Introduction: Hypertension (HTN) is a major cardiovascular disease that can cause and be worsened by renal damage and inflammation. We previously reported that renal lymphatic endothelial cells (LECs) increase in response to HTN and that augmenting lymphangiogenesis in the kidneys reduces blood pressure and renal pro-inflammatory immune cells in mice with various forms of HTN. Our aim was to evaluate the specific changes that renal LECs undergo in HTN. Methods: We performed single-cell RNA sequencing. Using the angiotensin II-induced and salt-sensitive mouse models of HTN, we isolated renal CD31+ and podoplanin+ cells. Results: Sequencing of these cells revealed three distinct cell types with unique expression profiles, including LECs. The number and transcriptional diversity of LECs increased in samples from mice with HTN, as demonstrated by 597 differentially expressed genes (p < 0.01), 274 significantly enriched pathways (p < 0.01), and 331 regulons with specific enrichment in HTN LECs. These changes demonstrate a profound inflammatory response in renal LECs in HTN, leading to an increase in genes and pathways associated with inflammation-driven growth and immune checkpoint activity in LECs. Conclusion: These results reinforce and help to further explain the benefits of renal LECs and lymphangiogenesis in HTN.

Hypertension (HTN) affects nearly half of all adults and leads to heart disease, stroke, and other severe cardiovascular conditions [1, 2]. The kidneys are actively involved in blood pressure (BP) regulation and are especially susceptible to HTN-related organ damage since they possess sensitive microvasculature that can be easily damaged by elevated BP, leading to immune cell activation and accumulation and chronic inflammation. As this process continues, pro-inflammatory and cytotoxic immune cells begin producing additional cytokines and attacking healthy cells [3, 4]. This process creates a loop of damage and inflammation that propagates the original issue of heightened BP. A great deal of work has been done to determine the contributions of immune cells in the kidneys to the development and maintenance of HTN. Notable experiments involve the depletion or adoptive transfer of immune cell subsets to and from hypertensive and normotensive animals, with several immune cell types taken from hypertensive animals being able to promote a pro-hypertensive state in normotensive animals [5, 6].

A natural control mechanism for this self-damaging feedback loop exists in the form of lymphatic endothelial cells (LECs), which are capable of trafficking or inactivating immune cells to reduce their inflammatory and cytotoxic potential in the surrounding tissue. Our lab has previously demonstrated that renal lymphatic vessels expand and grow as a result of HTN and hypertensive stimuli and that augmenting lymphangiogenesis in the kidney can reduce BP or prevent the development of HTN [7‒10]. However, little is known about the specific transcriptional changes that renal LECs undergo in response to HTN or how these changes enable them to compensate for elevated BP.

Single-cell RNA sequencing (scRNAseq) is an increasingly popular technique for providing an immense amount of transcriptional data in a variety of settings with incredible resolution [11]. This degree of resolution allows for a more refined understanding of transcriptomic changes and how specific cell populations can alter more granularly, rather than all at once. Additionally, new methods for analyzing these patterns and alterations allow for highly accurate predictions of cellular functionality that go beyond basic differential expression [12, 13].

In order to better evaluate the activity and response of renal LECs in HTN, we utilized scRNAseq on renal cell populations taken from hypertensive mice using two different mouse models of HTN. To combat the relative rarity of LECs in the kidney, we used a combination of the pan-endothelial marker CD31 and the lymphatic marker podoplanin, which are common markers to locate and specify renal LECs and lymphatic vessels [14, 15]. Single-cell transcriptomics allow for an exceptional degree of resolution in determining how individual cell types respond on a functional level to a condition and we sought to use this approach to better dissect how renal LECs respond to the murine angiotensin II-induced (A2HTN) and salt-sensitive (SSHTN) models of HTN.

Murine HTN Models

Male C57/BL6 mice were purchased from Jackson Laboratories and were between 10 and 14 weeks of age at the start of each model. We utilized the A2HTN and SSHTN models as described previously (n = 6 for each group) [7, 9]. Briefly, for the A2HTN model, mice were implanted with subcutaneous osmotic pumps under anesthesia. The pumps delivered either 1,000 ng/kg/min of angiotensin II in saline or pure saline for 3 weeks prior to euthanasia by isoflurane followed by organ retrieval. For the SSHTN model, mice were primed with 0.5 g/L of LNAME in their drinking water for 2 weeks followed by a 2-week washout period. Afterward, mice received either 4% salt chow or normal chow diet for 3 weeks prior to euthanasia and organ retrieval. Animal use protocols were approved by the IACUC of Texas A&M University.

BP Measurements

Systolic BPs were measured by tail cuff using the IITC Life Science BP acquisition system as described previously [16]. The chamber and restraints were warmed to 34°C and mice were allowed to acclimate for 5 min prior to measurement. BPs were taken prior to surgery and on days 7, 14, and 21 for the A2HTN groups or at the end of the washout period and on days 7, 14, and 21 during the 4% salt diet for the SSHTN groups.

Single Cell Suspension and CD31+/Podoplanin+ Enrichment

At the end of each model, the animals were euthanized and the kidneys from all animals in a given group were pooled and finely minced. Enzymatic digestion was performed as described previously using a combination of Collagenase D (2.5 mg/mL, Roche) and Dispase II (1 mg/mL, Sigma-Aldrich) and was accompanied by mechanical dissociation through light shaking at 10-min intervals [7, 17]. The cell suspension was filtered using 70 μm and 40 μm filters and washed twice to remove dead cells. Cells were then incubated with biotinylated rat anti-mouse CD31 antibodies (BD Biosciences, catalog number: 553371) and separated using the Invitrogen CELLection Biotin Binder Kit (Thermo Fisher Scientific, catalog number: 11533D). This process was repeated using biotinylated goat anti-mouse podoplanin antibodies (R&D Systems, catalog number: BAF3244) and the Invitrogen CELLection Biotin Binder Kit, then filtered and washed again. Cells were counted and assessed for viability using the Vi-CELL XR Cell Counter (Beckman Coulter Life Sciences).

Library Preparation, Sequencing, and Transcriptome Alignment

Libraries were prepared with the 10X Genomics Chromium Next GEM Single Cell 3′ Reagent Kit V3.1 using its standard protocol and cDNA from each sample was verified with the Agilent High Sensitivity D5000 ScreenTape assay, quantified, normalized, and then sequenced on an Illumina NovaSeq 6000 with an S4 flow cell by the Texas A&M Institute for Genome Sciences and Society. The resulting FASTQ files were aligned with Cell Ranger 7.0.0 via 10X Genomics Cloud Analysis using the 10 mm (2020 revision) mouse genome assembly.

Quality Control, Clustering, and Cell Type Determination

The files for each group were loaded into Seurat version 4.1 [18]. For quality control, genes and cells were removed from each sample using parameters derived from previous work in scRNAseq analysis of renal endothelium [19]. Cells with <300 or >3,000 genes and cells with a mitochondrial gene ratio above 10% were removed and genes expressed by <50 cells and mitochondrial genes were removed. Additionally, DoubletFinder 2.0.3 was run to exclude likely doublets in the sample [20]. Cells were clustered using 20 principle components for PCA and each cluster’s cell type identity was determined based on a list of genes derived from bulk RNAseq and scRNAseq data of renal epithelium, blood and lymphatic endothelium, and immune cells [21‒23]. Based on the expression and/or absence of known markers and their 100 most highly expressed genes, we identified groups of LECs, myeloid immune cells, and an unexpected group of multipotent cells, then labeled the clusters accordingly.

Differential Gene Expression and Gene Set Enrichment Analysis of LECs

Due to the relatively small number of LECs in the control samples and the lack of significant differences in the BP and gene expression between the two models’ controls, the two control samples were merged and reclustered into a single control group to improve clustering resolution for the purpose of comparative analyses. Differential expression between hypertensive (HTN) and normotensive control (Ctrl) samples was performed in Seurat with default parameters. Gene set enrichment analysis (GSEA) was performed using Fast Gene Set Enrichment Analysis (FGSEA 1.20) and the mouse curated and ontology gene sets from the Molecular Signatures Database via msigdbr 7.5.1 (https://www.gsea-msigdb.org/gsea/msigdb/) [24].

Regulon Analysis

The Python implementation of SCENIC (pySCENIC 0.11.2 in Python 3.8) and mouse transcription factor motifs (v9) were used to find differentially active positive regulons [25]. Regulon prediction was performed using default parameters and the resulting matrices were used to determine regulon specificity scores between HTN and Ctrl groups. Regulons are referred to in the text, figures, and tables by the identified genes that regulate the full group of enriched genes.

Blood Pressure

Weekly tail cuff BP readings after the start of each model (days 7, 14, and 21) were elevated in HTN groups over their respective controls (Table 1; p < 0.001), similar to previously reported values [7, 9, 10]. The A2HTN control group (A2 Ctrl) and SSHTN control group (SS Ctrl) showed no significant differences in BP, which aligns with previous data showing minimal differences between these control groups.

Table 1.

Tail cuff systolic BP readings by group (values given as mean ± SEM) before and during each model

GroupDay 0Day 7Day 14Day 21
A2HTN 107±0.91 153.75±1.89 156.43±2.06 168.55±1.22 
A2 Ctrl 107.25±0.75 107.2±1.04 107.75±0.78 102.52±1.84 
SSHTN 105.25±1.11 139.88±1.07 140.17±1.14 141.25±1.75 
SS Ctrl 105.25±0.85 106±0.82 106.25±1.11 104.5±0.65 
GroupDay 0Day 7Day 14Day 21
A2HTN 107±0.91 153.75±1.89 156.43±2.06 168.55±1.22 
A2 Ctrl 107.25±0.75 107.2±1.04 107.75±0.78 102.52±1.84 
SSHTN 105.25±1.11 139.88±1.07 140.17±1.14 141.25±1.75 
SS Ctrl 105.25±0.85 106±0.82 106.25±1.11 104.5±0.65 

Alignment, Quality Control, and Clustering of scRNAseq Samples

Samples were grouped into merged HTN and merged Ctrl aggregate groups to determine model-neutral effects of HTN. After FASTQ alignment, the samples were loaded into Seurat for quality control for cell health and doublets, then the original and merged groups were clustered and cluster-specific gene markers were analyzed [18]. A total of ∼15,000 cells were present across all samples and several distinct and consistent clusters were identified in each sample and indicated the presence of multiple cell types in addition to multiple cellular states per cell type.

Cell Type Cluster Identification and Verification

Considering the dual enrichment for CD31+ and podoplanin+ and the variety of known cell types present in the kidney, as well as the markers they express as mRNA and cell surface markers, we determined that there were renal LECs, myeloid immune cells (MICs), and a third, unusual cell type which we dubbed support cells (SCs) present in these samples. Clusters in each sample were labeled as one of these cell types using known cell type-specific markers from bulk RNAseq and scRNAseq (Fig. 1 and online suppl. Fig. S1, S2; for all online suppl. material, see https://doi.org/10.1159/000539721; Tables 2, 3 and online suppl. Tables S1–S7) [19, 21‒23, 26‒41]. We noted a relative scarcity of LECs in the SS Ctrl sample, which had led to issues with clustering resolution, and chose to utilize the merged Ctrl group for all further comparisons involving a control sample to avoid this issue and the potential bias it could introduce. The top 200 most variable genes per cell type were determined across all samples to help verify the assigned labels (Fig. 2; online suppl. Table S8).

Fig. 1.

TSNE plots after quality control showing clusters (top row) and assigned cell type labels (bottom row). All samples (a, d), hypertensive samples (b, e), and normotensive samples (c, f).

Fig. 1.

TSNE plots after quality control showing clusters (top row) and assigned cell type labels (bottom row). All samples (a, d), hypertensive samples (b, e), and normotensive samples (c, f).

Close modal
Table 2.

Top cluster marker genes for each cluster with all samples merged

ClusterGeneAverage log2FCAdjusted p valueGene function/associations
Gpx3 0.498644212 8.87E−191 Secreted reactive oxygen species scavenger and antioxidant 
Magi2 0.78243152 Tight junction protein in podocytes 
Slc34a1 0.840827456 1.42E−185 Sodium/phosphate cotransporter 2A 
Plvap 3.011301882 Permeability of endothelial cells 
Fabp4 3.015830222 Regulator of lipolysis and inducible by lipopolysaccharide 
Tpt1 2.318354037 1.11E−237 Calcium-binding protein involved in growth 
S100a4 3.341049133 Associated with fibroblasts and activated macrophages 
Plat 3.949391223 Tissue-type plasminogen activator that converts plasminogen to plasmin 
Kcnd3 2.210474898 5.16E−76 Voltage-gated ion channels involved in transport of electrolytes 
Ogn 2.068872057 8.28E−176 Associated with fibroblasts in the kidney 
10 C1qb 4.672472573 Subunit of C1q, which is part of the first step of the classical complement pathway 
11 Meis2 1.642192491 7.06E−24 Homeobox transcription factor and regulator 
12 Vwf 4.290790239 Glycoprotein involved in platelet adhesion and commonly produced by endothelium 
ClusterGeneAverage log2FCAdjusted p valueGene function/associations
Gpx3 0.498644212 8.87E−191 Secreted reactive oxygen species scavenger and antioxidant 
Magi2 0.78243152 Tight junction protein in podocytes 
Slc34a1 0.840827456 1.42E−185 Sodium/phosphate cotransporter 2A 
Plvap 3.011301882 Permeability of endothelial cells 
Fabp4 3.015830222 Regulator of lipolysis and inducible by lipopolysaccharide 
Tpt1 2.318354037 1.11E−237 Calcium-binding protein involved in growth 
S100a4 3.341049133 Associated with fibroblasts and activated macrophages 
Plat 3.949391223 Tissue-type plasminogen activator that converts plasminogen to plasmin 
Kcnd3 2.210474898 5.16E−76 Voltage-gated ion channels involved in transport of electrolytes 
Ogn 2.068872057 8.28E−176 Associated with fibroblasts in the kidney 
10 C1qb 4.672472573 Subunit of C1q, which is part of the first step of the classical complement pathway 
11 Meis2 1.642192491 7.06E−24 Homeobox transcription factor and regulator 
12 Vwf 4.290790239 Glycoprotein involved in platelet adhesion and commonly produced by endothelium 
Table 3.

Top cluster marker genes for each cluster in normotensive (Ctrl) and hypertensive (HTN) samples

SampleClusterGeneAverage log2FCAdjusted p valueGene function/associations
Ctrl Slc8a1 0.291696787 9.66E−12 Sodium/calcium exchange 
Slc34a1 0.419890473 6.68E−11 Sodium/phosphate cotransport 
Setbp1 0.394531057 1.30E−51 SET-binding transcription factor associated with multiple kidney cell types 
Fabp4 5.164852557 Regulator of lipolysis and inducible by lipopolysaccharide 
C1qa 5.030738954 Subunit of C1q, which is part of the first step of the classical complement pathway 
Efcc1 2.711040656 5.24E−56 Calcium ion binding 
Flt1 2.302050467 5.28E−13 Encodes VEGFR1, which acts as a decoy receptor for VEGF to regulate endothelial growth 
HTN Gpx3 1.514905487 6.53E−277 Secreted reactive oxygen species scavenger and antioxidant 
Fabp4 2.004034553 Regulator of lipolysis and inducible by lipopolysaccharide 
Slc12a1 1.414953211 Sodium-potassium-chloride cotransporter associated with Loop of Henle 
Plvap 2.148853318 Permeability of endothelial cells 
Pde4d 1.764841827 5.31E−176 Binds to mTORC1 and contributes to its downstream signaling 
Vim 3.37467389 Marker associated with epithelial to mesenchymal transition 
Gm37350 3.520657826 Long non-coding RNA with unknown function 
Rps29 1.421202152 3.07E−51 Part of a ribosomal subunit and required for handling of rRNA 
SampleClusterGeneAverage log2FCAdjusted p valueGene function/associations
Ctrl Slc8a1 0.291696787 9.66E−12 Sodium/calcium exchange 
Slc34a1 0.419890473 6.68E−11 Sodium/phosphate cotransport 
Setbp1 0.394531057 1.30E−51 SET-binding transcription factor associated with multiple kidney cell types 
Fabp4 5.164852557 Regulator of lipolysis and inducible by lipopolysaccharide 
C1qa 5.030738954 Subunit of C1q, which is part of the first step of the classical complement pathway 
Efcc1 2.711040656 5.24E−56 Calcium ion binding 
Flt1 2.302050467 5.28E−13 Encodes VEGFR1, which acts as a decoy receptor for VEGF to regulate endothelial growth 
HTN Gpx3 1.514905487 6.53E−277 Secreted reactive oxygen species scavenger and antioxidant 
Fabp4 2.004034553 Regulator of lipolysis and inducible by lipopolysaccharide 
Slc12a1 1.414953211 Sodium-potassium-chloride cotransporter associated with Loop of Henle 
Plvap 2.148853318 Permeability of endothelial cells 
Pde4d 1.764841827 5.31E−176 Binds to mTORC1 and contributes to its downstream signaling 
Vim 3.37467389 Marker associated with epithelial to mesenchymal transition 
Gm37350 3.520657826 Long non-coding RNA with unknown function 
Rps29 1.421202152 3.07E−51 Part of a ribosomal subunit and required for handling of rRNA 
Fig. 2.

Variance plots showing the top 10 most variable genes by cell type (LECs [a], SCs [b], and MICs [c]). The full gene lists are available in online supplementary Table S8.

Fig. 2.

Variance plots showing the top 10 most variable genes by cell type (LECs [a], SCs [b], and MICs [c]). The full gene lists are available in online supplementary Table S8.

Close modal

LEC-Specific Gene Expression Alterations in HTN

To focus on the effects of HTN on LECs, we isolated and reclustered the LECs present in each group then reanalyzed the cluster-specific markers (Fig. 3 and online suppl. Fig. S3–S4; Tables 4, 5 and online suppl. Table S9–S13) [42‒49]. Broadly speaking, there was a marked increase in the number of LECs present in the HTN samples over the Ctrl samples as well as an increase in the heterogeneity of cellular states, which aligns with known pathological alterations in LECs [8, 50‒53].

Fig. 3.

a, b TSNE cluster plot and feature plot of LECs from all samples. The features shown here were the most consistent and selective LEC cluster markers across all samples.

Fig. 3.

a, b TSNE cluster plot and feature plot of LECs from all samples. The features shown here were the most consistent and selective LEC cluster markers across all samples.

Close modal
Table 4.

Top cluster marker genes for each LEC cluster with all samples merged

ClusterGeneAverage log2FCAdjusted p valueGene function/associations
Itm2b 0.903931521 1.30E−170 Ubiquitously expressed and regulates urate influx 
Igfbp5 0.955341214 5.17E−76 IGF binding protein involved in cellular metabolism 
Cmss1 1.259288653 9.97E−116 Translation as part of small ribosomal subunit 
S100a4 2.559622945 2.13E−280 Associated with fibroblasts and activated macrophages 
Gm37350 3.501234612 Long non-coding RNA with unknown function 
Gsn 1.21461466 1.32E−57 Involved in assembly of actin filaments and associated with fibroblasts 
Mgp 4.70486961 2.20E−191 Suppresses proliferation of T cells and inhibits production of inflammatory cytokines 
Nlgn1 2.614150491 7.77E−187 Associated with nervous system development and synapse production 
ClusterGeneAverage log2FCAdjusted p valueGene function/associations
Itm2b 0.903931521 1.30E−170 Ubiquitously expressed and regulates urate influx 
Igfbp5 0.955341214 5.17E−76 IGF binding protein involved in cellular metabolism 
Cmss1 1.259288653 9.97E−116 Translation as part of small ribosomal subunit 
S100a4 2.559622945 2.13E−280 Associated with fibroblasts and activated macrophages 
Gm37350 3.501234612 Long non-coding RNA with unknown function 
Gsn 1.21461466 1.32E−57 Involved in assembly of actin filaments and associated with fibroblasts 
Mgp 4.70486961 2.20E−191 Suppresses proliferation of T cells and inhibits production of inflammatory cytokines 
Nlgn1 2.614150491 7.77E−187 Associated with nervous system development and synapse production 
Table 5.

Top cluster marker genes for each LEC cluster in normotensive (Ctrl) and hypertensive (HTN) samples

SampleClusterGeneAverage log2FCAdjusted p valueGene function/associations
Ctrl Plpp1 3.045046159 2.11E−33 Hydrolysis and cellular intake of lipids and a marker for endothelial cells 
Fxyd2 4.28985122 1.94E−23 Associated with distal convoluted tubule and reduces ion binding affinity of sodium-potassium ATPase 
HTN Itm2b 0.940919126 3.68E−164 Ubiquitously expressed and regulates urate influx 
Igfbp5 0.989341957 1.76E−65 IGF binding protein involved in cellular metabolism 
Igfbp7 1.653626736 1.04E−94 IGF binding protein involved in cellular metabolism 
Gm37350 3.491965373 Long non-coding RNA with unknown function 
S100a4 2.517749248 6.69E−223 Associated with fibroblasts and activated macrophages 
Cmss1 0.963677914 1.62E−43 Translation as part of small ribosomal subunit 
Pcdh7 2.865016673 2.19E−160 Cadherin family member involved with cell-to-cell adhesion and recognition 
Htra1 2.671634266 1.32E−181 Serine proteases involved in cellular growth and IGF binding protein cleavage 
SampleClusterGeneAverage log2FCAdjusted p valueGene function/associations
Ctrl Plpp1 3.045046159 2.11E−33 Hydrolysis and cellular intake of lipids and a marker for endothelial cells 
Fxyd2 4.28985122 1.94E−23 Associated with distal convoluted tubule and reduces ion binding affinity of sodium-potassium ATPase 
HTN Itm2b 0.940919126 3.68E−164 Ubiquitously expressed and regulates urate influx 
Igfbp5 0.989341957 1.76E−65 IGF binding protein involved in cellular metabolism 
Igfbp7 1.653626736 1.04E−94 IGF binding protein involved in cellular metabolism 
Gm37350 3.491965373 Long non-coding RNA with unknown function 
S100a4 2.517749248 6.69E−223 Associated with fibroblasts and activated macrophages 
Cmss1 0.963677914 1.62E−43 Translation as part of small ribosomal subunit 
Pcdh7 2.865016673 2.19E−160 Cadherin family member involved with cell-to-cell adhesion and recognition 
Htra1 2.671634266 1.32E−181 Serine proteases involved in cellular growth and IGF binding protein cleavage 

Differentially expressed genes (DEGs) were identified with Seurat using the merged control LEC group and an adjusted p value of <0.01 for all comparisons. When comparing all HTN and all Ctrl LECs, there were a total of 597 DEGs (486 ↑, 112 ↓; online suppl. Table S14). Among the top 25 most significant DEGs are genes related to growth, inflammation, and cytoskeletal remodeling (Table 6) [42, 43, 54‒69].

Table 6.

Top 25 differentially expressed genes by adjusted p value in hypertensive LECs versus normotensive LECs

GeneAverage log2FCPCT1PCT2Δ PCTAdjusted p value
Gphn −1.349155745 0.928 0.961 −0.033 6.15E−45 
Wfdc15b −2.042218012 0.006 0.116 −0.11 4.43E−42 
Fxyd2 −3.609638629 0.13 0.417 −0.287 2.47E−39 
Cdk8 −1.148227748 0.855 0.9 −0.045 3.98E−31 
Ly6e 0.858126251 0.957 0.653 0.304 8.31E−31 
Cmss1 −0.938748689 0.99 0.992 −0.002 3.85E−30 
Ptprb 0.85123309 0.908 0.564 0.344 3.81E−26 
Gm10076 0.877467653 0.686 0.255 0.431 6.97E−25 
Keg1 −1.379986448 0.037 0.189 −0.152 2.15E−24 
Hsp90ab1 0.694761984 0.945 0.714 0.231 1.71E−23 
Hspb1 0.830427029 0.849 0.483 0.366 1.65E−21 
Nrp1 0.740709283 0.919 0.629 0.29 2.03E−21 
Pecam1 0.759466224 0.868 0.506 0.362 2.17E−21 
Cyyr1 0.789947597 0.797 0.436 0.361 1.86E−20 
Kng2 −1.179047648 0.029 0.151 −0.122 3.45E−19 
Atp1b1 −1.728931885 0.073 0.239 −0.166 1.08E−18 
Sparc 0.750310006 0.884 0.595 0.289 1.16E−18 
Mef2c 0.751728185 0.763 0.402 0.361 2.12E−18 
Fabp4 0.900380012 0.853 0.602 0.251 3.54E−18 
Mmrn2 0.737492292 0.633 0.274 0.359 4.31E−18 
Ptprm 0.817038218 0.793 0.483 0.31 6.11E−18 
AY036118 −0.885621194 0.948 0.938 0.01 8.27E−18 
Rdx 0.757823755 0.856 0.552 0.304 8.68E−18 
Chrm3 0.988604657 0.736 0.413 0.323 1.92E−17 
Cdh5 0.68805324 0.817 0.459 0.358 2.41E−17 
GeneAverage log2FCPCT1PCT2Δ PCTAdjusted p value
Gphn −1.349155745 0.928 0.961 −0.033 6.15E−45 
Wfdc15b −2.042218012 0.006 0.116 −0.11 4.43E−42 
Fxyd2 −3.609638629 0.13 0.417 −0.287 2.47E−39 
Cdk8 −1.148227748 0.855 0.9 −0.045 3.98E−31 
Ly6e 0.858126251 0.957 0.653 0.304 8.31E−31 
Cmss1 −0.938748689 0.99 0.992 −0.002 3.85E−30 
Ptprb 0.85123309 0.908 0.564 0.344 3.81E−26 
Gm10076 0.877467653 0.686 0.255 0.431 6.97E−25 
Keg1 −1.379986448 0.037 0.189 −0.152 2.15E−24 
Hsp90ab1 0.694761984 0.945 0.714 0.231 1.71E−23 
Hspb1 0.830427029 0.849 0.483 0.366 1.65E−21 
Nrp1 0.740709283 0.919 0.629 0.29 2.03E−21 
Pecam1 0.759466224 0.868 0.506 0.362 2.17E−21 
Cyyr1 0.789947597 0.797 0.436 0.361 1.86E−20 
Kng2 −1.179047648 0.029 0.151 −0.122 3.45E−19 
Atp1b1 −1.728931885 0.073 0.239 −0.166 1.08E−18 
Sparc 0.750310006 0.884 0.595 0.289 1.16E−18 
Mef2c 0.751728185 0.763 0.402 0.361 2.12E−18 
Fabp4 0.900380012 0.853 0.602 0.251 3.54E−18 
Mmrn2 0.737492292 0.633 0.274 0.359 4.31E−18 
Ptprm 0.817038218 0.793 0.483 0.31 6.11E−18 
AY036118 −0.885621194 0.948 0.938 0.01 8.27E−18 
Rdx 0.757823755 0.856 0.552 0.304 8.68E−18 
Chrm3 0.988604657 0.736 0.413 0.323 1.92E−17 
Cdh5 0.68805324 0.817 0.459 0.358 2.41E−17 

Upregulated genes are shown in red and downregulated genes are shown in blue. PCT1 refers to the percentage of cells in the HTN samples expressing the gene, while PCT2 gives the percentage of expression for cells in Ctrl samples. Δ PCT gives the change in percentage from Ctrl samples to HTN samples.

In the top 25 DEGs, CD31 (Pecam1) and neuropilin-1 (Nrp1) were upregulated by 69% and 67% respectively and both have functions related to both endothelial growth and immunoregulation [70, 71]. In addition, lymphocyte antigen 6 family member E (Ly6e), a gene known to enhance immune checkpoint activity as well the growth-related effects of hypoxia-inducible factor-1 alpha subunit (HIF-1α; Hif1a), was also significantly upregulated with an 81% increase in expression and was expressed in 95% of LECs in HTN (up from 65% in Ctrl) [72, 73]. Other immune checkpoint and immunomodulatory genes were also upregulated in HTN, including butyrophilin-like 9 (Btnl9) (86% increase), TRAIL (Tnfsf10) (42% increase), galectin 9 (Lgals9) (31% increase), and CD47 (26% increase) [74, 75]. Similar to a previous report involving the effects of Leishmania major infection, we also noted an increase in H2-K1, H2-D1, and beta-2-microglobulin (B2m), which are all a part of MHC class I antigen presentation in LECs, as well as increases in guanylate-binding protein (GBP) genes, which is induced by interferon signaling and can lead to inflammasome activation [76‒78].

Comparisons for each individual model were also conducted, using A2HTN or SSHTN LECs instead of the merged HTN LEC group. A2HTN LECs had a total of 1,070 DEGs (948 ↑, 122 ↓; online suppl. Table S15) whereas SSHTN LECs only had 315 DEGs (244 ↑, 71 ↓; online suppl. Table S16). The DEGs for each model were largely similar to the merged HTN comparison, though Ly6e’s upregulation was even more exaggerated in the A2HTN LECs and was accompanied by upregulation of additional immune checkpoint genes, including PD-L1 (Cd274) (24% increase), CD200 (30% increase), nectin cell adhesion molecule 2 (Nectin2) (30% increase), and B2m (87% increase) [75, 79‒81]. Adding to the upregulation of MHC I-related genes seen in the merged HTN comparison, there was upregulation of MHC class II genes (notably CD74, H2-Ab1, and H2-Aa) in A2HTN LECs. In SSHTN, heat shock protein 90 alpha family class B member 1 (Hsp90ab1) increased by 93% (vs. a 22% increase in A2HTN) and heat shock protein family B (small) member 1 (Hspb1) increased by 114% (vs. 32% in A2HTN), possibly indicating a preference for heat shock protein signaling for growth in the presence of both excessive BP and sodium [54, 57, 60, 82]. When comparing LECs from the two models directly, additional growth and development genes are upregulated in A2HTN over SSHTN, including CD151, which has been associated with lymphangiogenesis and invasion in cancer, and B2m and Ly6e are increased by 48% and 32% respectively (online suppl. Table S17) [83].

Inflammatory and Immune Pathways Are Enriched in LECs in HTN

GSEA was performed using FGSEA with the identified DEGs and the curated gene sets (M2) and ontology gene sets (M5) from the mouse collections of the Molecular Signatures Database. Pathways were filtered to only contain entries with adjusted p values <0.01 and are shown sorted by normalized enrichment score (NES) in figures. Comparing all hypertensive LECs to normotensive LECs, we identified 56 significantly enriched pathways (18 ↑, 38 ↓) for the M2 gene sets and 218 (62 ↑, 156 ↓) for the M5 gene sets (Fig. 4; online suppl. Tables S18, S19). A2HTN LECs compared to normotensive LECs yielded 75 (35 ↑, 40 ↓) and 181 (51 ↑, 130 ↓) enriched pathways for the M2 and M5 sets respectively, while SSHTN LECs had 58 (14 ↑, 44 ↓) and 227 (58 ↑, 169 ↓) pathways enriched over Ctrl LECs (online suppl. Fig. S5; online suppl. Tables S20–S23). 19 (17 ↑ in A2HTN, 2 ↑ in SSHTN) and 12 (10 ↑ in A2HTN, 2 ↑ in SSHTN) pathways for M2 and M5 sets were differentially enriched between the two HTN models (online suppl. Tables S24–S25). Overall, the enriched pathways indicate a strong inflammatory and immune-type response, the induction of robust angiogenic/lymphangiogenic growth mechanisms, and an overall shift in metabolic processes in HTN. A2HTN LECs again show an exaggerated inflammatory response in addition to an increase in immune markers whereas SSHTN LECs seem to be more characterized by the downregulated pathways, as all pathways common to the two models were more downregulated in SSHTN LECs than in their A2HTN counterparts.

Fig. 4.

GSEA NES plots for the top 25 M2 (a) and M5 (b) pathways by NES from the DEGs between hypertensive LECs and normotensive LECs. The full GSEA results are available in online supplementary Tables S18 and S19.

Fig. 4.

GSEA NES plots for the top 25 M2 (a) and M5 (b) pathways by NES from the DEGs between hypertensive LECs and normotensive LECs. The full GSEA results are available in online supplementary Tables S18 and S19.

Close modal

HTN Leads to Enhanced Inflammatory Regulon Activation in LECs

Utilizing the resolution provided by scRNAseq, new approaches have been developed to identify regulons, which consist of a group of genes which are expressed as a unit and under the control of a single master gene (usually a transcription factor). By evaluating co-expression of experimentally-validated master genes and groups of their subordinate genes in different conditions, differential regulon enrichment can be identified and help provide more detailed context to known alterations in transcription factor activity and how they relate to the broader pathophysiology of a disease, including how pathologically altered genes influence one another. To determine if any gene expression changes in LECs could be tied to transcription factor activity related to HTN, regulons and regulon specificity scores (RSSs), which measure how specific a regulon’s activity is to a given group, cluster, or condition, were identified using pySCENIC for each group and each comparison respectively [25, 84].

RSS values were determined for merged hypertensive, A2HTN, and SSHTN LECs versus normotensive LECs, as well as comparing LECs from each model (online suppl. Tables S26–S29). A total of 605 regulons were identified for the HTN versus Ctrl LEC comparison, with 331 having RSS values >0.5 for HTN LECs, indicating preference for activation in HTN, and 64 having RSS values >0.5 for Ctrl LECs, indicating preference for activation in controls. A2HTN versus Ctrl LECs showed 602 regulons (342 for A2HTN, 72 for Ctrl) and SSHTN versus Ctrl LECs gave 583 regulons (345 for SSHTN, 57 for Ctrl). Comparing the two HTN models yielded 613 regulons, with 192 showing preference for A2HTN and 263 with preference for SSHTN. The regulons with the highest RSSs for each hypertensive versus normotensive comparison were plotted by expression and RSS (Fig. 5 and online suppl. Fig. S6) and clustermaps were generated for all regulons in each group (online suppl. Fig. S7).

Fig. 5.

SCENIC RSS heatmap (a) and specificity score plot (b) comparing hypertensive LECs and normotensive LECs. The specificity score plot shows the five regulons with the highest score per group in red and the full RSS list is available in online supplementary Table S26.

Fig. 5.

SCENIC RSS heatmap (a) and specificity score plot (b) comparing hypertensive LECs and normotensive LECs. The specificity score plot shows the five regulons with the highest score per group in red and the full RSS list is available in online supplementary Table S26.

Close modal

The top five regulons by RSS for HTN LECs were MAF BZIP transcription factor F (Maff), high mobility group nucleosomal binding domain 3 (Hmgn3), HIV type I enhancer-binding protein zinc finger 1 (Hivep1), Ras responsive element binding protein 1 (Rreb1), and Hif1a. Rreb1 and Hif1a are both major drivers for growth in LECs in inflammatory conditions, while Hivep1 negatively regulates NF-κB signaling in monocytes and macrophages [85‒90]. Additionally, Maff has anti-inflammatory functions and can alter the function of neighboring monocytes and Hmgn3 is upregulated by TGFB1 and the high-mobility group (HMG) family of proteins, which includes Hmgn3, has a strong association with immune checkpoint genes [91‒97].

Models and Cell Types

The A2HTN and SSHTN models used here generated robust increases in systolic BP which align with previous studies from our lab and others and are known to be sufficient to cause serious renal vascular injury [9, 98, 99]. The cell surface marker enrichment for CD31+/podoplanin+ cells was successful in increasing the proportion of LECs present in the sample, but it also had the effect of pulling in MICs and SCs [14, 15, 100]. While unexpected, these cell types are both known to express these markers under different conditions and may represent distinct subsets of larger populations that weakly express similar markers to LECs or even participate in inflammatory processes akin to LECs. The SCs we identified share many characteristics with mesenchymal stem cells, which are known to have anti-inflammatory effects and can even reduce BP and improve kidney function, while MICs can have a wide variety of pro-inflammatory, anti-inflammatory, and pro-lymphangiogenic effects [6, 101‒104].

Comparisons with Known LEC Alterations in HTN

Our previous studies have shown an increase in LECs and LEC markers in the kidneys in HTN and our results here seem to reinforce this at the single-cell expression level. Not only do we see an increase in LECs and genes known to be involved in lymphangiogenesis in the HTN samples, but we see an increase in the diversity of transcription factors and genes expressed by LECs, though some LEC markers appear to differ at single-cell resolution and across different organs [22, 105, 106]. However, while there were multiple clear LEC clusters in the HTN samples, we had far fewer cells that could be grouped into obvious LEC clusters in control samples, which likely reflects a relative scarcity of LECs in kidneys under normal conditions. Additionally, most DEGs when comparing hypertensive and normotensive LECs were upregulated rather than downregulated (ranging from 77 to 89% of all DEGs being upregulated in HTN, depending on the comparison), possibly indicating the LECs exist in a fairly quiescent state under normal conditions, then react strongly in response to inflammation.

Canonical LEC Genes

Regarding canonical LEC and lymphangiogenic markers in these samples, we observed that SRY-box transcription factor 18 (Sox18) is upregulated by 65% in merged HTN LECs and is up by 105% in A2HTN LECs, but does not have any sort of regulon specificity between the groups. On the other hand, COUP-TF II (Nr2f2) is not differentially expressed, but has specificity scores for HTN groups ranging from 0.721 to 0.767. Similarly, the master lymphatic transcription factor prospero homeobox 1 (Prox1) is not strongly or differentially expressed in these samples, yet it is among the top variable genes for LECs in this dataset and has a slightly higher RSS for A2HTN compared to controls (0.250 vs. 0.167), and VEGFC also ranks in the top 100 variable genes for both LECs and MICs.

Inflammatory LEC Growth in HTN

Conversely, while many of the DEGs, pathways, and regulons presented here are related to inflammation, growth, and expansion in endothelial cells, they are not a complete match for the traditional suite of genes associated with lymphangiogenesis. Instead, many of them are more comparable to studies involving various forms of inflammatory processes in LECs in other conditions or states and may indicate that these new lymphatics are being generating by and expanding through a continuous flood of HIF-1α, VEGF, and TLR signaling, rather than the more coordinated signaling that triggers and sustains lymphangiogenesis during development [73, 107‒110]. Notably, HIF-1α and its downstream effectors can contribute to lymphangiogenesis and lymphatic vessel malformation when upregulated [88]. PI3K (Pik3ca), which is one of HIF-1α’s effectors, has a well-known mutation in cancers that exaggerates lymphangiogenesis and lymphatic malformations, with recent scRNAseq analysis of this mutation’s activity revealing a number of similarities to the genes and pathways we have presented here [111].

Role of Immune Checkpoints on LECs in HTN

In addition to growth-related effects, HIF-1α can also directly stimulate the expression of immune checkpoint ligands, including PD-L1, which can regulate and reduce the number of cytotoxic T cells in an area [85]. Immune checkpoint inhibitors as part of cancer treatment have been observed to have negative effects on systemic and pulmonary HTN, though this is somewhat disputed [112‒114]. However, the relationship between HTN and activation of the immune system is well-established and is a topic of ongoing research, particularly regarding how the immune system can directly impact BP [115, 116]. CD8+ T cells are involved in the onset and worsening of HTN and other cardiovascular diseases through their pro-inflammatory and cytotoxic functions, so reducing their numbers and activity in the renal interstitium through lymphatic immune checkpoint signaling would help mitigate further inflammation and damage, potentially even lowering BP as a result [3, 4, 117]. Through the upregulation of genes involved in immune checkpoints and immunoregulation, including Ly6e, Btnl9, Lgals9, B2m, Tnfsf10, and Cd274, renal LECs (particularly in the A2HTN model) appear to be increasing their immunosuppressive potential, which is likely a natural compensatory response to limit the excessive and maladaptive inflammation that is present in the kidneys in HTN.

Summary and Future Directions

To better understand how renal LECs are altered by and influence the progression of HTN, we performed scRNAseq on CD31+/podoplanin+ renal cells taken from A2HTN and SSHTN mice and their respective controls. We identified LEC, SC, and MIC populations and analyzed the changes in gene expression in LECs due to HTN through differential gene expression, GSEA, and regulon analysis. There was a marked increase in the number and activity of LECs in the HTN groups, including the upregulation of inflammatory growth pathways that are similar to what is seen in other inflammatory conditions. Additionally, multiple genes related to immune checkpoint and immunoregulatory signaling were upregulated in LECs in HTN, providing a new potential role for renal LECs in counteracting the development and maintenance of HTN and HTN-related kidney damage.

In the future, we plan to evaluate how the inflammatory growth seen here impacts the function of LECs and their role in immune cell trafficking. As this type of growth has been associated with lymphatic hyperplasia and malformations, there may be a substantial impact on clearance and trafficking, leading to an accumulation of pro-inflammatory immune cells in the kidneys. Also, with the increase in immune checkpoint genes, we will investigate how lymphatic immune checkpoint and immunoregulatory signaling regulates the immune cell populations responsible for the onset and continuation of HTN. Better understanding of the pathological changes in renal LECs and immune cells that influence the development and progression of HTN will help provide novel approaches for treating severe HTN.

This study protocol was reviewed and approved by the IACUC of Texas A&M University, Approval No. 2022-0083.

The authors have no conflicts of interest to declare.

This work was funded by NIH RO1 (DK120493) to B.M. Mitchell and Texas A&M University T3 to J.M. Rutkowski.

Justin G. McDermott: conceptualization, data curation, formal analysis, investigation, methodology, project administration, resources, software, writing – original draft, and writing – review and editing. Bethany L. Goodlett and Shobana Navaneethabalakrishnan: data curation and investigation. Heidi A. Creed: conceptualization, data curation, and investigation. Joseph M. Rutkowski: conceptualization, funding acquisition, and project administration. Brett M. Mitchell: conceptualization, funding acquisition, project administration, resources, and writing – review and editing.

The data that support the findings of this study are publicly available in NCBI GEO (GSE 236410) at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE236410. Any additional inquiries can be addressed by the corresponding author.

1.
Chobufo
MD
,
Gayam
V
,
Soluny
J
,
Rahman
EU
,
Enoru
S
,
Foryoung
JB
, et al
.
Prevalence and control rates of hypertension in the USA: 2017–2018
.
Int J Cardiol Hypertens
.
2020
;
6
:
100044
.
2.
NCD Risk Factor Collaboration (NCD-RisC)
.
Worldwide trends in hypertension prevalence and progress in treatment and control from 1990 to 2019: a pooled analysis of 1201 population-representative studies with 104 million participants
.
Lancet Lond Engl
.
2021
;
398
(
10304
):
957
80
.
3.
Abais-Battad
JM
,
Rudemiller
NP
,
Mattson
DL
.
Hypertension and immunity: mechanisms of T cell activation and pathways of hypertension
.
Curr Opin Nephrol Hypertens
.
2015
;
24
(
5
):
470
4
.
4.
Ren
J
,
Crowley
SD
.
Role of T-cell activation in salt-sensitive hypertension
.
Am J Physiol Heart Circ Physiol
.
2019
;
316
(
6
):
H1345
53
.
5.
Lopez Gelston
CA
,
Mitchell
BM
.
Recent advances in immunity and hypertension
.
Am J Hypertens
.
2017
;
30
(
7
):
643
52
.
6.
Navaneethabalakrishnan
S
,
Smith
HL
,
Arenaz
CM
,
Goodlett
BL
,
McDermott
JG
,
Mitchell
BM
.
Update on immune mechanisms in hypertension
.
Am J Hypertens
.
2022
;
35
(
10
):
842
51
.
7.
Balasubbramanian
D
,
Gelston
CAL
,
Lopez
AH
,
Iskander
G
,
Tate
W
,
Holderness
H
, et al
.
Augmenting renal lymphatic density prevents angiotensin II-induced hypertension in male and female mice
.
Am J Hypertens
.
2020
;
33
(
1
):
61
9
.
8.
Kneedler
SC
,
Phillips
LE
,
Hudson
KR
,
Beckman
KM
,
Lopez Gelston
CA
,
Rutkowski
JM
, et al
.
Renal inflammation and injury are associated with lymphangiogenesis in hypertension
.
Am J Physiol-Ren Physiol
.
2017
;
312
(
5
):
F861
9
.
9.
Lopez
GCA
,
Balasubbramanian
D
,
Abouelkheir
GR
,
Lopez
AH
,
Hudson
KR
,
Johnson
ER
, et al
.
Enhancing renal lymphatic expansion prevents hypertension in mice
.
Circ Res
.
2018
;
122
(
8
):
1094
101
.
10.
Sims
BM
,
Goodlett
BL
,
Allbee
ML
,
Pickup
EJ
,
Chiasson
VL
,
Arenaz
CM
, et al
.
Time restricted feeding decreases renal innate immune cells and blood pressure in hypertensive mice
.
J Hypertens
.
2022
;
40
(
10
):
1960
8
.
11.
Jovic
D
,
Liang
X
,
Zeng
H
,
Lin
L
,
Xu
F
,
Luo
Y
.
Single-cell RNA sequencing technologies and applications: a brief overview
.
Clin Transl Med
.
2022
;
12
(
3
):
e694
.
12.
Zappia
L
,
Phipson
B
,
Oshlack
A
.
Exploring the single-cell RNA-seq analysis landscape with the scRNA-tools database
.
PLoS Comput Biol
.
2018
;
14
(
6
):
e1006245
.
13.
Zappia
L
,
Theis
FJ
.
Over 1000 tools reveal trends in the single-cell RNA-seq analysis landscape
.
Genome Biology
.
2021
;
22
(
1
):
301
.
14.
Kriehuber
E
,
Breiteneder-Geleff
S
,
Groeger
M
,
Soleiman
A
,
Schoppmann
SF
,
Stingl
G
, et al
.
Isolation and characterization of dermal lymphatic and blood endothelial cells reveal stable and functionally specialized cell lineages
.
J Exp Med
.
2001
;
194
(
6
):
797
808
.
15.
Lokmic
Z
,
Ng
ES
,
Burton
M
,
Stanley
EG
,
Penington
AJ
,
Elefanty
AG
.
Isolation of human lymphatic endothelial cells by multi-parameter fluorescence-activated cell sorting
.
J Vis Exp
.
2015
(
99
):
52691
.
16.
Balasubbramanian
D
,
Baranwal
G
,
Clark
MCC
,
Goodlett
BL
,
Mitchell
BM
,
Rutkowski
JM
.
Kidney-specific lymphangiogenesis increases sodium excretion and lowers blood pressure in mice
.
J Hypertens
.
2020
;
38
(
5
):
874
85
.
17.
Goodlett
BL
,
Kang
CS
,
Yoo
E
,
Navaneethabalakrishnan
S
,
Balasubbramanian
D
,
Love
SE
, et al
.
A kidney-targeted nanoparticle to augment renal lymphatic density decreases blood pressure in hypertensive mice
.
Pharmaceutics
.
2021
;
14
(
1
):
84
.
18.
Hao
Y
,
Hao
S
,
Andersen-Nissen
E
,
Mauck
WM
,
Zheng
S
,
Butler
A
, et al
.
Integrated analysis of multimodal single-cell data
.
Cell
.
2021
;
184
(
13
):
3573
87.e29
.
19.
Dumas
SJ
,
Meta
E
,
Borri
M
,
Goveia
J
,
Rohlenova
K
,
Conchinha
NV
, et al
.
Single-cell RNA sequencing reveals renal endothelium heterogeneity and metabolic adaptation to water deprivation
.
J Am Soc Nephrol
.
2020
;
31
(
1
):
118
38
.
20.
McGinnis
CS
,
Murrow
LM
,
Gartner
ZJ
.
DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors
.
Cell Syst
.
2019
;
8
(
4
):
329
37.e4
.
21.
Clark
JZ
,
Chen
L
,
Chou
CL
,
Jung
HJ
,
Lee
JW
,
Knepper
MA
.
Representation and relative abundance of cell-type selective markers in whole-kidney RNA-Seq data
.
Kidney Int
.
2019
;
95
(
4
):
787
96
.
22.
Pasut
A
,
Becker
LM
,
Cuypers
A
,
Carmeliet
P
.
Endothelial cell plasticity at the single-cell level
.
Angiogenesis
.
2021
;
24
(
2
):
311
26
.
23.
Petrova
TV
,
Mäkinen
T
,
Mäkelä
TP
,
Saarela
J
,
Virtanen
I
,
Ferrell
RE
, et al
.
Lymphatic endothelial reprogramming of vascular endothelial cells by the Prox-1 homeobox transcription factor
.
EMBO J
.
2002
;
21
(
17
):
4593
9
.
24.
Korotkevich
G
,
Sukhov
V
,
Budin
N
,
Shpak
B
,
Artyomov
MN
,
Sergushichev
A
.
Fast gene set enrichment analysis
.
Bioinformatics
.
2016
.
25.
Aibar
S
,
González-Blas
CB
,
Moerman
T
,
Huynh-Thu
VA
,
Imrichova
H
,
Hulselmans
G
, et al
.
SCENIC: single-cell regulatory network inference and clustering
.
Nat Methods
.
2017
;
14
(
11
):
1083
6
.
26.
Fujimoto
N
,
He
Y
,
D’Addio
M
,
Tacconi
C
,
Detmar
M
,
Dieterich
LC
.
Single-cell mapping reveals new markers and functions of lymphatic endothelial cells in lymph nodes
.
PLoS Biol
.
2020
;
18
(
4
):
e3000704
.
27.
He
B
,
Chen
P
,
Zambrano
S
,
Dabaghie
D
,
Hu
Y
,
Möller-Hackbarth
K
, et al
.
Single-cell RNA sequencing reveals the mesangial identity and species diversity of glomerular cell transcriptomes
.
Nat Commun
.
2021
;
12
(
1
):
2141
.
28.
Jafree
DJ
,
Moulding
D
,
Kolatsi-Joannou
M
,
Perretta Tejedor
N
,
Price
KL
,
Milmoe
NJ
, et al
.
Spatiotemporal dynamics and heterogeneity of renal lymphatics in mammalian development and cystic kidney disease
.
Elife
.
2019
;
8
:
e48183
.
29.
Kalucka
J
,
de Rooij
LPMH
,
Goveia
J
,
Rohlenova
K
,
Dumas
SJ
,
Meta
E
, et al
.
Single-cell transcriptome atlas of murine endothelial cells
.
Cell
.
2020
;
180
(
4
):
764
79.e20
.
30.
Kirita
Y
,
Wu
H
,
Uchimura
K
,
Wilson
PC
,
Humphreys
BD
.
Cell profiling of mouse acute kidney injury reveals conserved cellular responses to injury
.
Proc Natl Acad Sci
.
2020
;
117
(
27
):
15874
83
.
31.
Schupp
JC
,
Adams
TS
,
Cosme
C
,
Raredon
MSB
,
Yuan
Y
,
Omote
N
, et al
.
Integrated single-cell atlas of endothelial cells of the human lung
.
Circulation
.
2021
;
144
(
4
):
286
302
.
32.
Chen
L
,
Chou
CL
,
Knepper
MA
.
Targeted single-cell RNA-seq identifies minority cell types of kidney distal nephron
.
J Am Soc Nephrol
.
2021
;
32
(
4
):
886
96
.
33.
Whitlock
JH
,
Soelter
TM
,
Howton
TC
,
Wilk
EJ
,
Oza
VH
,
Lasseigne
BN
.
Cell-type-specific gene expression and regulation in the cerebral cortex and kidney of atypical Setbp1 S858R Schinzel Giedion Syndrome mice
.
J Cell Mol Med
.
2023
;
27
(
22
):
3565
77
.
34.
Sun
N
,
Zhao
X
.
Therapeutic implications of FABP4 in cancer: an emerging target to tackle cancer
.
Front Pharmacol
.
2022
;
13
:
948610
.
35.
Jourde-Chiche
N
,
Fakhouri
F
,
Dou
L
,
Bellien
J
,
Burtey
S
,
Frimat
M
, et al
.
Endothelium structure and function in kidney health and disease
.
Nat Rev Nephrol
.
2019
;
15
(
2
):
87
108
.
36.
Karaman
S
,
Paavonsalo
S
,
Heinolainen
K
,
Lackman
MH
,
Ranta
A
,
Hemanthakumar
KA
, et al
.
Interplay of vascular endothelial growth factor receptors in organ-specific vessel maintenance
.
J Exp Med
.
2022
;
219
(
3
):
e20210565
.
37.
Nirgude
S
,
Choudhary
B
.
Insights into the role of GPX3, a highly efficient plasma antioxidant, in cancer
.
Biochem Pharmacol
.
2021
;
184
:
114365
.
38.
Denzer
L
,
Muranyi
W
,
Schroten
H
,
Schwerk
C
.
The role of PLVAP in endothelial cells
.
Cell Tissue Res
.
2023
;
392
(
2
):
393
412
.
39.
Good
KV
,
Vincent
JB
,
Ausió
J
.
MeCP2: the genetic driver of rett syndrome epigenetics
.
Front Genet
.
2021
;
12
:
620859
.
40.
Jeong
MH
,
Urquhart
G
,
Lewis
C
,
Chi
Z
,
Jewell
JL
.
Inhibition of phosphodiesterase 4D suppresses mTORC1 signaling and pancreatic cancer growth
.
JCI Insight
.
2023
;
8
(
13
):
e158098
.
41.
Mirabello
L
,
Macari
ER
,
Jessop
L
,
Ellis
SR
,
Myers
T
,
Giri
N
, et al
.
Whole-exome sequencing and functional studies identify RPS29 as a novel gene mutated in multicase Diamond-Blackfan anemia families
.
Blood
.
2014
;
124
(
1
):
24
32
.
42.
Manouchehri
N
,
Salinas
VH
,
Hussain
RZ
,
Stüve
O
.
Distinctive transcriptomic and epigenomic signatures of bone marrow-derived myeloid cells and microglia in CNS autoimmunity
.
Proc Natl Acad Sci
.
2023
;
120
(
6
):
e2212696120
.
43.
Wang
F
,
Cai
B
,
Li
KC
,
Hu
XY
,
Lu
YJ
,
Wang
Q
, et al
.
FXYD2, a γ subunit of Na⁺, K⁺-ATPase, maintains persistent mechanical allodynia induced by inflammation
.
Cell Res
.
2015
;
25
(
3
):
318
34
.
44.
Chung
JJ
,
Goldstein
L
,
Chen
YJJ
,
Lee
J
,
Webster
JD
,
Roose-Girma
M
, et al
.
Single-cell transcriptome profiling of the kidney glomerulus identifies key cell types and reactions to injury
.
J Am Soc Nephrol
.
2020
;
31
(
10
):
2341
54
.
45.
Mandal
AK
,
Mount
DB
.
Interaction between ITM2B and GLUT9 links urate transport to neurodegenerative disorders
.
Front Physiol
.
2019
;
10
:
1323
.
46.
Haywood
NJ
,
Slater
TA
,
Matthews
CJ
,
Wheatcroft
SB
.
The insulin like growth factor and binding protein family: novel therapeutic targets in obesity & diabetes
.
Mol Metab
.
2019
;
19
:
86
96
.
47.
Zhang
S
,
Fu
X
.
The clinical significance and biological function of PCDH7 in cervical cancer
.
Cancer Manag Res
.
2021
;
13
:
3841
7
.
48.
Janosevic
D
,
Myslinski
J
,
McCarthy
TW
,
Zollman
A
,
Syed
F
,
Xuei
X
, et al
.
The orchestrated cellular and molecular responses of the kidney to endotoxin define a precise sepsis timeline
.
Elife
.
2021
;
10
:
e62270
.
49.
Tang
Y
,
Kwiatkowski
DJ
,
Henske
EP
.
Midkine expression by stem-like tumor cells drives persistence to mTOR inhibition and an immune-suppressive microenvironment
.
Nat Commun
.
2022
;
13
(
1
):
5018
.
50.
Donnan
MD
,
Kenig-Kozlovsky
Y
,
Quaggin
SE
.
The lymphatics in kidney health and disease
.
Nat Rev Nephrol
.
2021
;
17
(
10
):
655
75
.
51.
Petrova
TV
,
Koh
GY
.
Biological functions of lymphatic vessels
.
Science
.
2020
;
369
(
6500
):
eaax4063
.
52.
Seeger
H
,
Bonani
M
,
Segerer
S
.
The role of lymphatics in renal inflammation
.
Nephrol Dial Transplant
.
2012
;
27
(
7
):
2634
41
.
53.
Wiig
H
,
Swartz
MA
.
Interstitial fluid and lymph formation and transport: physiological regulation and roles in inflammation and cancer
.
Physiol Rev
.
2012
;
92
(
3
):
1005
60
.
54.
Batulan
Z
,
Pulakazhi Venu
VK
,
Li
Y
,
Koumbadinga
G
,
Alvarez-Olmedo
DG
,
Shi
C
, et al
.
Extracellular release and signaling by heat shock protein 27: role in modifying vascular inflammation
.
Front Immunol
.
2016
;
7
:
285
.
55.
Bekkar
A
,
Isorce
N
,
Snäkä
T
,
Claudinot
S
,
Desponds
C
,
Kopelyanskiy
D
, et al
.
Dissection of the macrophage response towards infection by the Leishmania-viral endosymbiont duo and dynamics of the type I interferon response
.
Front Cell Infect Microbiol
.
2022
;
12
:
941888
.
56.
Biaoxue
R
,
Xiling
J
,
Shuanying
Y
,
Wei
Z
,
Xiguang
C
,
Jinsui
W
, et al
.
Upregulation of Hsp90-beta and annexin A1 correlates with poor survival and lymphatic metastasis in lung cancer patients
.
J Exp Clin Cancer Res
.
2012
;
31
(
1
):
70
.
57.
Charmpilas
N
,
Kyriakakis
E
,
Tavernarakis
N
.
Small heat shock proteins in ageing and age-related diseases
.
Cell Stress Chaperones
.
2017
;
22
(
4
):
481
92
.
58.
Choi
KJ
,
Nam
JK
,
Kim
JH
,
Choi
SH
,
Lee
YJ
.
Endothelial-to-mesenchymal transition in anticancer therapy and normal tissue damage
.
Exp Mol Med
.
2020
;
52
(
5
):
781
92
.
59.
Frye
M
,
Dierkes
M
,
Küppers
V
,
Vockel
M
,
Tomm
J
,
Zeuschner
D
, et al
.
Interfering with VE-PTP stabilizes endothelial junctions in vivo via Tie-2 in the absence of VE-cadherin
.
J Exp Med
.
2015
;
212
(
13
):
2267
87
.
60.
Kaigorodova
EV
,
Zavyalova
MV
,
Bogatyuk
MV
,
Tarabanovskaya
NA
,
Slonimskaya
EM
,
Perelmuter
VM
.
Relationship between the expression of phosphorylated heat shock protein beta-1 with lymph node metastases of breast cancer
.
Cancer Biomark
.
2015
;
15
(
2
):
143
50
.
61.
Li
J
,
Jiang
X
,
Li
C
,
Che
H
,
Ling
L
,
Wei
Z
.
Proteomic alteration of endometrial tissues during secretion in polycystic ovary syndrome may affect endometrial receptivity
.
Clin Proteomics
.
2022
;
19
(
1
):
19
.
62.
Liang
J
,
Chen
M
,
Hughes
D
,
Chumanevich
AA
,
Altilia
S
,
Kaza
V
, et al
.
CDK8 selectively promotes the growth of colon cancer metastases in the liver by regulating gene expression of TIMP3 and matrix metalloproteinases
.
Cancer Res
.
2018
;
78
(
23
):
6594
606
.
63.
Ning
YL
,
Yang
ZQ
,
Xian
SX
,
Lin
JZ
,
Lin
XF
,
Chen
WT
.
Bioinformatics analysis identifies hub genes and molecular pathways involved in sepsis-induced myopathy
.
Med Sci Monit
.
2020
;
26
:
e919665
.
64.
Seibert
TA
,
Hibbert
B
,
Chen
YX
,
Rayner
K
,
Simard
T
,
Hu
T
, et al
.
Serum heat shock protein 27 levels represent a potential therapeutic target for atherosclerosis: observations from a human cohort and treatment of female mice
.
J Am Coll Cardiol
.
2013
;
62
(
16
):
1446
54
.
65.
Sychev
ZE
.
Unraveling the proteome of endothelial cells latently infected with KSHV reveals reliance on peroxisome lipid metabolism and redox regulation
.
University of Washington
;
2017
.
66.
Yan
Y
,
Xing
C
,
Xiao
Y
,
Shen
X
,
Zhang
Z
,
He
C
, et al
.
Discovery and anti-inflammatory activity evaluation of a novel CDK8 inhibitor through upregulation of IL-10 for the treatment of inflammatory bowel disease in vivo
.
J Med Chem
.
2022
;
65
(
10
):
7334
62
.
67.
Zhao
Y
,
Tao
L
,
Jiang
D
,
Chen
X
,
Li
P
,
Ning
Y
, et al
.
The -144C/A polymorphism in the promoter of HSP90beta is associated with multiple organ dysfunction scores
.
PLoS One
.
2013
;
8
(
3
):
e58646
.
68.
Zhou
K
,
Jiang
T
,
Liu
Y
,
Zhao
Z
,
Huang
L
,
Li
G
.
FXYD2 mRNA expression represents a new independent factor that affects survival of glioma patients and predicts chemosensitivity of patients to temozolomide
.
BMC Neurol
.
2021
;
21
(
1
):
438
.
69.
Zhou
L
,
Kiss
E
,
Demmig
R
,
Kirsch
J
,
Nawrotzki
RA
,
Kuhse
J
.
Binding of gephyrin to microtubules is regulated by its phosphorylation at Ser270
.
Histochem Cell Biol
.
2021
;
156
(
1
):
5
18
.
70.
Chaudhary
B
,
Khaled
YS
,
Ammori
BJ
,
Elkord
E
.
Neuropilin 1: function and therapeutic potential in cancer
.
Cancer Immunol Immunother
.
2014
;
63
(
2
):
81
99
.
71.
Newman
DK
,
Fu
G
,
Adams
T
,
Cui
W
,
Arumugam
V
,
Bluemn
T
, et al
.
The adhesion molecule PECAM-1 enhances the TGF-β-mediated inhibition of T cell function
.
Sci Signal
.
2016
;
9
(
418
):
ra27
.
72.
AlHossiny
M
,
Luo
L
,
Frazier
WR
,
Steiner
N
,
Gusev
Y
,
Kallakury
B
, et al
.
Ly6E/K signaling to TGFβ promotes breast cancer progression, immune escape, and drug resistance
.
Cancer Res
.
2016
;
76
(
11
):
3376
86
.
73.
Yeom
CJ
,
Zeng
L
,
Goto
Y
,
Morinibu
A
,
Zhu
Y
,
Shinomiya
K
, et al
.
LY6E: a conductor of malignant tumor growth through modulation of the PTEN/PI3K/Akt/HIF-1 axis
.
Oncotarget
.
2016
;
7
(
40
):
65837
48
.
74.
Chyuan
IT
,
Hsu
PN
.
TRAIL regulates T cell activation and suppresses inflammation in autoimmune diseases
.
Cell Mol Immunol
.
2020
;
17
(
12
):
1281
3
.
75.
Yu
Y
,
Tang
H
,
Franceschi
D
,
Mujagond
P
,
Acharya
A
,
Deng
Y
, et al
.
Immune checkpoint gene expression profiling identifies programmed cell death ligand-1 centered immunologic subtypes of oral and squamous cell carcinoma with favorable survival
.
Front Med
.
2021
;
8
:
759605
.
76.
Praefcke
GJK
.
Regulation of innate immune functions by guanylate-binding proteins
.
Int J Med Microbiol
.
2018
;
308
(
1
):
237
45
.
77.
Santambrogio
L
,
Berendam
SJ
,
Engelhard
VH
.
The antigen processing and presentation machinery in lymphatic endothelial cells
.
Front Immunol
.
2019
;
10
:
1033
.
78.
Venugopal
G
,
Bird
JT
,
Washam
CL
,
Roys
H
,
Bowlin
A
,
Byrum
SD
, et al
.
In vivo transcriptional analysis of mice infected with Leishmania major unveils cellular heterogeneity and altered transcriptomic profiling at single-cell resolution
.
PLoS Negl Trop Dis
.
2022
;
16
(
7
):
e0010518
.
79.
Busch
E
,
Ahadova
A
,
Kosmalla
K
,
Bohaumilitzky
L
,
Pfuderer
PL
,
Ballhausen
A
, et al
.
Beta-2-microglobulin mutations are linked to a distinct metastatic pattern and a favorable outcome in microsatellite-unstable stage IV gastrointestinal cancers
.
Front Oncol
.
2021
;
11
:
669774
.
80.
Kloor
M
,
Michel
S
,
Buckowitz
B
,
Rüschoff
J
,
Büttner
R
,
Holinski-Feder
E
, et al
.
Beta2-microglobulin mutations in microsatellite unstable colorectal tumors
.
Int J Cancer
.
2007
;
121
(
2
):
454
8
.
81.
Ozcan
M
,
Janikovits
J
,
von Knebel Doeberitz
M
,
Kloor
M
.
Complex pattern of immune evasion in MSI colorectal cancer
.
OncoImmunology
.
2018
;
7
(
7
):
e1445453
.
82.
Zhang
S
,
Sun
Y
,
Yuan
Z
,
Li
Y
,
Li
X
,
Gong
Z
, et al
.
Heat shock protein 90β inhibits apoptosis of intestinal epithelial cells induced by hypoxia through stabilizing phosphorylated Akt
.
BMB Rep
.
2013
;
46
(
1
):
47
52
.
83.
Detchokul
S
,
Newell
B
,
Williams
ED
,
Frauman
AG
.
CD151 is associated with prostate cancer cell invasion and lymphangiogenesis in vivo
.
Oncol Rep
.
2014
;
31
(
1
):
241
7
.
84.
Suo
S
,
Zhu
Q
,
Saadatpour
A
,
Fei
L
,
Guo
G
,
Yuan
GC
.
Revealing the critical regulators of cell identity in the mouse cell atlas
.
Cell Rep
.
2018
;
25
(
6
):
1436
45.e3
.
85.
Balamurugan
K
.
HIF-1 at the crossroads of hypoxia, inflammation, and cancer: hypoxia, Inflammation, and Cancer
.
Int J Cancer
.
2016
;
138
(
5
):
1058
66
.
86.
Boehme
JT
,
Morris
CJ
,
Chiacchia
SR
,
Gong
W
,
Wu
KY
,
Kameny
RJ
, et al
.
HIF-1α promotes cellular growth in lymphatic endothelial cells exposed to chronically elevated pulmonary lymph flow
.
Sci Rep
.
2021
;
11
(
1
):
1468
.
87.
Ducoli
L
,
Agrawal
S
,
Sibler
E
,
Kouno
T
,
Tacconi
C
,
Hon
CC
, et al
.
LETR1 is a lymphatic endothelial-specific lncRNA governing cell proliferation and migration through KLF4 and SEMA3C
.
Nat Commun
.
2021
;
12
(
1
):
925
.
88.
Han
T
,
Yan
J
,
Chen
H
,
Ji
Y
,
Chen
J
,
Cui
J
, et al
.
HIF-1α contributes to tube malformation of human lymphatic endothelial cells by upregulating VEGFR-3
.
Int J Oncol
.
2019
;
54
(
1
):
139
51
.
89.
Matsumoto
H
,
Scicluna
BP
,
Jim
KK
,
Falahi
F
,
Qin
W
,
Gürkan
B
, et al
.
HIVEP1 is a negative regulator of NF-κB that inhibits systemic inflammation in sepsis
.
Front Immunol
.
2021
;
12
:
744358
.
90.
Morgani
SM
,
Su
J
,
Nichols
J
,
Massagué
J
,
Hadjantonakis
AK
.
The transcription factor Rreb1 regulates epithelial architecture, invasiveness, and vasculogenesis in early mouse embryos
.
Elife
.
2021
;
10
:
e64811
.
91.
Katsuoka
F
,
Yamamoto
M
.
Small maf proteins (MafF, MafG, MafK): history, structure and function
.
Gene
.
2016
;
586
(
2
):
197
205
.
92.
Massrieh
W
,
Derjuga
A
,
Doualla-Bell
F
,
Ku
CY
,
Sanborn
BM
,
Blank
V
.
Regulation of the MAFF transcription factor by proinflammatory cytokines in myometrial cells
.
Biol Reprod
.
2006
;
74
(
4
):
699
705
.
93.
Onodera
K
,
Shavit
JA
,
Motohashi
H
,
Katsuoka
F
,
Akasaka
J
,
Engel
JD
, et al
.
Characterization of the murine mafF gene
.
J Biol Chem
.
1999
;
274
(
30
):
21162
9
.
94.
Saliba
J
,
Coutaud
B
,
Solovieva
V
,
Lu
F
,
Blank
V
.
Regulation of CXCL 1 chemokine and CSF 3 cytokine levels in myometrial cells by the MAFF transcription factor
.
J Cell Mol Med
.
2019
;
23
(
4
):
2517
25
.
95.
Sorin
S
,
Pokaew
N
,
Vaeteewoottacharn
K
,
Waraasawapati
S
,
Pairojkul
C
,
Sashida
G
, et al
.
Overexpression of HMGN3 nucleosome binding protein is associated with tumor invasion and TGF-? expression in cholangiocarcinoma
.
ScienceAsia
.
2022
;
48
(
5
):
518
.
96.
Wang
M
,
Liu
F
,
Fang
B
,
Huo
Q
,
Yang
Y
.
Proteome-scale profiling reveals MAFF and MAFG as two novel key transcription factors involved in palmitic acid-induced umbilical vein endothelial cell apoptosis
.
BMC Cardiovasc Disord
.
2021
;
21
(
1
):
448
.
97.
Wu
Z
,
Huang
Y
,
Yuan
W
,
Wu
X
,
Shi
H
,
Lu
M
, et al
.
Expression, tumor immune infiltration, and prognostic impact of HMGs in gastric cancer
.
Front Oncol
.
2022
;
12
:
1056917
.
98.
Crowley
SD
,
Gurley
SB
,
Herrera
MJ
,
Ruiz
P
,
Griffiths
R
,
Kumar
AP
, et al
.
Angiotensin II causes hypertension and cardiac hypertrophy through its receptors in the kidney
.
Proc Natl Acad Sci
.
2006
;
103
(
47
):
17985
90
.
99.
Gomolak
JR
,
Didion
SP
.
Angiotensin II-induced endothelial dysfunction is temporally linked with increases in interleukin-6 and vascular macrophage accumulation
.
Front Physiol
.
2014
;
5
:
396
.
100.
Jafree
DJ
,
Long
DA
.
Beyond a passive conduit: implications of lymphatic biology for kidney diseases
.
J Am Soc Nephrol
.
2020
;
31
(
6
):
1178
90
.
101.
Franchi
F
,
Peterson
KM
,
Xu
R
,
Miller
B
,
Psaltis
PJ
,
Harris
PC
, et al
.
Mesenchymal stromal cells improve renovascular function in polycystic kidney disease
.
Cell Transplant
.
2015
;
24
(
9
):
1687
98
.
102.
Oliveira-Sales
EB
,
Varela
VA
,
Maquigussa
E
,
Borges
FT
,
Shimoura
CG
,
Gomes
G
, et al
.
Renovascular hypertension: effects of mesenchymal stem cells in the contralateral hypertensive kidney in rats
.
Clin Exp Hypertens
.
2016
;
38
(
7
):
586
93
.
103.
Oliveira-Sales
EB
,
Varela
VA
,
Bergamaschi
CT
,
Campos
RR
,
Boim
MA
.
Effects of mesenchymal stem cells in renovascular hypertension: renovascular hypertension and mesenchymal stem cells
.
Exp Physiol
.
2015
;
100
(
5
):
491
5
.
104.
Zhao
Y
,
Zhu
X
,
Zhang
L
,
Ferguson
CM
,
Song
T
,
Jiang
K
, et al
.
Mesenchymal stem/stromal cells and their extracellular vesicle progeny decrease injury in poststenotic swine kidney through different mechanisms
.
Stem Cells Dev
.
2020
;
29
(
18
):
1190
200
.
105.
Oliver
G
,
Srinivasan
RS
.
Endothelial cell plasticity: how to become and remain a lymphatic endothelial cell
.
Development
.
2010
;
137
(
3
):
363
72
.
106.
Tabrizi
ZB
,
Ahmed
NS
,
Horder
JL
,
Storr
SJ
,
Benest
AV
.
Transcription factor control of lymphatic quiescence and maturation of lymphatic neovessels in development and physiology
.
Front Physiol
.
2021
;
12
:
672987
.
107.
Lane
R
.
The contribution of lymphatic and blood endothelial cell PD-L1 to leukocyte accumulation in skin
.
Oregon Health & Science University
;
2019
.
108.
Milam
KE
,
Parikh
SM
.
The angiopoietin-Tie2 signaling axis in the vascular leakage of systemic inflammation
.
Tissue Barriers
.
2015
;
3
(
1–2
):
e957508
.
109.
Parikh
SM
.
The angiopoietin-tie2 signaling Axis in systemic inflammation
.
J Am Soc Nephrol
.
2017
;
28
(
7
):
1973
82
.
110.
Wang
B
,
Ji
G
,
Naeem
H
,
Wang
J
,
Kantharidis
P
,
Powell
D
, et al
.
The use of targeted Next generation sequencing to explore candidate regulators of TGF-β1’s impact on kidney cells
.
Front Physiol
.
2018
;
9
:
1755
.
111.
Petkova
M
,
Kraft
M
,
Stritt
S
,
Martinez-Corral
I
,
Ortsäter
H
,
Vanlandewijck
M
, et al
.
Immune-interacting lymphatic endothelial subtype at capillary terminals drives lymphatic malformation
.
J Exp Med
.
2023
;
220
(
4
):
e20220741
.
112.
Minegishi
S
,
Kinguchi
S
,
Horita
N
,
Namkoong
H
,
Briasoulis
A
,
Ishigami
T
, et al
.
Immune checkpoint inhibitors do not increase short-term risk of hypertension in cancer patients: a systematic literature review and meta-analysis
.
Hypertension
.
2022
;
79
(
11
):
2611
21
.
113.
Palassin
P
,
Maria
ATJ
,
Ladhari
C
,
Quantin
X
,
Montani
D
,
Bourdin
A
, et al
.
Pulmonary hypertension reported with immune checkpoint inhibitors: a pharmacovigilance study
.
Cancer Immunol Immunother
.
2022
;
71
(
12
):
3093
7
.
114.
Turker
I
,
Sharma
A
,
Huang
S
,
Johnson
DB
,
Alexander
MR
.
Combination immune checkpoint inhibitor therapy is associated with increased blood pressure in melanoma patients
.
Hypertension
.
2023
;
80
(
3
):
e43
5
.
115.
Caillon
A
,
Paradis
P
,
Schiffrin
EL
.
Role of immune cells in hypertension
.
Br J Pharmacol
.
2019
;
176
(
12
):
1818
28
.
116.
Singh
MV
,
Chapleau
MW
,
Harwani
SC
,
Abboud
FM
.
The immune system and hypertension
.
Immunol Res
.
2014
;
59
(
1–3
):
243
53
.
117.
Sun
Y
,
Li
L
,
Wu
Y
,
Yang
K
.
PD-1/PD-L1 in cardiovascular disease
.
Clin Chim Acta
.
2020
;
505
:
26
30
.