 3I, [192]Extended Data Figure 6A). This is similar to the histopathological changes noted in fibrotic lungs and lungs of mice with kidney fibrosis ([193]Extended Data Figure 6B-[194]E). There was no appreciable change in the kidney parenchyma following rAng2 administration ([195]Extended Data Figure 6F) or in the kidneys of mice with lung or kidney fibrosis ([196]Extended Data Figure 6G-[197]H). Exogenous administration of Ang2 (rAng2) was sufficient to significantly enhance lung colonization of intravenously administered 4T1 cancer cells ([198]Figure 3J, [199]Extended Data Figure 6I), with no localization observed in the kidney ([200]Extended Data Figure 6J). These results are consistent with the increased 4T1 lung colonization observed in the context of lung or kidney fibrosis ([201]Figure 1O-[202]P). Collectively, these results support the effect of Ang2 on lung vascular permeability and its consequent impact on lung metastasis without measurable impact in the kidney. Ang2 neutralization reverses kidney fibrosis-induced enhanced lung metastasis To determine the rate limiting role of Ang2 on lung metastasis, we evaluated primary tumor growth kinetics and metastatic disease in mice treated with anti-Ang2 neutralizing antibodies (anti-Ang2) ^[203]25. Evaluation of 4T1 primary tumors revealed similar growth kinetics in control IgG treated mice with or without kidney fibrosis, while anti-Ang2 treatment significantly reduced tumor burden in both groups (with and without kidney fibrosis) by approximately 30% ([204]Figure 4A). These results are consistent with previous reports ^[205]25 and likely reflect the impact of anti-Ang2 on intratumoral vascular permeability. In the absence of fibrosis, mice with 4T1 orthotopic tumors treated with either control IgG or anti-Ang2 presented with similar amounts of lung metastasis ([206]Figure 4B-[207]D). Anti-Ang2 treatment reduced the lung metastasis area and the number of microscopic lung nodules in mice with kidney fibrosis to the baseline levels observed in control (no kidney fibrosis) groups ([208]Figure 4B-[209]D). Histopathological analyses of the kidneys revealed that anti-Ang2 treatment did not impact kidney fibrosis when compared to the lgG control group ([210]Figure 4E-[211]G). Our results support a rate-limiting role for systemic circulating Ang2 released by kidney fibrosis, with an impact on lung metastasis associated with mammary carcinoma. Figure 4. Ang2 neutralization is sufficient to revert lung metastasis induced by kidney fibrosis. Figure 4. [212]Open in a new tab (A) Orthotopic 4T1 tumor volume over time +/− kidney fibrosis (KF) and IgG or anti-Ang2 treatment, n=9 mice per group. Arrows indicate a ~30% reduction in tumor volume at experimental endpoint with anti-Ang2 compared to IgG (ctrl groups). (B-C) Quantification of metastatic area (B) and number of microscopic lung nodules (C) in the indicated groups, n=9 mice per group. (D) Representative lung H&E of mice in the indicated groups. Scale bar, 25 μm. (E) Representative images of kidneys with H&E, MTS, and Sirius Red staining from the indicated groups. Scale bar, 25 μm. (F-G) Quantification of positive MTS (F) and Sirius Red stained area (G) in the indicated groups, n=9 mice per group. (H) Relative mRNA expression levels of Cldn2, Cldn3, Cldn4, Cldn5, Cldn7, Cldn10, Cldn17, Cldn19, Jam2, Jam3, Occludin and Tjp1 in healthy kidney, lung, and liver tissues. Expression value is normalized to endogenous Pecam1 (CD31) levels and is presented as 2^-ΔCt, n=3 mice per group. Data are presented as mean values +/− SEM. For A: two-way ANOVA with Tukey’s multiple comparisons test. For B, C, F and G: one-way ANOVA test with Tukey’s multiple comparisons test. P values are listed, ns: not significant. Ang2–Foxo1 signaling suppresses Claudin 5 in lung endothelial cells Ang2 has been previously implicated in breast cancer metastasis via the impairment of tight junction (TJ) integrity ^[213]32. Claudins (Cldns) are critical structural and functional components of TJ intercellular adhesion complexes ^[214]33. Evaluation of key TJ components, including Cldn2, Cldn3, Cldn4, Cldn5, Cldn7, Cldn10, Cldn17, Cldn19, Jam2, Jam3, Occludin and Tjp1, showed that Cldn5 was uniquely expressed at higher levels in healthy lungs when compared to healthy liver and kidney ([215]Figure 4H). Single-cell RNA transcriptome sequencing (scRNA-seq) of murine endothelial cells across 11 tissues ^[216]34, revealed that Cldn5 was highly expressed in endothelial cells from the lung, brain, and testes, and could also be found in kidney and liver endothelial cells ([217]Figure 5A-[218]B). Notably, Cldn5 was uniquely enriched in endothelial cells of both murine and human scRNA-seq analyses of healthy lungs ^[219]35 ([220]Figure 5C-[221]E, [222]Extended Data Figure 7A-[223]C), whereas the expression of Claudins in the kidney endothelium revealed more diversity, with not only Clnd5, but also Cldn2, 3, 4, 6, 7, 8, 10, 12, 16, and 19 noted in murine kidney endothelial cells ^[224]36 ([225]Figure 5F-[226]G, [227]Extended Data Figure 7D), and CLDN2–5, 7, 8, 10, 12, 15 and 23 in human kidney endothelial cells ^[228]37 ([229]Extended Data Figure 7E). Immunolabeling experiments confirmed the expression of Cldn5 in CD31-expressing endothelial cells in the liver, brain, kidney, and lung tissues ([230]Figure 5H, [231]Supplementary Figure 3A-[232]D). Immunolabeling studies showed Cldn5 was more frequently associated with CD31^+ vascular endothelial cells rather than Lyve1^+ lymphatic endothelial cells ([233]Figure 5 H-[234]I, [235]Extended Data Figure 8A-[236]C, [237]Supplementary Figures 4A-[238]B). In addition to Cldn5, other Claudins are expressed in murine kidney endothelial cells ([239]Figure 5G, [240]Extended Data Figure 8D-[241]G). Cldn2 and Cldn10 were also expressed in both endothelial and epithelial TJs in the kidney and brain parenchyma ^[242]38. Immunolabeling for Cldn2 and Cldn10 co-localized with CD31^+ vascular endothelial cells in both the cortex and medulla ([243]Extended Data Figure 8H-[244]I, [245]Supplementary Figure 5A-[246]D). scRNA-seq data also pointed to diverse Cldn genes expressed epithelial and endothelial cells in murine kidney, whereas lung and brain endothelial cells show specific Cldn5 expression ([247]Extended Data Figure 8D-[248]G). Collectively, these studies support a role for Cldn5 in the lung (and brain) endothelial TJs, and a role for multiple Claudins, including Cldn5, Cldn2 and Cldn10 in kidney endothelial TJs. Given the enriched Cldn5 expression in lung endothelial TJs, we sought to determine whether the increased lung vascular permeability induced by Ang2 was, in part, a result of Cldn5 downregulation as previously supported in other studies ^[249]39,[250]40. The lungs of mice with lung fibrosis, kidney fibrosis, or rAng2 treatment consistently revealed a reduced endothelial Cldn5 expression when compared to control lungs ([251]Extended Data Figure 8J-[252]K, [253]Supplementary Figure 6A-[254]D). Inhibition of Ang2 signaling in kidney fibrosis with neutralizing anti-Ang2 administration failed to show a reduction in endothelial Cldn5 ([255]Extended Data Figure 8J-[256]K, [257]Supplementary Figure 6A-[258]D). Taken together, the results support Ang2 downregulate Cldn5 in vivo in murine lung endothelial cells. Figure 5. Differential Claudin expression levels in lung and kidney. [259]Figure 5. [260]Open in a new tab (A-B) t-SNE plots (A) and violin plots (B) of scRNA-seq evaluation of Cldn5 mRNA levels in endothelial cells from the listed organs. (C-D) t-SNE plot of sRNA-seq of murine lung cells (C) with a focus on endothelial cells (EC) showing the relative mRNA levels for Cldn2, Cldn5 and Cldn10 (D). (E) tSNE plots and analysis of Claudin 5 and CD31 colocalization percentage in mouse lung endothelial cells (EC). (F-G) t-SNE plot of sRNA-seq of murine kidney cells (F) with a focus on endothelial cells (EC) showing the relative mRNA levels for Cldn2, Cldn5 and Cldn10 (G). (H) Colocalization of CD31 (green) and Claudin 5 (red) in WT liver, brain, and kidney. Scale bar, 25 μm (individual images in [261]Supplementary Figures 3, [262]4A). (I) Colocalization of Lyve-1 (green) and Claudin 5 (red) in lung. Scale bar, 25 μm (individual images in [263]Supplementary Figure 4B). Data are presented as mean values +/− SEM. To define the signaling events associated with Ang2 mediated downregulation of Cldn5, we evaluated FOXO1 in lung endothelial cells treated with rAng2. Previous studies reported that, in an inflammatory state with down regulation of Tie1, Ang2 antagonist action on Tie2 leads to FOXO1 activation in endothelial cells ^[264]40. FOXO1 activation and nuclear translocation results in downregulation of Cldn5 in endothelial cells ^[265]39. Murine lung microvascular endothelial cells showed transcriptional downregulation of Cldn5 upon Ang2 treatment ([266]Figure 6A-[267]B). Cldn5 protein levels were also downregulated in lung endothelial cells following rAng2 treatment ([268]Figure 6C-[269]E), and this was associated with a nuclear enrichment in FOXO1 ([270]Figure 6F). Jam2, Cldn3 and Cldn4 mRNA levels were unaltered ([271]Figure 6G). Decreased expression of Cldn5 was also noted in brain endothelial cell following rAng2 treatment ([272]Figure 6H), corroborating previous findings ^[273]32. Figure 6. Ang2 downregulates Claudin 5 via nuclear enrichment of FOXO1. [274]Figure 6. [275]Open in a new tab (A) Experimental design for the evaluation of Ang2 mediated Claudin 5 downregulation in the lung endothelial cells. (B) Relative Cldn5 mRNA levels in lung endothelial cells treated with the rAng2 (PBS control normalized to 1) for the indicated timepoints, n=3 biologically independent experiments (n=3 PBS and n=3 rAng2 with each 2 technical replicates). (C-D) Relative Claudin 5 protein levels in lung endothelial cells treated with rAng2 for the indicated timepoints, n=5 biologically independent experiments (n=5 PBS and n=5 rAng2). Uncropped blots are shown in the Image Source Data File. (E) Representative immunofluorescence staining of Claudin 5 in lung endothelial cells after treatment with rAng2 for 24h. n=1 biologically independent experiment. Scale bars, 25 μm. (F) FOXO1 protein levels in nuclear and cytoplasmic fractions at the indicated timepoints, with alpha-tubulin and Lamin A/C as markers for cytoplasmic and nuclear fraction, respectively. n=1 biologically independent experiment. Uncropped blots are shown in the Image Source Data File. (G) Relative Jam2, Cldn3 and Cldn4 mRNA levels of in lung endothelial cells after treatment with rAng2 for 24h, n=3 biologically independent experiments (with each 3 technical replicates). (H) Relative Cldn5 mRNA levels of in brain endothelial cells after treatment with the rAng2 for 24h, n=3 biologically independent experiments (with each 2 technical replicates). Data are presented as mean values +/− SEM. Unpaired two-tailed t test, with Welch’s correction applied for unequal variances (determined by F-test). P values are listed, ns: not significant. Claudin 5 tight junction compromised by Ang2 enhances lung metastasis To determine the direct contribution of Cldn5 reduction by Ang2 in lung metastasis, we performed rescue experiments where Cldn5 was overexpressed via the intratracheal administration of Cldn5 overexpressing adenovirus (Ad-Claudin 5) before the intravenous infusion of 4T1 cancer cells, with or without rAng2 administration ([276]Figure 7A). Following the in vitro validation of the reagents ([277]Extended Data Figure 9A-[278]B; Ad-ctrl: control adenovirus), in vivo studies revealed a decrease in Cldn5/CD31 double positive area after rAng2 treatment by immunofluorescence labeling, while overexpression of Cldn5 induced by intratracheal Ad-Claudin 5 prevented such downregulation ([279]Figure 7B-[280]C, [281]Supplementary Figure 7). Consistent with our previous findings ([282]Figure 3I-[283]J), rAng2 treatment significantly increased 4T1 lung colonization ([284]Figure 7D-[285]E). Concurrent overexpression of Cldn5 in this context, however prevented the increased 4T1 lung colonization ([286]Figure 7D-[287]E). Figure 7. Critical role of Claudin 5 in 4T1 cell extravasation in lung. Figure 7. [288]Open in a new tab (A) Experimental design for the evaluation of Cldn5 overexpression (Ad-Claudin 5) in mice with 4T1 i.v. injection +/− rAng2 treatment. (B) Representative colocalization of Claudin 5 and CD31 in the lung of mice in the indicated groups (individual images in [289]Supplementary Figure 7). Scale bars, 50 μm. (C) Quantification of percentage of Claudin 5/CD31 double positive area per visual field in the lung of mice in the indicated groups, n=4 to 5 mice per group. (D) Representative lung H&E of mice in the indicated groups. Scale bar, 25 μm. (E) Quantification of lung colonization area of mice in the indicated groups, n=4 to 5 mice per group. (F) Experimental design for the evaluation of the downregulation of Claudin 5 (AAV-shRNA) in mice with 4T1 orthotopic tumors. Dox: doxycycline in the drinking water (Dox on). (G) Representative colocalization of Claudin 5 and CD31 in the lung of mice in the indicated groups individual images in [290]Supplementary Figure 8). Scale bar, 50 μm. (H) Quantification of percentage of Claudin 5/CD31 double positive area per visual field in the lung of mice in the indicated groups, n=4 to 5 mice per group. (I) Representative lung H&E of mice in the indicated groups. Scale bar, 25 μm. (J) Quantification of lung metastatic area in mice in the indicated groups, n=4 to 5 mice per group. Data are presented as mean values +/− SEM. One-way ANOVA with Tukey ‘s multiple comparisons test. P values are listed. Next, the downregulation of Cldn5, using doxycycline controlled Cldn5 shRNA AAV administered intratracheally was performed to determine whether the suppression of Cldn5 was sufficient to increase lung metastasis of 4T1 mammary carcinoma ([291]Figure 7F, [292]Extended Data Figure 9C). A decrease in Cldn5/CD31 double positive area was confirmed using immunolabeling ([293]Figure 7G-[294]H, [295]Supplementary Figure 8). The downregulation of Cldn5 in the lung was found to be sufficient to increase lung metastatic disease when compared to controls ([296]Figure 7I-[297]J), with no impact on the growth kinetics of the primary 4T1 tumor growth ([298]Extended Data Figure 9D). Additionally, reduced CLDN5 mRNA expression in metastatic lungs was found in patients with breast cancer compared to healthy lung ([299]Extended Data Figure 9E). Taken together, our results support a role for vascular Cldn5 in the lung as one of the gatekeepers of metastasis. Claudin 2/10 suppression enables metastasis to the kidney We next assessed whether the loss of Cldn2 and Cldn10, along with Ang2 mediated suppression of Cldn5, would be sufficient to generate a metastasis-permissive vascular microenvironment due to compromised vascular permeability in the kidney. We employed CRISPR/Cas9 knockdown of Cldn2 and Cldn10 expression in kidneys ^[300]41, and confirmed that this approach yields a significant reduction in Cldn2 and Cldn10 in mouse kidney endothelial cells ([301]Extended Data Figure 9F-[302]J). Fluorescence-activated cell sorting (FACS) purified CD31^+ kidney endothelial cells revealed lower Cldn2 and Cldn10 mRNA levels compared to E-Cadherin^+ epithelial cells ([303]Extended Data Figure 9H). Twenty-six days after administration of CRISPR/Cas9 constructs directly into the renal cortex to suppress Cldn2 or Cldn10 in mice with 4T1 tumors, we confirmed a reduction in Cldn2 and Cldn10 in the FACS purified CD31^+ endothelial cells, albeit with limited downregulation in E-Cadherin^+ epithelial cells ([304]Extended Data Figure 9I-[305]J, [306]Supplementary Figure 9A). 4T1 tumors showed similar primary tumor growth kinetics ([307]Extended Data Figure 9K-[308]L) and lung metastasis when Cldn2/10 were downregulated in the kidney ([309]Extended Data Figure 9M-[310]O). Importantly, kidney metastasis, evaluated by histopathology and immunolabeling for GFP^+ cancer cells, was detected in mice with Cldn2/10 downregulation compared to controls ([311]Extended Data Figure 10A-[312]E), effectively demonstrating the formation of metastasis to a non-organotropic location for 4T1 mammary carcinoma. This experiment was repeated with inclusion of additional experimental arm, in which mice were also subjected to kidney fibrosis ([313]Figure 8A). In this context, lung metastasis was increased with kidney fibrosis without impact from the suppression of Cldn2/10, and no impact on primary tumor growth was also observed ([314]Figure 8B-[315]C, [316]Extended Data Figure 10F). Suppression of Cldn2/10 in the kidney revealed kidney metastasis, which was further increased with kidney fibrosis ([317]Figure 8D-[318]G, [319]Extended Data Figure 10G, [320]Supplementary Figure 9B). Figure 8. Knockdown of Claudin 2/10 in kidney fibrosis enables renal metastasis. Figure 8. [321]Open in a new tab (A) Experimental design for the evaluation of the downregulation of Claudin 2 and Claudin 10 in kidney together with induction of kidney fibrosis in mice with 4T1 orthotopic tumors. (B) Representative lung H&E of mice in the indicated groups. Scale bar, 25 μm. (C) Quantification of lung metastatic area. LentiCRISPRv2 control vector without fibrosis, n=14; LentiCRISPRv2 control vector with fibrosis, n=11; LentiCRISPRv2 Cldn2 and Cldn10 without fibrosis, n=12 mice; LentiCRISPRv2 Cldn2 and Cldn10 with fibrosis, n=10 mice. (D) Representative kidney H&E of mice in the indicated groups. Scale bar, 50 μm (upper), 25 μm (lower). (E-F) Representative lung (E) and kidney (F) images for GFP immunolabeling in the indicated groups and associated quantification. E: scale bar, 50 μm, F: scale bar, 25 μm. (G) Percentage of mice with kidney lesion in the indicated groups. (H) Graph abstract. Data are presented as mean values +/− SEM. For C, E: one-way ANOVA with Tukey’s multiple comparisons test. For F: Kruskal–Wallis with Dunn’s multiple comparison test. For G: two-sided Chi-square. P values are listed, ns: not significant. Intravascular cancer cells were observed in the kidney ([322]Extended Data Figure 10H, [323]Supplementary Figure 10A-[324]B) but the metastatic growth in the kidney was detected in the parenchyma only upon vascular tight junction compromise ([325]Extended Data Figure 10I, [326]Supplementary Figure 10C). CD31 staining revealed that the vessel numbers were unchanged and suppression of Cldn 2/10 in kidney had no impact on primary mammary tumor vasculature ([327]Extended Data Figure 10J-[328]K). Collectively, our data indicates that destabilization of kidney endothelial cell TJs and the compromised vascular permeability resulting from suppressing Cldn2/10 enables the atypical formation of metastasis to the kidney, which is enhanced with inflammatory fibrosis. Our findings show that Cldn5 in the lung is uniquely susceptible to Ang2 mediated vascular destabilization and lung metastasis, and organotropism of metastasis, at least in part, is driven by endothelial TJ tissue specific composition ([329]Figure 8H). Discussion Organ specific tropism of metastasis associated with different cancers has long been a subject of intrigue and scientific investigation ^[330]42. The question as to why certain cancers rarely result in metastasis to certain organs but do so with high frequency to other organs remains under explored. Here, we employed mouse models of metastatic mammary carcinoma to evaluate the molecular underpinnings of frequent breast cancer metastasis to the lung but not to the kidney. Collectively, our studies demonstrate that when the extravasation step is bypassed, mammary carcinoma cells become equally well adapted to grow in the lungs and kidneys, and form established tumors. This finding suggests that cancer cell intrinsic mechanisms are not sufficient to enable their growth into tumors in the kidney, as shown with the models studied here. In different spontaneous metastasis models of mammary carcinoma that depend on extravasation of cancer cells, it remains clear how growth of secondary tumors in the kidney is prevented despite robust growth of metastatic tumors in the lung. In this experimental setting, the absence of metastasis to the kidney seems to depend on the vasculature-mediated barrier against extravasation of cancer cells, rather than on cancer cell intrinsic mechanisms and pre-metastatic niche. Previous studies that conducted extravasation and colonization assays using i.v. infusion of melanoma cells and ectopic tissue implantation revealed the critical role of the local environment in influencing lung and kidney metastasis ^[331]43. However, this approach involved the injection of several-fold higher numbers of cultured melanoma cells directly into circulation, unlike the number of circulating cancer cells originating from a primary tumor ^[332]43. Hence, the rationale behind this selective pattern of tumor growth is likely more intricate than tissue damage alone that creates a conducive microenvironment. This also suggests the involvement of different vascularization patterns on influencing metastasis^[333]43. Ang2 expression is upregulated by hypoxia, a known feature of the primary tumor microenvironment and inflammatory fibrotic microenvironment (IFM). In this study, the induction of IFM in either the lung or kidney leads to secretion of Ang2 along with the simultaneous activation of inflammatory signaling networks. Notably, vasculature response played a critical role in amplifying these inflammatory signals, leading to the formation of a pro-metastatic vascular niche ^[334]44,[335]45. Our results support that Ang2 increases vascular permeability in the lung but not the kidney, leading to efficient extravasation of the cancer cells into the lung and enabling metastatic colonization in the lung but not the kidney. Despite the findings reported by Chang et al. that Ang2 inhibition using a peptide-Fc fusion (L1–10) attenuated kidney fibrosis ^[336]46, our experiments using anti-Ang2 neutralizing antibodies did not yield any significant impact on renal fibrosis. This discrepancy in results could be attributed to variations in treatment modality, dosing, timing, and readouts. Robust mammary carcinoma cell growth and tumor formation upon direct injection into the renal tissue support that a mechanism maintaining vascular integrity, even when endothelial cells are exposed to Ang2, is a major factor underlying the lack of metastasis to the kidneys. Importantly, many host factors can contribute to emergence of metastasis in a given tissue and here we demonstrate that molecular features of vasculature also plays a causal role in restricting metastasis^[337]43. Single cell analyses of endothelial cells from the lung and the kidney revealed that the endothelial TJ in the lung predominantly contains Cldn5, which can be downregulated by Ang2, while the endothelial TJ in the kidney contains low levels of Cldn5 along with two non-Ang2 responsive Claudins–Cldn2 and Cldn10. Ang2, a known inducer of lung vascular permeability ^[338]47, regulates Cldn5 downregulation via engagement of FOXO1 signaling ^[339]39,[340]40. A positive correlation has been reported between pulmonary fibrosis and circulating Ang2 levels ^[341]48, and Ang2 can directly provoke lung vascular hyperpermeability and induce pulmonary edema ^[342]47,[343]49,[344]50. Ang2 is a known ligand of the receptor tyrosine kinase Tie2, and the activation of Tie1/2 signaling by angiopoietins is not only important for vascular homeostasis, but also critical in the tumor angiogenic switch and metastasis ^[345]51. Specifically, circulating Ang2 levels are associated with disease progression and survival in metastatic malignant breast cancer ^[346]30,[347]52, and a positive correlation of Ang2 and a pro-metastatic outcome has been reported ^[348]25,[349]32,[350]53. Our data support a direct role of Ang2 in lung vascular permeability, making this organ specifically permissive to metastasis. Direct suppression of Cldn5 in the lung vasculature results in increased lung metastasis. Rescue studies with overexpression of Cldn5 in the context of elevated Ang2 further implicates Cldn5 amongst the gatekeepers of lung metastasis. CRISPR mediated suppression of Cldn2 and Cldn10, along with Ang2 mediated suppression of Cldn5 in the kidney vasculature, enabled extravasation of the cancer cells and resulted in mammary carcinoma metastasis to the kidney. These studies directly implicate a role for vascular endothelial cells as potential rate-limiting regulators of organ specific metastasis. Our proof-of-concept studies support that heterogeneity within endothelial cell junctions in different organs is likely a key contributor of organotropism associated with mammary carcinoma and support further testing of Ang2 targeting to control lung metastasis. Methods All research presented here complies with ethical regulation, namely the Institutional Animal Care and Use Committee of MDACC. Further information on research design is available in the Nature Research Reporting Summary linked to this article. Cell Culture 4T1-GFP cells (4T1-turbo GFP and 4T1-luc-GFP) were modified using 4T1 BALB/c mammary carcinoma epithelial cells (Expasy RRID: CVCL 0125, from the Characterized Cell Line Core laboratory at MD Anderson Cancer Center), transduced to stably express TurboGFP (Origene, TR300007), or infected to stably express eGFP and luciferase (as previously described ^[351]16. 4T1 cells were maintained in RPMI 1640 medium supplemented with 10% heat-inactivated FBS (Gemini), penicillin (100 U/mL) and streptomycin (100 μg/mL) (Corning, VWR45000–652). When needed, culture medium was also supplemented with 1 μg/mL puromycin (Sigma-Aldrich, P8833). HEK293T/17 cells were provided by Dr. Lynda Chin, MD Anderson Cancer Center, and maintained in DMEM medium supplemented with 10% heat-inactivated FBS (Gemini), streptomycin (100 μg/mL) solution (Corning, VWR45000–652) and penicillin (100 U/mL) at 37°C and 5% CO[2] as described before^[352]41. Mouse kidney endothelial cells (MKECs) and NIH-3T3 cells were kindly provided by Dr. Isaiah J. Fidler, MD Anderson Cancer Center. MKECs were evaluated for microscopic morphology without further characterization, and they were maintained in endothelial cell medium (Cell Biologics, M1168) together with the endothelial cell medium supplement kit (Cell Biologics, M1168-Kit). NIH-3T3 cells were maintained in DMEM medium supplemented with 10% heat-inactivated FBS (Gemini), penicillin (100 U/mL) and streptomycin (100 μg/mL) solution (Corning, VWR45000–652). C57BL/6 mouse primary lung microvascular endothelial cells (Cell Biologics, C57–6011) and C57BL/6 mouse primary brain microvascular endothelial cells (Cell Biologics, C57–6023) were maintained in complete mouse endothelial cell medium (Cell Biologics, M1168) on gelatin (Cell Biologics, 6950) coated culture dishes. The cells were incubated in 0.25% FBS for 24h before treatment with murine recombinant Ang2 (rAng2, R&D Systems, 7186-AN-025, 100 ng/mL) or PBS (control) for the indicated timepoints (6h, 12h, and 24h) and harvested for RNA isolation based on previous studies ^[353]47. For the primary lung microvascular endothelial cells, the cells were treated with rAng2 (100 ng/mL) or PBS for different timepoints before starvation in 0.25% FBS for 24h and harvested into lysis buffer for western blot. HEK293T/17 cells were validated by STR analysis at the UT MD Anderson Cytogenetics and Cell Line Authentication Core. 4T1 and NIH-3T3 cells identity were validated via CellCheck (IDEXX BioResearch). All the cells were routinely screened to confirm the absence of mycoplasma contamination. C57BL/6 mouse primary lung microvascular endothelial cells (Cell Biologics, C57–6011) were found positive for mycoplasma and were treated, and subsequently found negative for mycoplasma prior to use in experiments. Virus For the overexpression of Cldn5, Ad-CMV-CLDN5v2 with His tag (Vigene Biosciences, Catalog number VH809267, Lot 100001) and Adeno-CMV-luc control (Vigene Biosciences, Catalog number 1000, Lot A1022t#6M13) were purchased from the listed vendor. For the downregulation of Cldn5 gene expression, AAV9-mCldn5-o4-shRNA (Vector Biolab, custom production, shAAV-180716#17) and AAV9-anti-Luc-Control-shRNA (Vector Biolab, custom production, shAAV-Lot180716#18) were purchased from the listed vendor. For the overexpression of Cldn5, NIH-3T3 (5 × 10^4) cells were transduced with 10 μL (1 × 10^7 PFU/μL, total 1× 10^8 PFU) Ad-CMV-CLDN5v2 or 10 μL (1 × 10^7 PFU/μL, total 1 × 10^8 PFU) Adeno-CMV-luc for 48 hours followed by RNA isolation and protein extraction. Control parental cells were not transduced. For the downregulation of Cldn5, NIH-3T3 cells were transduced with AAV9-mCldn5-shRNA. NIH-3T3 (5 × 10^4) cells were seeded in complete medium with or without doxycycline (1 μg/mL), parental cells without any treatment were used as control. After 24 hours, the cells were transduced with 5 μL (1.1 × 10^10 genome copies (GC)/μL, with 10-fold dilution, total 5.5 × 10^9 GC) AAV9-mCldn5-o4-shRNA or 5 μL (1.1 × 10^10 GC/μL, with 10-fold dilution, total 5.5 × 10^9 GC) AAV9-anti-Luc-Control-shRNA. RNA was isolated 72 hours after the transduction. To downregulate Cldn2 and Cldn10 gene expression using CRISPR-Cas9, Cldn2 or Cldn10 targeting sgRNAs were designed using an online tool ([354]http://crispr.dfci.harvard.edu/SSC/). The sequences used are shown in [355]Supplementary Table 5. LV/Cas9-Cldn2-sgRNA1,2 and LV/Cas9-Cldn10-sgRNA1,2,3 were generated by employing BsmBI to insert each sgRNA into the RNA scaffold of lentiCRISPRv2 vector (Addgene #52961). The generation of lentiviruses and concentration measurements were performed as previously described ^[356]41. The titration of lentivirus was determined by utilizing the Lenti-X qRT-PCR Titration Kit (LV900, Applied Biological Materials Inc) in accordance with manufacturer’s instructions. For the transduction in vitro, mouse kidney endothelial cells (MKEC) were maintained in endothelial cell medium (Cell Biologics, M1168) and incubated with 5 μg/mL polybrene (EMD Millipore TR-1003-G, CA, US) and 5 μL (2 × 10^8 IU/μL) viral solution together with for 60 hours. Fresh RPMI medium supplemented with puromycin (1 μg/mL) was replaced every two days post-transduction before the establishment of LV/Cas9-Cldn2-sgRNA1,2-MKEC and LV/Cas9-Cldn10-sgRNA1,2,3-MKEC stable cell lines. After 5 to 7 days, RNA was isolated for subsequent quantitative real-time PCR analyses (described in a separate section below). Mice Source data is listed for all tumor experiments in the associated source data file for each Main and Extended Data Figure in this study. For orthotopic implantation, BALB/c virgin female mice (Charles River, strain code 028) between the ages of 6 to 12 weeks were inoculated with 4T1 cells in each mammary pad (5 × 10^5 4T1 cells in the right or left mammary fat pad). Tumor burden was assessed using Vernier calipers and volumes were calculated as previously described ^[357]13. Surface lung nodules were counted at autopsy. When the sum of averaged tumors on each flank reached 500 mm^3, fibrotic insults were induced in the lung via intratracheal administration of bleomycin (25 μL of 0.15U in saline solution, Sigma-Aldrich, B5507–15), and in the kidney via surgical manipulation (unilateral ureteral obstruction surgery, UUO) ^[358]54. Full-thickness cutaneous wound surgery was performed as previously described ^[359]55. For the cancer cell colonization studies, 5 × 10^5 4T1 cells were injected intravenously (4T1 i.v.) into the retro-orbital venous sinus of BALB/c virgin female mice aged between 6 to 12 weeks. 4T1 i.v. injection was performed on the same day as the intratracheal bleomycin, UUO surgery, and cutaneous wound surgery. MMTV-PyMT (Pymt) transgenic mice backcrossed on the BALB/c background spontaneously develop tumors in their mammary fat pads between the ages of 8 to 12 weeks ^[360]56. Fibrotic events were induced in the lung and kidney when the cumulative tumor volumes reached 500 mm^3, and the cutaneous wounding was generated as described above. For both orthotopic implantation and MMTV-PyMT transgenic mice, all the mice were euthanized before or upon reaching a tumor volume of approximately 2,000 mm^3, as permitted by the Institutional Animal Care and Use Committee of MDACC (with a goal of a maximum of 2,000 mm^3 using regular evaluation of tumor size with calipers and prompt euthanasia if volume measurement were found to exceed this limit). Following i.v. cancer cell injection, the mice were euthanized on day 10 after lung and kidney fibrosis induction, and on day 14 after wound surgery. For the ultrasound-guided intracardiac injection, BALB/c virgin female mice aged between 6 to 12 weeks were separated into 2 groups receiving either UUO or sham surgery (Day 0). The next day, 4T1 cancer cells (5 × 10^5) were injected into the left cardiac ventricle via using ultrasound machine (VisualSonics Vevo-2100, Day 1). All the mice were euthanized after 10 days (Day 10). 4T1 (5 × 10^5) cells were injected under the renal capsule of BALB/c virgin female mice aged between 6 to 12 weeks. The mice were separated into 2 cohorts receiving either UUO or sham surgery to the injected kidney. All the mice were euthanized after 10 days. The mice were euthanized before or at early signs of moribundity. In some experiments, mice received recombinant murine Ang2 (100 μg/mL in 100 μL PBS) or PBS (100 μL) control intraperitoneally on Day 1 and Day 3 (2 times) and subsequently euthanized on Day 4 (24h after the last rAng2 administration). In mice with 4T1 i.v., rAng2 or PBS was intraperitoneally injected one day before and one day after i.v. injection of 4T1 cells. In experiments in which Ang2 was neutralized, kidney fibrotic events were induced by UUO when mice presented with an averaged tumor burden of 500 mm^3 and mice were treated with control isotype-matched IgG (10 mg/kg BW in 200 μL PBS) or anti-Ang2 neutralizing antibody (murinized LC06, Roche Diagnostics, 10 mg/kg BW in 200 μL PBS) once a week (2 times) as previously reported ^[361]25. Overexpression of Claudin 5 was induced in the lung via intratracheal administration of 20 μL (1 × 10^7 PFU/μL) Ad-CMV-CLDN5v2 and 20 μL (1 × 10^7 PFU/μL) Adeno-CMV-luc. After 24 hours (day 1), the mice were treated with intraperitoneal injection of rAng2 (as described above) or PBS. On day 3, the mice were injected i.v. with 4T1 cells (5 × 10^5) cells. On day 4, the mice were treated once more with rAng2 or PBS. Down-regulation of Claudin 5 in the 4T1 orthotopic model was achieved by intratracheal administration of AAV9-mCldn5-o4-shRNA. On day 1, the mice were implanted orthotopically with 4T1 cells (5 × 10^5) cells. After 10 days, the mice were intratracheally administered 20 μL (1.1 × 10^10 GC/μL) AAV9-mCldn5-o4-shRNA or 20 μL (1.1 × 10^10 GC/μL) AAV9-anti-Luc-Control-shRNA. Doxycycline (0.2% doxycycline with 4% sucrose in water) was administered from the day of tumor implantation until the mice were euthanized. For the knockdown of Claudin 2 and Claudin 10 in vivo, LV/Cas9-Cldn2-sgRNA1, 2 and LV/Cas9-Cldn10-sgRNA1, 2, 3 virus solutions (4 μL each, with a concentration ranging from 3 × 10^8 to 4 × 10^9 IU/μL) were combined in 20 μL (for a final combined concentration of approximately 3 × 10^9 IU/μL in 20 μL) and injected into the renal cortex. LV/Cas9-control sgRNA (3 × 10^9 IU/μL in 20 μL) was used as control. After 26 days, the kidneys of some of the mice were harvested for analyses. Other mice were subsequently implanted orthotopically with 4T1 cells (5 × 10^5) as described above. Some of the mice were also subjected to UUO or sham surgery 10 days after orthotopic cancer cells inoculation. Although rare, mice were excluded from the experimental cohort if UUO was found to have failed (no fibrosis due to failed ligation of the ureter). To assess vascular permeability, mice were administered with FITC-conjugated 70 kDa dextran via retro-orbital venous sinus injection 5 min prior to euthanasia. All the mice involved in the study were maintained under standard housing conditions at UT MD Anderson Cancer Center (MDACC) animal facilities: mice were housed in individually ventilated cages with a 12h light/12 h dark cycle (06:00/18:00), a temperature range of at 21°C to 23°C and humidity levels between 40 to 60%. The procedures conducted on mice were reviewed and approved by Institutional Animal Care and Use Committee of MDACC. Histology Formalin-fixed mouse lung, liver, kidney and other tissues (5 μm) were cut for different staining, including hematoxylin and eosin (H&E), Picrosirius red, and Masson Trichrome staining (MTS, using Gomori’s Trichrome Stain Kit (Leica Biosystems, 38016SS2) or Trichrome, Masson, Aniline Blue Stain Kit (Newcomer Supply, 9179B). Images of an entire lung lobe(s) or sagittal kidney sections were obtained using the Panoramic 250 Flash III slide scanner or ZEISS Axio Scan.Z1 AxioScan. Metastatic area is defined as the metastatic disease burden in the lung of a mouse with a primary tumor. Colonization area refers to the cancer disease burden in the lung of a mouse following a systemic injection of cancer cells. Lung metastasis or colonization were determined through histopathological analysis, and the metastatic area was quantified as a percentage of total lung area via using Panoramic Viewer software or Image J. One or two lung H&E sections per mouse was evaluated, as detailed in the source data. Liver metastases were identified using histopathological analysis from H&E staining and liver metastatic burden was quantified by counting the average number of nodules from randomly selected visual fields images captured by Leica DM 1000 LED microscope equipped with a DFC295 microscope camera (Leica) with LAS version 4.4 software (Leica). The kidney tumor burden was measured using histopathological analysis of H&E staining and the tumor area was quantified using Panoramic Viewer software and Image J software. The tumor area was measured as a percentage of the total kidney area. Pleural thickness was measured at 8 random points at the circumference of the lung lobe by H&E staining. Pulmonary edema was identified from H&E staining in both septal and alveolar area, characterized by noticeable interstitial and intra-alveolar fluid accumulation throughout the lungs as previously reported ^[362]57. Edema area was quantified by determining the percentage of total lung area affected using Panoramic Viewer software. Immunofluorescence and Immunohistochemistry Kidney tissues were fixed in 4% paraformaldehyde at 4°C for overnight. After fixation, they were transferred to 30% sucrose solution for 24 hours before being embedding in OCT compound. Deparaffinized and rehydrated FFPE sections (5 μm) followed antigen retrieval with heated citric acid buffer (pH 6.0) or TE buffer (pH 9.0) for 15 minutes at 98°C, were blocked with blocking solution (1% BSA in PBS) for 1 hour before the incubation with different primary antibodies (as detailed in the Nature Research Reporting Summary linked to this article) overnight at 4°C. Samples were incubated with conjugated secondary antibodies (as detailed in the Nature Research Reporting Summary linked to this article), followed by DAPI (Vectashield, H1200) or Hoechst 33342 (Invitrogen, H3570, 1:2,000) staining. Random visual fields were selected, and representative pictures were acquired with Zeiss Axio Observer.Z1 or LSM800 confocal laser scanning microscope using ZEN software (Zeiss). We evaluated the entire kidney tissue for GFP signal and analyzed kidneys from all the mice in the study and did not observe any GFP positive cells in kidneys. The percentage of Claudin 5 CD31 double positive area out of total CD31 positive area were also quantified by ImageJ software. Randomly selected visual fields images were captured and the quantification was performed on 200x or 400x images, as listed in the figure legends. Vascular leakage assay was performed and quantified as previously described ^[363]25. Briefly, OCT-embedded tissues were stained with CD31 (Dianova, Dia310M, 1:40) before the incubation with Alexa Fluor 647 anti-rat antibody (Invitrogen A21472, 1:1000). Representative images were captured via Zeiss Axio Observer.Z1 and quantified as the total FITC positive field normalized to CD31 field by Image J. For the visualization of Claudin 5 expression in lung endothelial cells, cells were cultured on gelatin-coated 8-well chambers (Thermo Scientific^™ 154534) for overnight. The cells were then starved in 0.25% FBS for 24 hours prior to the treatment with rAng2 (100 ng/mL) or PBS (control) for 24 hours. Afterwards, cells were fixed with 100% methanol for 5 minutes and permeabilized with PBST (+0.5% Triton X-100/PBS) for an additional 5 minutes. Subsequently, the cells were incubated with blocking buffer (5% BSA in PBS+0.5% Triton X-100/PBS) for 1 hour at RT before overnight incubation with Claudin 5 antibody (Abcam 131259, 1:100) at 4°C. Following the primary antibody incubation, the cells were treated with Anti-Rabbit 488 secondary antibody (Jackson ImmnoResearch, 111546047, 1:200) before staining with Hoechst 33342 (Invitrogen, H3570, 1:2,000) for 10 minutes. The media chamber was carefully removed, and representative pictures were acquired with LSM800 confocal laser scanning microscope using ZEN software (Zeiss). For the immunohistochemistry staining of CD31, it was performed as described previously ^[364]41. FFPE mouse tumor tissues (5 μm) were processed before antigen retrieval with heated citric acid buffer (pH 6.0) for 15 minutes at 98°C. After incubation with 3% H[2]O[2] in PBS for 15 minutes, the samples were blocked with 1% BSA in PBST before incubation with CD31 (Dianova, Dia310M, 1:40) antibody overnight at 4°C. Then the samples were incubated with 4plus Biotinylated Goat Anti-Rat IgG (Biocare GR607H) for 30 minutes and a subsequent 30-minute incubation with 4plus Streptavidin HRP Label (Biocare HP604H) at room temperature. Subsequently, the slides were stained with DAB (Life Technologies 34065) and counterstained with hematoxylin before being cover-slipped. Randomly selected visual fields images (200X) imaged with the Leica DM 1000 LED microscope were captured and quantified with ImageJ (NIH, Bethesda, MD). Microarray analysis Total RNA was extracted from healthy kidney (n=3 mice), fibrotic kidney (n=2 mice), healthy lungs (n=3 mice), fibrotic lungs (n=3 mice), and lungs from mice with fibrotic kidneys (n=3 mice) using TRIzol reagent (Life Technologies, 15596026) according to the manufacturer’s instructions. An equal amount of RNA was submitted from each cohort to the Advanced Technology Genomics Core at MD Anderson. The microarray data was deposited into Gene Expression Omnibus ([365]GSE199178). Gene expression analysis was performed using Mouse Ref6 Gene Expression BeadChip (Illumina), and the Limma package (version 3.44.3) from R (version 4.0.2) was used to identify differentially expressed genes for each comparison (p<0.05, abs (fold change) >=1.5). Pathway enrichment analysis was conducted by WebGestalt 2019 ([366]http://www.webgestalt.org/) ^[367]58. Benjamini and Hochberg’s procedure was used for multiple test correction ^[368]59. Significant pathways were defined as those with an FDR< 0.05. Quantitative real-time PCR analyses RNA from cells were extracted using TRIzol reagent (Life Technologies 15596026) according to the manufacturer’s instructions. cDNA was synthetized using high-capacity cDNA reverse transcription kit (Applied Biosystems 4368814). Gene expression was determined using Power SYBR Green PCR Master Mix (Applied Biosystems) on QuantStudio^™ 7 Flex Real-Time PCR System (ThermoFisher Scientific). Primer sequences for the target genes are shown in [369]Supplementary Table 6. Gapdh, Actb or Cd31 were used to normalize the data. The relative mRNA expression is presented as fold change (2^-ΔΔCt), normalizing the control to 1, or presented as 2^-ΔCt. Statistical analyses were performed on the ΔCt values. Western blot analysis NIH3T3 cells were transduced with Ad-CMV-CLDN5v2 (with His tag) or Adeno-CMV-luc for 48 hours. The cells were lysed with RIPA buffer (Thermo Scientific, 89900) with EDTA free protease inhibitor (Roche, 4693159001) and the lysates were resolved on 4–12% Bis-Tris protein gels (Thermo Fisher Scientific, NP0321PK2) as previously described^[370]60. Control parental cells without any treatment were used. The proteins were transferred with a semidry trans-blot turbo transfer system (Bio-Rad, 1704150) into methanol activated PVDF membrane (Bio-Rad 1704272). The membranes were incubated with 5 % non-fat milk (LabScientific M0841) before incubation with different primary antibodies (as detailed in the Nature Research Reporting Summary linked to this article) overnight at 4°C. The membranes were washed and incubated with mouse (R&D system, HAF007, 1:2000) or Rabbit IgG (Abcam, ab16284, 1:2000) horseradish peroxidase-conjugated secondary antibody for 1 hour at room temperature. Amersham^™ Hyperfilm^™ ECL (GE Healthcare, 28906835) and West-Q Pico ECL Solution (GenDEPOT, W3652020) were utilized for protein detection as described previously ^[371]41. The band density from different independent experiments were quantified using ImageJ as previously described ^[372]41, and statistical analyses were performed using difference in intensity between β-actin and Claudin 5 on three distinct experiments. For the evaluation of Claudin 5 protein levels in lung microvascular endothelial cells treated with rAng2, the cells were lysed with RIPA buffer and the lysates were resolved as described above. For the evaluation of Foxo1, the cells were lysed with subcellular protein fractionation kit for cultured cells (Thermo Scientific^™ 78840). The cytoplasmic and nuclear protein fractions were isolated based on manufacturer’s instructions and the lysates were separated on 4–12% Bis-Tris protein gels (Thermo Fisher Scientific, NP0321PK2) before transfer onto a PVDF membrane (Bio-Rad 1704272). The membranes were incubated with 5 % non-fat milk (LabScientific M0841) before incubation with different primary antibodies (as detailed in the Nature Research Reporting Summary linked to this article) overnight at 4°C. The membranes were washed and incubated with Rabbit IgG (Abcam, ab16284, 1:2000) horseradish peroxidase-conjugated secondary antibody for 1 hour at room temperature and proteins were detected as described above. β-actin served as the internal control for data normalization. Relative protein expression is reported as fold change, with the control set to 1. Statistical analyses were conducted on the ratio of target protein band intensity relative to β-actin band intensity. Uncropped blots are shown in the Image Source Data File. Cytokine profiling and Ang2 ELISA Blood was collected at the time of euthanasia and centrifuged at 300g for 10 minutes. The serum was extracted and stored at −80°C for further use. Cytokine profiling was performed using the Mouse XL Cytokine Array Kit (R&D, ARY028) based on the manufacturer’s instructions. Cytokine array scans are shown in the Image Source Data File.Ang2 protein levels in serum samples were quantified by Angiopoietin 2 Mouse ELISA kit (Abcam, ab171335) according to the manufacturer’s protocol. Flow cytometry Fresh kidneys were harvested and minced into small pieces prior to digestion with collagenase IV (Gibco, 17104019, 4 mg/mL) and dispase II (Gibco, 17105041, 4 mg/mL) in RPMI medium for 1 hour at 37°C. The mixture was then filtered through a cell strainer (70 μm) and the collected cells were resuspended in PBS/2% FBS. Afterwards, the cells were stained with anti-CD31-PE (BioLegend, 102508, 1:100) and anti-E-cadherin-eFluor^™ 660 (eBioscience, 50–3249-82, 1:100) in PBS/2% FBS on ice for 30 min. Samples were filtered through a 40 μm mesh and sorted with BD FACS AriaTM II sorter (South Campus Flow Cytometry Core Lab of MD Anderson Cancer Center). Singlets were then gated for CD31 and E-cadherin positivity, with endothelial cells captured as CD31^+ E-cadherin^- and epithelial cells captured as CD31^- E-cadherin^+. Cells were collected in RPMI medium with PBS/10% FBS before RNA isolation. For the evaluation of immune cells infiltration in the lung and kidney after induction of fibrosis, analysis was performed as previously reported ^[373]54. Lung fibrosis was induced via intratracheal administration of bleomycin, and kidney fibrosis was induced via UUO as described above. Lung and kidney were minced and digested in 10 mL Liberase TL (Roche 05401020001, 0.1 mg/mL) and DNase I (Roche, 10104159001, 0.2 mg/mL) in RPMI-1640 medium for 30 minutes at 37°C. The tissue lysate was filtered through cell strainer (100 μm) and washed with PBS/2% FBS buffer prior to immunostaining. The resulting single cell suspension was incubated with different antibodies (as detailed in the Nature Research Reporting Summary linked to this article) on ice for 30 minutes in dark. For the staining of intracellular marker, cells were fixed and permeabilized using the Foxp3/Transcription Factor Staining Buffer Set (eBioscience 00552300) according to manufacturer’s instructions before staining with anti-Foxp3-Alexa Fluor^™700 (eBioscience 56577382, 1:50) in permeabilization buffer. Data were acquired on BD LSR Fortessa X-20 Cell Analyzer (BD Biosciences) and the CD45^+ cells were analyzed for the expression of the different immune populations using FlowJo. Detection of circulating cancer cells To detect circulating tumor cells in lung and kidney, 5 × 10^5 4T1 cells were injected intravenously (4T1 i.v.) into the retro-orbital venous sinus of BALB/c virgin female mice. After 10 days, mice were euthanized with CO[2] and a perfusion with 150 mL of PBS was conducted via insertion into the left ventricle of the heart. Subsequently, fresh lung and kidney tissues were collected for flow cytometry analysis and genomic DNA isolation. For flow cytometry, fresh lung and kidneys were minced into small pieces prior to digestion with collagenase IV (Gibco, 17104019, 4 mg/mL) and dispase II (Gibco, 17105041, 4 mg/mL) in RPMI-1640 medium at 37°C for 1h, followed by treatment with DNase I (Roche 10104159001, 0.2 mg/mL). After the filtration of the cells via cell strainer (70 μm), the cells were resuspended in PBS/2% FBS and 10mM EDTA before the filtration through a 40 μm mesh before the centrifugation at 500g for 5 minutes. Cells were then incubated with 3 mL ACK lysis buffer (Gibco A1049201) at RT for red cell lysis and wash with PBS/2% FBS and 10mM EDTA for twice before centrifugation at 500g for 5 minutes. Then the cells were stained with eFluor^™ 780 live/dead fixable viability dye (eBioscience 65086514, 1:1000) on ice for 10 minutes before the resuspension with PBS/2% FBS in PBS. Flow cytometry analysis for GFP^+ cancer cells was performed using BD LSR Fortessa X-20 Cell Analyzer (BD Biosciences). The data were recorded as the percentage of GFP^+ cells per 10^6 total cells. For genomic DNA isolation, DNA was isolated from lung and kidney tissues using the DNeasy Blood & Tissue Kits (Qiagen, 69506), 4T1-luc-GFP cells derived DNA was served as the positive control, and fresh lung and kidney tissues from WT mice were served as the negative control. For Q-PCR analysis of GFP, the DNA was amplified using Power SYBR Green PCR Master Mix (Applied Biosystems) on QuantStudioTM 7 Flex Real-Time PCR System (ThermoFisher Scientific). Neurofibromatosis type 1 (Nf1) was used to normalize the data and presented as 2^-ΔCt. Single cell RNA sequencing analysis The raw counts and metadata of scRNA-seq of murine endothelial cells ^[374]34, murine lung tissue ^[375]35, human lung tissue ^[376]35, murine kidney tissue ^[377]36, and human kidney tissue ^[378]37, murine brain tissue ^[379]34 and human brain tissue ^[380]61 were obtained from the database as described in the original studies. The data was processed using R (version 4.0.0) package Seurat (version 4.0.3). Principal component analysis (PCA) was applied to perform dimensionality reduction on the expression matrices. The ‘FindNeighbors’ function identified nearest neighbors among cells within the PCA space, while the ‘FindClusters’ function grouped cells using the Louvain algorithm. Visualization of the clusters resulting from t-Distributed Stochastic Neighbor Embedding (tSNE) dimension reduction was achieved through the ‘RunTSNE’ function. The cell type annotations were adopted from the original studies to label the clusters for visualization purpose. ‘DimPlot’ function was used to show the tSNE plots of cell clusters. ‘VlnPlot’ function was used for displaying the expression of genes in different tissues. ‘FeaturePlot’ was employed to display the expression of genes in cell clusters. RNA sequencing analysis The raw reads from bulk RNA sequencing, capturing transcriptome data from breast cancer samples with lung metastasis and corresponding normal lung tissues, and were obtained from the database as described in the original studies ([381]GSE193103)^[382]62. The data was processed using R software (version 4.2.2) along with the DESeq2 package (version 1.38.3) for data normalization and differential gene expression analysis. Statistics & Reproducibility No statistical methods were used to pre-determine sample sizes but our sample sizes are similar to those reported in previous publications^[383]13,[384]16,[385]25,[386]54,[387]55. Mice and samples were randomized prior to group allocations whenever applicable. No data were excluded from the analyses with the following rare exceptions: 1) mice in which UUO was found to have failed and 2) sample processing for flow cytometry failed quality control assessment. Data collection and analysis were not performed blind to the conditions of the experiments. Data are represented as mean ± SEM and statistical analysis was conducted using GraphPad Software. Detailed information on statistical tests used and definition of P values are listed in the Figure Legends. Extended Data Extended Data Fig. 1: Lung and kidney fibrosis induction. Extended Data Fig. 1: [388]Open in a new tab (a) Representative H&E staining, Sirius Red staining, and αSMA immunolabeling of the lung and kidneys of mice with and without fibrosis in the respective organs. Scale bar, 100 μm. (b) Quantification of positive Sirius Red and αSMA^+ stained area in the indicated organs and groups, n = 5 to 6 mice per group. (c, d) Relative Fn1 and Col1a1 mRNA levels in the respective organs and groups, n = 3 mice per group. KF: kidney fibrosis; LF: lung fibrosis. (e) Representative images for GFP immunolabeling and quantification of percent of the GFP positive area in lung with or without lung fibrosis. Scale bar, 50 μm, n = 6 mice per group. (f) Representative images for GFP immunolabeling of tumor in the mice with or without lung fibrosis. n = 1 mouse per group. Scale bar, 25 μm. (g) Representative liver H&E images in 4T1 tumor orthotopic bearing mice after induction of lung fibrosis. Scale bar, 50 μm. Quantification of metastatic liver nodules in the indicated groups, n = 6 mice per group. (h) Representative images for GFP immunolabeling and quantification of percent of the GFP positive area in liver of mice with or without lung fibrosis. Scale bar, 50 μm, n = 6 mice per group. (i, j) Representative H&E images of heart (i) and intestine (j) in 4T1 tumor orthotopic bearing mice after induction of lung fibrosis. Scale bar, 50 μm. (k) Orthotopic 4T1 tumor volume over time in mice with or without kidney fibrosis, n = 6 mice per group. (l, m) Representative H&E images (l) of the lung of 4T1 orthotopic tumor bearing mice with or without kidney fibrosis and respective quantification (m) of metastatic area. Scale bar, 25 μm, n = 6 mice per group. (n) Representative images for GFP immunolabeling and quantification of percent of the GFP positive area in lung with or without kidney fibrosis. Scale bar, 50 μm, n = 6 mice per group. (o) Representative images for GFP immunolabeling for tumor in the mice with or without kidney fibrosis. Scale bar, 25 μm. (p) Representative liver H&E images in 4T1 tumor orthotopic bearing mice after induction of kidney fibrosis. Scale bar, 50 μm. Quantification of metastatic liver nodules in the indicated groups, n = 6 mice per group. (q) Representative images for GFP immunolabeling and quantification of percent of the GFP positive area in liver of mice with or without kidney fibrosis. Scale bar, 50 μm, n = 6 mice per group. (r) Representative H&E images of heart and intestine of 4T1 tumor orthotopic bearing mice after induction of kidney fibrosis. Scale bar, 50 μm. KF: kidney fibrosis; LF: lung fibrosis. Data are presented as mean values +/− SEM. For k: two-way ANOVA with Sidak’s multiple comparisons test. For all other panels: Unpaired two-tailed t test with Welch’s correction applied for unequal variances (determined by F-test). P values are listed, ns: not significant. Extended Data Fig. 2. Fibrosis does not promote non-tropic metastasis. Extended Data Fig. 2 [389]Open in a new tab (a) Representative H&E images of the kidneys of 4T1 orthotopic tumor bearing mice +/− lung or kidney fibrosis. Scale bar, 25 μm. (b) Number of mice with microscopic kidney tumors in the indicated groups (c) Representative images for GFP immunolabeling and quantification of percent of the GFP positive area in kidney +/− kidney fibrosis. Scale bar, 50 μm. (d) Number of mice with microscopic kidney tumors in the indicated groups. (e) Representative kidney H&E images in MMTV-PyMT transgenic mouse model with lung or kidney fibrosis. Scale bar, 25 μm. (f) Number of mice with microscopic kidney tumors in the indicated groups. (g) Representative immunofluorescence images of PyMT positive cancer area in tumor, lung, and kidney after kidney fibrosis in MMTV-PyMT transgenic mice. Scale bar, 25 μm. (h–j) Representative images and quantification of positive MTS and Sirius Red staining from the indicated groups. Scale bar, 25 μm, n = 6–7 mice per group. Data are presented as mean values +/− SEM. Unpaired two-tailed t test, with Welch’s correction applied for unequal variances (determined by F-test). P values are listed, ns: not significant. Extended Data Fig. 3. Intracardiac cancer cell injection does not alter metastatic disease in renal fibrosis & Wound healing does not impact tropic and non-tropic site metastasis. Extended Data Fig. 3 [390]Open in a new tab (a) Representative GFP and CD31 staining of the kidneys from mice with 4T1 orthotopic inoculation and 4T1 i.v. administration with or without kidney fibrosis. Scale bar, 25 μm. (b) Quantification of GFP^+ cancer cells from lung after 4T1 i.v. administration for 10 days with or without perfusion by flow cytometry. n = 3 mice per group. Gating strategy shown in [391]Supplementary Fig. 1A. (c) Quantification of GFP^+ cancer cells from kidneys after 4T1 i.v. administration for 10 days with or without perfusion by flow cytometry, n = 3 mice per group. Gating strategy shown in [392]Supplementary Fig. 1B. (d, e) QPCR quantification of GFP DNA (expressed 2^-ΔCt after normalization to Nf1) from the lung (d) and kidney (e) of mice after 4T1 i.v. administration for 10 days with or without perfusion, n = 3 mice per group. (f) Representative H&E images and quantification of lung after intracardiac injection of 4T1 cancer cells together with induction of kidney fibrosis. Control, n = 5 mice; kidney fibrosis, n = 9 mice, Scale bar, 25 μm. (g) Representative H&E images and quantification of metastatic liver nodules after intracardiac injection of 4T1 cancer cells together with induction of kidney fibrosis. Control, n = 5 mice; kidney fibrosis, n = 9 mice. Scale bar, 50 μm. (h) Representative H&E images and number of mice with microscopic kidney tumors in the indicated groups after Intracardiac injection of 4T1 cancer cells together with induction of kidney fibrosis. Scale bar, 25 μm. (i) Representative images for GFP immunolabeling and number of mice with microscopic kidney tumors in the indicated groups after Intracardiac injection of 4T1 cancer cells together with induction of kidney fibrosis. Scale bar, 50 μm. (j) Orthotopic 4T1 tumor volume over time +/− cutaneous wound, n = 6 mice per group. (k) Representative H&E images of the lung of 4T1 orthotopic tumor bearing mice +/− cutaneous wound. Scale bar, 25 μm. (l) Quantification of metastatic area and surface lung nodules in 4T1 orthotopic tumor bearing mice with and without cutaneous wounds, n = 6 mice per group. (m) Tumor volume over time in MMTV-PyMT mice +/− cutaneous wound, n = 5 to 6 mice per group. (n) Representative lung H&E images from in MMTV-PyMT mice +/− cutaneous wound. Scale bar, 25 μm. (o) Quantification of metastatic area and surface lung nodules in MMTV-PyMT mice +/− cutaneous, n = 5 to 6 mice per group. (p) Wound closure rate in 4T1 orthotopic tumor bearing mice (n = 10 mice) and no tumor control (n = 4 mice). (q) Representative lung H&E images from mice with 4T1 i.v. injection and +/− cutaneous wound. Scale bar, 25 μm. (r) Quantification of metastatic area and surface lung nodules in mice with 4T1 i.v. injection +/− cutaneous wound, n = 5 mice per group. Data are presented as mean values +/− SEM or individual values shown. For j, m: two-way ANOVA with Sidak’s multiple comparisons test. For p: mixed effect model with Sidak’s multiple comparison test. For other panels: Unpaired two-tailed t test, with Welch’s correction applied for unequal variances (determined by F-test). P values are listed, ns: not significant. Extended Data Fig. 4. Global gene expression profiling in fibrosis and serum cytokine analysis. [393]Extended Data Fig. 4 [394]Open in a new tab (a, b). Volcano plot and gene ontology (GO) enrichment ratio and false discovery rate (FDR) values associated with overlapped up-regulated and down-regulated genes. (c, d) GO enrichment ratio and FDR values in the lungs of mice with and without lung fibrosis (c) or with and without kidney fibrosis (d). (e, f) Overlapping up-regulated (e) or down regulated (f) genes in the lungs of healthy mice vs. mice with the lung fibrosis, and in the lungs of healthy mice vs mice with kidney fibrosis, with overlapped genes involved in vascular (f) and immune remodeling (e) pathways. (g) Cytokines profile of serum samples from healthy mice, mice with cutaneous wound, mice with lung fibrosis and kidney fibrosis, and heat map of the averaged relative cytokine levels in the indicated groups after normalization to healthy group (set to 1). Healthy, n = 2 mice; wound, n = 2 mice; lung fibrosis (LF), n = 3 mice; kidney fibrosis (KF), n = 3 mice. Cytokines showing minimal changes between healthy and wound (wound/healthy (W/H) ratio between 0.9 and 1.1) were then ranked based on levels in kidney fibrosis. Extended Data Fig. 5. Immune cells infiltration in lung and kidney after fibrosis induction. [395]Extended Data Fig. 5 [396]Open in a new tab (a) Percentages of CD45^+, CD3^+, CD4^+FoxP3^− effector T (Teff) cells, CD8^+, γδ^+, CD4^+FoxP3^+ regulatory T (Treg) cells, NK1.1^+ cells, CD11c^+, CD19^+, CD11b^+, CD11b^+Ly6C^+Ly6G^−and CD11b^+Ly6G^+ Ly6C^− cells in lungs of mice with lung fibrosis or kidney fibrosis. Healthy lung, n = 6 mice; lung fibrosis (LF), n = 5 mice; healthy kidney, n = 6 mice; kidney fibrosis (KF), n = 5 mice. (b) Percentages of CD45^+, CD3^+, CD4^+FoxP3^− effector T (Teff) cells, CD8^+, γδ^+, CD4^+FoxP3^+ regulatory T (Treg) cells, NK1.1^+ cells, CD11c^+, CD19^+, CD11b^+, CD11b^+Ly6C^+Ly6G^−and CD11b^+Ly6G^+Ly6C^− cells in kidneys of mice with lung fibrosis or kidney fibrosis. Healthy lung, n = 5 mice; lung fibrosis (LF), n = 6 mice; healthy kidney, n = 6 mice; kidney fibrosis (KF), n = 4 mice. (c) Colocalization of FITC-dextran (green) and CD31 (red) in the liver of mice with and without after rAng2 treatment. Scale bar, 20 μm. Data are presented as mean values +/− SEM. Unpaired two-tailed t test, with Welch’s correction applied for unequal variances (determined by F-test). P values are listed, ns: not significant. Extended Data Fig. 6. Recombinant Ang 2 enhances metastasis to the lung but not kidney. Extended Data Fig. 6 [397]Open in a new tab (a) Representative lung H&E of mice treated with PBS (control) or recombinant Ang2 (rAng2). Scale bar, 25 μm. (b, c) Representative lung H&E of mice after induction of lung (b) or kidney fibrosis (c). Scale bar, 25 μm. (d) Quantification of edema area per whole lung area, n = 5 to 6 mice per group. (e) Quantification of pleural thickening of lung, n = 5 to 6 mice per group. (f) Representative kidney H&E images of mice treated with PBS (control) or rAng2. Scale bar, 25 μm. (g, h) Representative kidney H&E images of mice after induction of lung (g) or kidney fibrosis (h). Scale bar, 25 μm. (i) Representative lung H&E of mice treated with PBS (control) or recombinant Ang2 (rAng2) with 4T1 cells i.v. injection. Scale bar, 25 μm. (j) Representative kidney H&E of mice treated with PBS (control) or recombinant Ang2 (rAng2) with 4T1 cells i.v. injection. Scale bar, 25 μm. Data are presented as mean values +/− SEM. Unpaired two-tailed t test, with Welch’s correction applied for unequal variances (determined by F-test). P values are listed. Extended Data Fig. 7. tSNE plots of Claudin genes in scRNA-seq data sets. [398]Extended Data Fig. 7 [399]Open in a new tab (a) tSNE plots of different Cldn genes in flow cytometry sorted CD31^+ endothelial cells from mouse lung tissue[400]34. (b) tSNE plots of different Cldn genes in endothelial cells from mouse lung tissue[401]35. (c) tSNE plots of Claudin family genes in the endothelial cells from human lung scRNA-seq data[402]35. (d) tSNE plots of Claudin family genes in the endothelial cells from mouse kidney scRNA-seq data[403]36. (e) tSNE plots of Claudin family genes in the endothelial cells from human kidney scRNA-seq data[404]37. Colors coded for the relative gene expressions. Extended Data Fig. 8. Claudins expression in distinct cell types. [405]Extended Data Fig. 8 [406]Open in a new tab (a) Different Claudin expressions in different endothelial cell types of mouse lung[407]1,[408]34. (b, c) tSNE plots of Claudin 5 in different cell types of in mouse lung (b) and highly expressed in lung endothelial cells (c)[409]35. (d) Different Claudins expression in different mouse lung cell types[410]35. (e) Expression of diverse Claudins in different mouse kidney cell types[411]36. (f, g) Diverse Claudins expression in different mouse[412]34 (f) and human brain cell types (g)[413]61. (h) Colocalization of Claudin 2 and CD31 in WT kidney cortex and medulla (individual images in [414]Supplementary Fig. 5a, [415]b). (i) Colocalization of Claudin 10 and CD31 in WT kidney cortex and medulla (individual images in [416]Supplementary Fig. 5c, [417]d). Scale bar, 50 μm (upper), 25 μm (lower). (j) Representative colocalization of Claudin 5 and CD31 in the lung of mice in the indicated groups. Scale bar, 20 μm. (k) Quantification of percentage of Claudin 5 and CD31 double positive area per visual field in the indicated groups, n = 5–9 mice per group. These images and graphs also shown in [418]Supplementary Fig. 6. Data are presented as mean values +/− SEM. Unpaired two-tailed t test, with Welch’s correction applied for unequal variances (determined by F-test). P values are listed, ns: not significant. Extended Data Fig. 9. Data Figure 9. Ang2 and Claudins signaling and Claudin2/10 suppression. [419]Extended Data Fig. 9 [420]Open in a new tab (a) Relative mRNA expression of Cldn5 in parental NIH-3T3 cells and NIH-3T3 cells treated with Adeno-CMV-luc (Ad-Ctrl) or Ad-CMV-CLDN5v2 (Ad-Claudin 5), n = 3 biologically independent experiments (with each 2 technical replicates). (b) Representative western blot and quantification of his-tagged Claudin 5 (His), Claudin 5, and β-actin in parental NIH-3T3 cells and NIH-3T3 cells treated with Adeno-CMV-luc (Ad-Ctrl) or Ad-CMV-CLDN5v2 (Ad-Claudin 5), n = 3 biologically independent experiments. Uncropped blots are shown in the Image Source Data File. (c) Relative mRNA expression of Cldn5 in NIH-3T3 cells with the indicated treatment. Dox on/off indicates doxycycline exposure, n = 4 biologically independent experiments (with each 2 technical replicates). (d) Orthotopic 4T1 tumor volume over time in the indicated groups, n = 5 mice per group. (e) CLDN5 mRNA levels in metastatic lungs of patients with breast cancer (n = 9) and healthy lungs (n = 6) ([421]GSE193103)62. (f, g) Relative mRNA expression of Cldn2 (f) and Cldn10 (g) in kidney endothelial cells treated with the indicated guide RNAs, n = 3 biologically independent experiments (with each 2 technical replicates). (h) Relative mRNA expression of Cldn2 and Cldn10 in sorted E-cadherin^+ epithelial cells and CD31^+ endothelial cells, n = 3 mice per group. Gating strategy is shown in [422]Supplementary Fig. 9. (i, j) Relative mRNA expression levels of Cldn2 and Cldn10 in sorted CD31^+ endothelial cells (i) and E-cadherin^+ epithelial cells (j) after treatment with the indicated guide RNAs, n = 3 mice per group. (k) Experimental design for the evaluation of the downregulation of Claudin 2 and Claudin 10 in the kidney of mice with 4T1 orthotopic tumors. (l) Orthotopic 4T1 tumor volume over time in the indicated groups. On day 14, LentiCRISPRv2 control vector, n = 5; LentiCRISPRv2 Cldn2 and Cldn10, n = 5; Day 18, LentiCRISPRv2 control vector, n = 5; LentiCRISPRv2 Cldn2 and Cldn10, n = 6; Day 22, LentiCRISPRv2 control vector, n = 8; LentiCRISPRv2 Cldn2 and Cldn10, n = 6 mice. (m) Representative H&E images of the lung of mice in the indicated groups. Scale bar, 25 μm. (n) Quantification of lung metastatic area. LentiCRISPRv2 control vector, n = 15; LentiCRISPRv2 Cldn2 and Cldn10, n = 20 mice. (o) Representative images for GFP immunolabeling and quantification of percent of the GFP positive area in lung. Scale bar, 50 μm, n = 9 mice per group. Data are presented as mean values +/− SEM, Min-to-Max in E. For a: one-way ANOVA test with Tukey’s multiple comparisons test. For d: two-way ANOVA with Sidak’s multiple comparisons test. For b, c, f, g: one-way ANOVA test with Dunnett’s multiple comparisons test. For e: two-tailed Mann-Whiney test. For i, j, n, o: Unpaired two-tailed t test, with Welch’s correction applied for unequal variances (determined by F-test). Extended Data Fig. 10. Knockdown of Claudin 2/10 redirects metastasis to the kidney. [423]Extended Data Fig. 10 [424]Open in a new tab (a) Representative H&E images of the kidney of mice in the indicated groups. Scale bar, 50 μm (upper), 25 μm (lower). (b) Percentage of mice with kidney lesion in the indicated groups. LentiCRISPRv2 control vector, n = 15; LentiCRISPRv2 Cldn2 and Cldn10, n = 20 mice. (c) Representative images for GFP immunolabeling in kidney. Scale bar, 50 μm (upper), 25 μm (lower). (d) Number of mice with GFP positive kidney. (e) Quantification of percent of the GFP positive area in the indicated groups, n = 12 mice per group. (f) Orthotopic 4T1 tumor volume over time in the indicated groups. LentiCRISPRv2 control vector without fibrosis, n = 14; LentiCRISPRv2 control vector with fibrosis, n = 11; LentiCRISPRv2 Cldn2 and Cldn10 without fibrosis, n = 12 mice; LentiCRISPRv2 Cldn2 and Cldn10 with fibrosis, n = 10 mice. (g) Number of mice with GFP positive kidney in the indicated groups. LentiCRISPRv2 control vector without fibrosis, n = 14; LentiCRISPRv2 control vector with fibrosis, n = 11; LentiCRISPRv2 Cldn2 and Cldn10 without fibrosis, n = 12 mice; LentiCRISPRv2 Cldn2 and Cldn10 with fibrosis, n = 10 mice. (h) Representative GFP and CD31 staining of the kidneys in mice with Claudin 2 and Claudin 10 down regulation together with or without kidney fibrosis. (i) Representative GFP and CD31 staining in the metastatic kidney with Claudin 2 and Claudin 10 downregulation. For i-j, individual images are shown in [425]Supplementary Fig. 10A-[426]C. Scale bar, 25 μm. (j) Representative images for CD31 and quantification of percent of the CD31 positive area in tumor when downregulate Claudin 2/10 in the kidney. Scale bar, 50 μm. LentiCRISPRv2 control vector, n = 5; LentiCRISPRv2 Cldn2 and Cldn10, n = 7 mice. (k) Representative images for CD31 and quantification of percent of the CD31 positive area in tumor when downregulate Claudin 2 and Claudin 10 with or without kidney fibrosis. Scale bar, 50 μm. LentiCRISPRv2 control vector without fibrosis, n = 5; LentiCRISPRv2 control vector with fibrosis, n = 4; LentiCRISPRv2 Cldn2 and Cldn10 without fibrosis, n = 4 mice; LentiCRISPRv2 Cldn2 and Cldn10 with fibrosis, n = 6 mice. Data are presented as mean values +/− SEM. For b: two-sided Chi-square. For f: two-way ANOVA with Sidak’s multiple comparisons test. For j: Unpaired two-tailed t test, with Welch’s correction applied for unequal variances (determined by F-test). For k: one-way ANOVA with Tukey’s multiple comparisons test. P values are listed, ns: not significant. Supplementary Material Supplementary Table [427]NIHMS2052537-supplement-Supplementary_Table.docx^ (1.8MB, docx) Supplementary Figures [428]NIHMS2052537-supplement-Supplementary_Figures.pdf^ (2.7MB, pdf) Acknowledgments