Skip to main content

The immune regulatory role of lymphangiogenesis in kidney disease

Abstract

The renal lymphatic system is critical for maintaining kidney homeostasis and regulating the immune response inside the kidney. In various kidney pathological situations, the renal lymphatic network experiences lymphangiogenesis, which is defined as the creation of new lymphatic vessels. Kidney lymphangiogenesis controls immunological response inside the kidney by controlling lymphatic flow, immune cell trafficking, and immune cell regulation. Ongoing study reveals lymphangiogenesis’s different architecture and functions in numerous tissues and organs. New research suggests that lymphangiogenesis in kidney disorders may regulate the renal immune response in various ways. The flexibility of lymphatic endothelial cells (LECs) improves the kidney’s immunological regulatory function of lymphangiogenesis. Furthermore, current research has shown disparate findings regarding its impact on distinct renal diseases, resulting in contradictory outcomes even within the same kidney condition. The fundamental causes of the various effects of lymphangiogenesis on renal disorders remain unknown. In this thorough review, we explore the dual impacts of renal lymphangiogenesis on several kidney pathologies, with a particular emphasis on existing empirical data and new developments in understanding its immunological regulatory function in kidney disease. An improved understanding of the immunological regulatory function of lymphangiogenesis in kidney diseases might help design novel medicines targeting lymphatics to treat kidney pathologies.

Introduction

The significance of lymphangiogenesis in various diseases has been extensively examined in recent literature. These studies have provided significant insights into the contrasting effects of lymphangiogenesis on disease pathophysiology. Lymphangiogenesis relies on the proliferation, migration, and differentiation of lymphatic endothelial cells (LECs). The cellular processes lead to the biosynthesis of lymphatic vessels, which transport excess fluid and regulate immune responses in the lymphatic system [1]. Recent studies utilizing genetic lineage tracing and single-cell RNA sequencing have demonstrated that stem/progenitor cells also play a crucial role in lymphangiogenesis [2]. Additionally, M1 macrophages have been shown to polarize and transdifferentiate into new LECs through activation of the vascular endothelial growth factor (VEGF-C)/vascular endothelial growth factor receptor 3 (VEGFR3) pathway [3]. Lymphatic vessel proliferation comprises healthy lymphangiogenesis (during wound healing and corpus luteum development) and pathological lymphangiogenesis. The latter is caused by pathological situations such as inflammation, tumors, and transplant rejection, among others [4,5,6]. Physiological and pathological lymphangiogenesis often entail the enlargement and sprouting of preexisting lymphatic vessels (LVs) rather than neolymphangiogenesis, which is more closely related to lymph node transfer [7]. The interaction between lymphangiogenesis and various clinical conditions has a complex effect on the organism. Advanced imaging and genetic approaches have made it possible to investigate specific structures and functions within the lymphatic systems in various diseases.

Lymphangiogenesis plays complex immune regulatory roles via various mechanisms, differing from the nuanced variations of microenvironments in tissues and organs. The newly formed lymphatic vessels can either enhance or inhibit the immune response [8, 9]. The lymphatic system maintains homeostasis and supports immune responses throughout various tissues and organs [10]. In both health and disease, the lymphatic system also plays a crucial role in regulating immune responses by directly influencing immune cells and coordinating their movement from tissues to draining lymph nodes (dLNs) [11]. The unique characteristics of lymphatic vessels in both health and disease demonstrate specificity related to tissue and organ types. The characteristics influence the varied outcomes of lymphangiogenesis in different disease contexts.

Within the kidney, lymphangiogenesis is closely linked to kidney tissue inflammation, fibrosis progression, and transplant rejection [12]. Evidence unveils that it can elicit dual-sided effects in various kidney pathologies [12, 13]. Studies have illuminated that kidney lymphangiogenesis exhibits an intricate immune regulatory mechanism capable of promoting or alleviating immune responses [14], depending on the specific kidney pathology under consideration. Emerging evidence suggests that, within kidney diseases, the distinct trafficking patterns of diverse immune cells and varying durations of different pathological conditions significantly contribute to the dual-sided effect of lymphangiogenesis [15, 16].

The kidney lymphatic system selectively transports renal interstitial fluid and immune cells. It actively contributes to the maintenance of kidney homeostasis and the orchestration of kidney immune response. Notably, preexisting lymphatic vessels within the kidney are predominantly distributed in the renal cortex and rare in the medulla. However, neo-synthesized lymphatic vessels can proliferate extensively throughout the kidney [17].

Lymphatic migrations of immune cells are regulated by Various chemokines, including chemokine (C–C motif) ligand 19 (CCL19), CCL21, and chemokine (C-X-C motif) ligand 12 (CXCL12). Additionally, several inflammatory and anti-inflammatory mediators, including Interleukin-1β (IL-1β), Tumor Necrosis Factor-α (TNF-α), Interleukin-10 (IL-10), and Transforming Growth Factor β (TGF-β), also involves in the regulation of lymphatic immune cell migrations [1].

To be specific, recent findings have uncovered that lymphatic vessels can suppress the expansion of CD8+ T cells [18, 19]. The interaction between Mac-1 on DCs and ICAM-1 on LECs mediates the adhesive interactions between DCs and LECs, thereby inhibiting the ability of DCs to induce T cell proliferation [20]. Moreover, chemokine receptor chemokine (C–C motif) receptor 7 (CCR7) expressed on DCs and its CCL21 produced by LEC are the main molecules involved in DC migration [21]. At the same time, reducing DCs is beneficial for slowing the progression of inflammation [22]. The reasons and mechanisms underlying this remarkable discrepancy in diverse kidney diseases require further investigation.

LECs and their immune regulatory role

LECs are crucial in immune responses during inflammation, tumour, and other pathological conditions. Different subsets of LECs, including peripheral capillary LECs and lymph node LECs, have distinct functions. The primary functions of peripheral capillary LECs include fluid drainage, leucocyte transport, and participation in lipid metabolism. They also actively regulate the endocytosis of antigens, mediating by clathrin and caveolin. Interestingly, capillary LECs exhibit phenotypic adaptations in varying microenvironments [23]. Therefore, they can dynamically orchestrate the trafficking and activities of various immune cells. Among the intricate process of immune cell trafficking, lymphatic vessel endothelial receptor-1 (LYVE-1) makes the first adhesive contact between migrating immune cells and lymphatic endothelium, initiating the entry and trafficking of immune cells within afferent lymphatic vessels [24]. Moreover, capillary LECs secrete various chemokines to drive immune cell intravasation through a complicated process of actomyosin-mediated immune motility and β2 integrin activation during inflammatory status [24]. Among these cytokines, CCL21 is one of the most important and well-studied regulators. By binding to heparan sulfate within the extracellular matrix, CCL21 generates a hypotactic concentration gradient to promote the migration of diverse leucocytes, such as DCs, neutrophils, and monocytes, through interacting with CCR7 expressed on these immune cells [25,26,27]. Furthermore, accumulating data suggests numerous cytokines and chemokine/receptor combinations are involved in lymphatic migration. Immunosuppressive substances like IL-10 and TGF-β may prevent immune cells from migrating through the lymphatic system [28,29,30]. LN LECs exert varied functions after transporting molecules and cells to dLNs. These cells are pivotal contributors to immune surveillance in both health and disease. LN LECs and specifically distributed rapidly classify molecules [31,32,33,34,35]. These two types of LECs subtly regulate innate and adaptive immune responses [21, 22, 36,37,38,39,40,41].

Cytokines and chemokines involved in lymphangiogenesis

Lymphangiogenesis is predominantly regulated by VEGF-C and VEGF-D, both of which directly bind with VEGFR-3 and the co-receptor neuropilin 2 (NRP2), expressed on the surface of LECs, subsequently inducing lymphangiogenesis [42]. Recent studies have revealed that several types of macrophages can promote lymphangiogenesis by secreting VEGF-C in various pathological conditions, including kidney damage and cardiac injury [43,44,45,46,47]. Cortical and medullary kidney tubules can secrete VEGF-C and VEGF-D within the kidney [48]. Studies suggest that VEGF-C has an essential role in the development of lymphangiogenesis, but its impact on the maintenance of lymphatic vessels might be limited [49]. Conversely, unlike VEGF-C, VEGF-D dominates the maintenance of lymphangiogenesis, which indicates a modulatory function of VEGF-D in its developmental stage [49]. Furthermore, TGF-β and connective tissue growth factor (CTGF) also contribute to the induction of lymphangiogenesis in kidney diseases, particularly in kidney inflammation and fibrosis [50]. Additionally, angiopoietins (Angs) are involved in the lymphangiogenesis mechanism. In fact, the Ang2/Tie/PI3K signaling pathway plays a crucial role in lymphangiogenesis; blocking this pathway leads to a decrease in VEGFR3 and inhibits lymphatic vessel formation [51]. Similarly, the transcription factors FOXC1 and FOXC2, which are part of the Forkhead box (FOX) family, positively regulate lymphangiogenesis. Studies have shown that FOXC1 and FOXC2 are essential for regulating the Ras/ERK signaling pathway during lymphangiogenesis, and the loss of FOXC1 and FOXC2 promotes excessive activation of ERK, leading to abnormal lymphangiogenesis [52].

The immune regulatory functions of lymphangiogenesis in kidney diseases

Acute kidney injury

In many AKI animal models and AKI patient biopsies, VEGF-C and VEGF-D expression increased, as did robust lymphangiogenesis. Following kidney damage, inflammatory mediators such as Interferon-gamma (IFN-γ), TNF-α, and TGF-β promote lymphangiogenesis via several mechanisms [48, 53,54,55,56]. Functional neo-lymphatic vessels can manifest the same role as preexisting renal lymphatic vessels, promoting inflammation resolution through drainage of retained fluid, clearance of cellular debris, removal of pro-inflammatory cytokines and cells, and mobilizing immune cells [11, 48]. Macrophages are highly adaptable to transfer into various distinct phenotypes within the local microenvironment. Among these macrophages, M1 macrophages are predominantly infiltrated during the AKI process, primarily promoting inflammation response and inducing kidney injury [57]. They enhance the synthesis of new lymphatic vessels in renal inflammation and fibrosis microenvironment due to elevated expression levels of VEGF-C induced by TGF-β [49]. Recent evidence has demonstrated that M1 macrophages can directly contribute to the synthesis of new lymphatic vessels through transdifferentiating into LECs [3, 58]. Increased levels of VEGF-C directly suppress macrophage autophagy, which prompts M1 macrophage polarization into LECs [3].

Lymphangiogenesis adversely affects the AKI process, exacerbating renal inflammation [56, 59, 60]. In general, AKI-induced lymphangiogenesis can exert a dual-sided impact on the kidney. In AKI, kidney lymphangiogenesis also acts as an immunological regulator to balance immunity and immune pathology despite clearing excessive fluids, noxious stimuli, and inflammatory cells. Neo-synthesized lymphatic vessels can either induce or suppress the immune response in AKI models, regulating local and systematic immune systems through diverse mechanisms [61]. Firstly, it significantly enhances lymphatic flow, actively participating in the early immune regulation process after AKI. During AKI’s initiation, maintenance, and regression process, abundant immune cells, including inflammatory monocyte, neutrophil, lymphocyte and natural killer cells, orchestrate the overall immune response [62]. Therefore, lymphangiogenesis can mitigate kidney tissue inflammation by properly removing infiltrated immune cells at the injury site (Tables 1, 2).

Table 1 The role of important immune cells that involve in kidney disease
Table 2 Two-sided consequences of immune regulation through lymphangiogenesis in kidney diseases

Despite lymphatic flow, some immune cells, including T cells, B cells, and DCs, can also directly regulate LECs-related signal pathways [63, 64], contributing to the inflammatory progress. Entry of naïve T cells to afferent lymphatics is regulated by the S1P (sphingosine-1-phosphate) receptor pathway [65], while memory T cells also possess CCR7, which binds with CCL21. Immune cells with CCR7 can also be regulated by the CCL21 gradient expressed by LECs, actively migrating to dLNs through afferent lymphatics [66]. Of note, current evidence has demonstrated that enhancing antigen-specific T helper cell 1 (Th1 cell) cell migration from tissues to dLNs accelerates the resolution of inflammation. In the setting of AKI, upgraded infiltration of Th cells (T helper cells), particularly T helper cell 17 (Th17 cell), has been observed [67]. Th17 cell, which aggravates tissue injury by recruiting neutrophils and other inflammatory cells, is the most abundant lymphocyte infiltrated at the injury site following AKI in mice [68, 69]. Additionally, intestinal flora-derived Th17 cells have been proved to migrate to the kidney in kidney disease. They enter peripheral blood circulation through lymphatic vessels regulated by the S1P-R1 pathway [16]. Subsequently, they return to the renal inflammation site through blood circulation, further exacerbating the inflammatory response [16]. Increased reduction of Th17 cells through kidney lymphangiogenesis may significantly mitigate kidney damage, alleviating AKI and the following progression to chronic kidney disease (CKD). However, further studies are required to demonstrate whether lymphangiogenesis can aggravate kidney damage by regulating these Th cells.

Moreover, due to the adaptiveness of LECs, lymphangiogenesis can also directly suppress the local CD8+ T cells during inflammation. This intricate mechanism has been well-studied in the setting of skin lymphangiogenesis. Lymphatic endothelial cells (LECs) largely express nonhematopoietic programmed death-ligand 1 (PD-L1) to limit local CD8+ T cell effectors to functioning in inflamed skin and melanoma [70]. Despite PD-L1-dependent inhibition of T cell antigen receptor (TCR) signaling, evidence supports that PD-L1 can regulate lymphocyte migration through endothelial and epithelial barrier tissues [70], which indicates that PD-L1 may directly regulate T cell transendothelial migration without antigen presentation mechanism. Similarly, the activated PD-1 signalling pathway in the kidney protects the ischemia–reperfusion-induced AKI mouse model [71]. At the beginning of inflammation, infiltrated antigen-specific CD8+ T cells produce IFN-γ, which directly induces PD-L1 expression in adjacent inflammation-induced lymphatic vessels. Abundant PD-L1 expressed by LECs limits the further accumulation of CD8+ T cells at the injury site, alleviating kidney inflammation.

In the context of AKI, lymphatic migration of immune cells affects local immunity bidirectionally. Current evidence has demonstrated that lymphangiogenesis can also be detrimental to AKI [11, 72]. This detrimental impact closely correlates with positive immune feedback (Fig. 1) that enhances immune cells’ constant migration and activation at the injury site. During kidney inflammation, the increased level of CCL21 that is overexpressed by preexisting LECs, along with other chemokine and integrin pathways, promotes kidney dLNs and spleen to recruit more CCR7+ immune cells through afferent lymphatic vessels [73, 74]. The significantly elevated recruitment leads to systemic expansion of lymphocytes [56]. Within kidney dLNs, CCR7+ DCs present antigens of injury sites to CD8+ T cells, promoting T cell proliferation in dLNs. After which, the activated CD8+ T cells return to injury tissue via blood circulation, releasing inflammatory cytokines including IFN-γ, TNF, TGF-β, and TonEBP (transcription factor tonicity-responsive enhancer-binding protein), and therefore aggravating the inflammatory infiltration and injury in the kidney. Also, it released inflammatory cytokines, further prompting kidney lymphangiogenesis and lymphatic, immune cell migration. This positive feedback between kidney dLNs and the injury tissue regulates the immune response in AKI. Both disrupting this loop (whether by removal of renal dLNs or inhibiting DCs recruitment and inhibiting kidney lymphangiogenesis can facilitate the progression of kidney injury [56, 59]. Besides DCs migration, T cell migration also plays an essential role in AKI. As regulatory T cells (Treg cells) reduce renal injury by inhibiting inflammation and facilitating tissue repair during AKI, obstructing the migration of Treg cells to dLNs reduces inflammation [75]. Therefore, lymphangiogenesis, which greatly promotes Treg cell migration from the injury site, can enhance the inflammation response, subsequently exacerbating tissue injury in AKI. Despite T cells, lymphangiogenesis can promote B cells egressing to dLNs [1], which may also contribute to the integral immune regulation impact of lymphangiogenesis in AKI.

Fig. 1
figure 1

Immune regulation roles of lymphangiogenesis in inflammation settings. a. During kidney inflammation, lymphangiogenesis is significantly induced, and LECs overexpress chemokine CCL21, which promotes recruitment of CCR7+ immune cells to kidney dLNs through lymphatic vessels. Increased migration of CCR7+ dendritic cells with antigen presented promotes antigen-specific CD8+ T cell proliferation and homing to inflammation site. These infiltrated CD8+ T cells released inflammatory cytokines including interferon γ (IFN-γ), tumor necrosis factor-α (TNF), transforming growth factor β (TGF-β) and transcription factor of tonicity-responsive enhancer-binding protein (TonEBP). These cytokines promotes macrophages and proximal tubular epithelial cells to express several factors including VEGF-C and VEGF-D that eventually further prompt kidney lymphangiogenesis. b. Constant inflammation microenvironment results in abnormally-structured lymphangiogenesis, which aggravates inflammation response in kidney. c. Immune regulation role of lymphangiogenesis functions differently in multiple pathological conditions, resulting in diverse immune microenvironments. In kidney fibrosis, reductions of B cells, Treg cells, IFN-γ-producing CD8+ T cells and CD11c+CD8+ T cells are shown. And in acute kidney injury, accumulations of Treg cells (Th17 cells) and local CD8+ T cells are inhibited. Of note, despite actively regulating immune cell migrations, during inflammation, infiltrated CD8+ T cells released IFN-γ, inducing PD-L1 expression by LECs, further inhibiting local CD8+ T cell effector function, reducing accumulation of local CD8+ T cell and alleviating kidney damage and progression of kidney fibrosis. Lymphangiogenesis significantly aids to this mechanism through enhanced immune cell trafficking. d. Lymphangiogenesis promote clearance of cellular debris, pro-inflammatory cytokines. In AKI, it significantly reduce the level of TGF-β to suppress the inflammatory response in kidney

To date, the integral effect of kidney lymphangiogenesis on AKI remains controversial. Despite protective or detrimental impacts, the argument that kidney lymphangiogenesis only acts as a passive response to AKI also exists. In an adriamycin-induced mouse model, it was proved that inhibited lymphangiogenesis did not affect renal inflammation and fibrosis progression [76]. Dynamic immune regulation mechanisms reacting to the different microenvironments of diverse AKI models and the duration of AKI may explain the contradictory outcomes of kidney lymphangiogenesis in AKI. Although these immune regulation mechanisms are widely triggered in a large number of diseases, evidence has suggested that lymphangiogenesis triggered by different stimuli differs in its immune regulation impacts to some extent. For instance, B cells-driven dLNs lymphangiogenesis significantly contributes to immune cell migration [77], while tumor-induced dLNs lymphangiogenesis mainly enhances lymph flow and metastasis [78, 79]. Nonetheless, currently, few studies focus on the complex mechanisms and the balance between opposite outcomes beneath AKI-induced lymphangiogenesis. Though lymphangiogenesis is found in different AKI models, it manifests divergent impacts on different models, including ischemia–reperfusion injury-induced, unilateral ureteral obstruction-induced and several toxin-induced models. However, its specific mechanisms are still unknown [80]. Given the varied pathogeneses underlying these models, future investigations need to elucidate lymphangiogenesis's distinct roles in these AKI models.

Additionally, since UUO can directly cause great urinary retention, which might confound the factors that trigger the initiation and maintenance of lymphangiogenesis, it might not be the ideal AKI model to study these intricate mechanisms involved in kidney lymphangiogenesis [48].

Hemolytic uraemic syndrome (HUS) is a group of disorders including AKI, thrombocytopenia and microangiopathic hemolytic anemia [81]. It is a rare but often life-threatening syndrome that various infective and non-infective reasons can induce. Shiga toxin-associated HUS is the most common type [82]. In general, all the non-infective types refer to atypical HUS, which is often associated with dysregulation of the complement system [83]. The consistent features of all types of HUS manifested in the kidney include aberrant immune cell populations and remarkable renal inflammation [81, 82, 84]. To date, classical treatments are limited to supportive options, and few targeted therapies are implemented in the clinic except anti-complement therapy. A recent study reported that ibrutinib and acalabrutinib (Bruton’s tyrosine kinase inhibitors) significantly reduced immune cell invasion and ameliorated disease progression [82]. Bruton’s tyrosine kinase inhibitor is crucial for innate immune response by regulating the recruitment and function of immune cells [85]. Therapies focused on promoting the growth of lymphatic vessels may be a viable alternative approach due to the unique feature of highly increased immune cell recruitment in the kidney. In contrast to other AKI scenarios, the process of lymphangiogenesis may significantly enhance the movement of immune cells, which in turn worsens the inflammatory response in the kidney. However, more studies are necessary due to limited research, especially on lymphangiogenesis in HUS.

Further studies are urgently needed to provide a clearer acknowledge of how newly synthesized lymphatic vessels act as an immune switch, specifically in the setting of AKI, which can yield novel insights to alleviate AKI through utilizing protective aspect of kidney lymphangiogenesis or avoiding detrimental actions of kidney lymphangiogenesis.

General function of lymphangiogenesis in chronic kidney disease

DN, IgAN, and LN are significant contributors to the progression of chronic kidney disease (CKD) and subsequent renal fibrosis [36]. CKD is a progressive condition characterized by gradually losing kidney function over time. The key pathological features of CKD are renal fibrosis and inflammation, which involve the excessive accumulation of extracellular matrix proteins in the kidney, leading to scarring and structural damage [40]. In CKD patients, diverse types of immune cells are infiltrated, mostly including macrophages, T cells, DCs, and mast cells [86]. The protective function of lymphangiogenesis is primarily exhibited through the clearance of local inflammatory factors and immune cells in the kidney.

Lymphangiogenesis reduces macrophage infiltration at the injury site and decreases the level of TGF-β, subsequently relieving intrarenal immune response and retarding the fibrosis progression [87]. Unlike AKI, M2 macrophages are the predominant phenotype within kidney fibrosis, which promotes tissue fibrosis in chronic kidney disease and kidney fibrosis [57]. Additionally, TGF-β serves as a master regulator during the progressive process of CKD. Physiologically, TGF-β in the kidney is responsible for the maturation of immune cells and regulating immune tolerance and response. However, overexpression of TGF-β induced by kidney inflammation disrupts the immune balance and accelerates the progression of kidney fibrosis. Additionally, lymphatic retention and interstitial fluid accumulation also increase expression level of TGF-β [88]. Therefore, clearing excessive fluid through kidney lymphangiogenesis also reduces TGF-β, which subsequently activates immune cells, including macrophages, reducing the progression of kidney fibrosis.

Besides clearing overexpressed TGF-β and infiltrated macrophages, recent studies elucidate lymphangiogenesis’s important immune regulation role by removing B cells in the kidney to mitigate fibrosis. B cells can affect kidney fibrosis through cytokine production and interactions with macrophages, T cells, and fibroblasts. Accumulating evidence demonstrates that depletion of B cells exhibits a protective effect towards kidney fibrosis in animal models. B cell-deficient mice were resistant to UUO-induced renal interstitial fibrosis [89]. Infiltrating B cells in kidney lesions exacerbate fibrosis by secreting various chemokines, including CCL2 and chemokine CCL7. Evidence demonstrates that enhanced recruitment of B cells in renal tissue exacerbates CKD via increasing macrophage infiltration [89], inhibiting T cell differentiation and activation [90, 91]. In CKD progress, lymphangiogenesis promotes B cell's egress to dLNs, decreasing B cell accumulation in the kidney. However, whether lymphangiogenesis can protect the kidney directly through reducing renal B cell accumulation and profibrotic chemokines secreted by B cells remains to be established. At first, overexpression of CCL21 by newly formed lymphatic vessels enhances the recruitment of CCR7+ DCs in dLNs. It eventually systematically promotes antigen-specific CD8+ T cells infiltration and kidney inflammation response, further inducing lymphangiogenesis. This chronic and systematic immune response significantly aggravates the inflammation and fibrosis progression. Suppressing the recruitment of CCR+7 DCs alleviates the infiltration of inflammatory cells infiltration and the progression of kidney fibrosis [16].

Reducing local immune cells due to lymphangiogenesis can also be maleficent in kidney fibrosis, inducing CKD. Studies have demonstrated that Treg cells protect the kidney against fibrosis progress [92], while Tγδ cells, Th17 cells, and CD4+ T cells present a profibrotic effect on the injured site [93, 94]. Therefore, reducing these anti-fibrotic immune cells due to kidney lymphangiogenesis can accelerate the progression of kidney fibrosis. Contrary to conventional acknowledge, subsets of CD8+ T cells, including IFN-γ-producing CD8+ T cells and CD11c+CD8+ T cells, also exert an anti-fibrotic and renal protective role in the kidney fibrosis setting. IFN-γ-producing CD8+ T cells inhibit the differentiation of CD4+ T cells into Th2 cells, subsequently controlling kidney inflammation and fibrosis, while CD11c+CD8+ T cells induce fibroblast apoptosis in obstructed kidney disease. These CD8+ T cells reduce myofibroblasts accumulation, which is one of the principal pathologies of CKD [95, 96].

Besides, sustained inflammation in the kidney may eventually result in the abnormal structure of newly synthesized lymphatic vessels [97], which destructs lymphatic vessels' functions, aggregating the progression of kidney inflammation and fibrosis (Fig. 1).

Diabetic kidney disease

Chronic hyperglycemia can lead to progressive diabetic kidney disease (DKD), which is the leading cause of end-stage renal disease (ESKD) in many countries. Excessive lipid accumulation in kidney tissue stimulates the expression of TGF-β and TNF-α, consequently resulting in an inflammatory response, eventually leading to severe diabetic renal damage [98].

As for the reason of lymphangiogenesis in DKD patients, it occurs due to a hyperglycemia-induced pro-inflammatory environment [99, 100], which creates a positive feedback loop between kidney tissue and dLNs, leading to an intensified inflammatory response in the kidneys [56, 74]. Additionally, the markedly elevated expression of VEGF-C in hyperglycemic conditions [53], while excessive ROS production from lipotoxicity induces apoptotic cell death, damaging the lymphatic endothelium and further promoting abnormal lymphangiogenesis [101].

In the context of the immune mechanisms of lymphangiogenesis in DKD, DC cells and macrophages play an indispensable role. Under hyperglycemia, the amount of DCs greatly increases, and danger-associated molecule patterns (DAMPs) interact with pattern recognition receptors on kidney DCs, activating CD8+ T cells, a feedback loop between kidney lymphangiogenesis and immune response. The macrophage population increases heavily in the glomeruli and tubulointerstitial within human type 2 diabetes. The intrarenal macrophages that were recruited primarily underwent polarization towards the M1 subset. This polarization resulted in heightened expressions of both systemic and renal cytokines, such as MCP-1 and TNF-α. Consequently, neo-lymphatic vessels developed, leading to the ultimate progression of glomerulosclerosis and tubulointerstitial fibrosis. Studies have demonstrated that macrophage infiltration prevention alleviates DKD progression [102].

In DKD condition, the newly synthesized lymphatic vessels in the kidney are dilated, with characteristics of hypertonicity and aberrant functions [97]. Notably, abnormal lymphangiogenesis might also partly damage preexisting lymphatic vessels’ function [99].

IgA nephropathy

Current studies have suggested that VEGF can manifest protective or detriment effects in glomerulonephritis [103,104,105]. These studies mainly focused on the roles of VEGF-induced angiogenesis in diverse chronic glomerulonephritis models; nonetheless, only a few studies targeted lymphangiogenesis. A recent study has provided evidence of a correlation between kidney lymphangiogenesis and clinical outcomes of IgA [106]. The increased lymphatic vessel density significantly correlates with more serious renal function injury and earlier progression to ESKD [48, 106]. Previous studies proved that the density of lymphatic vessels manifested as a promising prognostic value to predict the risk of ESKD for IgAN patients [106]. Similar to IgAN, a recent study demonstrated that the increased kidney lymphatic vessel density correlated with poor outcomes in crescentic glomerulonephritis [107]. Research has suggested that increased immune cell infiltration in crescentic glomerulonephritis is highly associated with kidney lymphangiogenesis [107, 108]. However, whether increased infiltration of immune cells induced by lymphangiogenesis plays a crucial part in IgAN remains uninvestigated. Emerging evidence has unveiled attributions of various immune cells, including Th cells, Treg cells, follicular helper T cells, and B cells, to the pathology of IgAN [109,110,111,112,113,114]. Within IgAN, these lymphocytes exert abnormal functions, which are significantly involved in the pathogenesis of IgAN, aggravating kidney inflammation and injury [111, 114, 115].

Dysfunctional newly synthesized lymphatic vessels contribute to the maleficent role of lymphangiogenesis in IgAN, similar to hyperglycemia-induced kidney disease. In addition, macrophages contribute to abnormal lymphangiogenesis in IgA glomerulonephritis. Emerging evidence demonstrates that CD137 ligand (CD137L)-secreted macrophages are present in IgA nephropathy, similar to other chronic kidney inflammatory diseases. CD137L interacts with CD137 on lymphatic endothelial cells, inducing lymphatic autophagy and lymphangiogenesis [116]. It may significantly contribute to the dysfunctional kidney lymphatic vessels, resulting in loss of transportation of inflammatory-associated molecules and obstruction of lymphatic routes for immune cell migration. Eventually, this CD137L-CD137 pathway drives fibrogenic responses, resulting in kidney fibrosis.

Lupus nephritis

Lupus nephritis is significantly associated with the progression of kidney inflammation and fibrosis. Still, only a few studies have focused on lymphangiogenesis's specific role in lupus nephritis. Inhibition of lymphangiogenesis in a mouse model of lupus nephritis (LN) distinctly alleviated the severity of the disease, but the effect of lymphangiogenesis was confounded in this model [117]. Additionally, a recent study found that kidney lymphangiogenesis induced the trafficking of LN-specific Mono/MΦ to both the entry and exit of the injured lesion [118]. The maleficent effect of lymphangiogenesis is probably related to the positive feedback between the renal inflammation site and the dLN, as mentioned above. However, whether the protective role of lymphangiogenesis can alleviate lupus nephritis remains unclear. In line with other chronic kidney diseases, lymphangiogenesis may mediate kidney injury through the clearance of immune and inflammation-related molecules and mediation in immune cell trafficking.

Hypertensive nephropathy

Patients and animal models with hypertension manifest a substantial increase of activated immune cells in the kidney [14, 119,120,121,122,123,124,125,126]. Infiltration of activated macrophages, DCs, B cells, and T cells distinctly aggravates renal injury and fibrosis, exacerbating sodium retention and ulteriorly elevating blood pressure [64, 127]. In general, the inflammatory response in the kidney, which is triggered by hypertension, further deteriorates both kidney function and hypertension condition [120].

In the setting of HTN, inflammation-associated kidney lymphangiogenesis is significantly induced [14, 119, 120, 128,129,130]. Previous studies demonstrated that HTN stimuli indirectly promote lymphangiogenesis instead of prompting LECs proliferation. HTN stimuli interact with various immune cell-secreted factors, sprouting lymphatic vessels [131]. Notably, increased extracellular ions in the kidney may directly activate macrophages to facilitate kidney lymphangiogenesis in hypertensive conditions. The mechanism above was discovered earlier in the dermis interstitium. Studies have demonstrated that increased osmolarity and extracellular salts within the skin directly activate TonEBP in macrophages and DCs, further inducing macrophages to express VEGF-C, which promotes lymphangiogenesis.

Given the critical role of lymphangiogenesis in fluid clearance and the significantly increased kidney lymphangiogenesis in HTN models, the interaction between the lymphatic system and HTN has drawn much attention. Furthermore, despite newly synthesized lymphatics' function in modulating renal fluid homeostasis, recent evidence supports that lymphangiogenesis can also influence HTN by regulating the immune response in the kidney. Enhancement of kidney lymphangiogenesis exerts a protective effect against hypertension, reducing renal immune cell accumulation and alleviating inflammation [14, 45, 120, 128, 130, 132, 133]. Despite clearance of excessive fluid, lymphangiogenesis also elevates drainage of infiltrated immune cells and pro-inflammatory cytokines secreted by these cells. In several hypertension mouse models with kidney-specific overexpression of VEGF-D (KiDVD+ mouse model), excessively accumulated immune cells, including macrophages in the kidney, were all reduced, subsequently preventing hypertension [132]. Furthermore, in hypertension conditions, kidney lymphangiogenesis also actively regulates the migration of immune cells, including macrophages, DCs, and T cells, via increased secretion of CCL21 and CCR7. An angiotensin II-induced hypertension (A2HTN) mouse model study has proved that lymphangiogenesis significantly reduces the CD11c+F4/80 monocyte renal population [130]. Activated monocytes in the kidney express pro-inflammatory cytokines and mediate T-cell activation and differentiation. Naïve T cells differentiate into Th1 or Th17 cells, which secrete pro-inflammatory and pro-hypertensive cytokines, causing sodium retention and hypertension [134]. Thus, reducing these monocyte populations in kidney attributes inhibits excessive inflammation response in the kidney due to hypertension.

However, evidence supports that renal-specific lymphangiogenesis cannot fully rescue kidney hypertensive condition but can only alleviate systemic blood pressure [135]. Additionally, as Treg cells can inhibit inflammation response and improve sodium retention within HTN [75, 136], in line with other kidney diseases, lymphangiogenesis-induced removal of Treg cells might aggravate kidney inflammation and hypertension conditions.

In the HTN setting, kidney lymphangiogenesis is limited in renal immune cell trafficking [132]. Additionally, due to current evidence, an uneven outcome in the transport level of different immune cell populations by newly formed renal lymphatic vessels has been discovered in hypertension models. The great involvement of T cells and M1 macrophages in hypertensive kidneys possibly results in limited transferring of these cells from kidney tissues through kidney lymphangiogenesis, compared with other renal immune cell populations.

Interestingly, unlike other kidney diseases, in hypertension condition, kidney lymphangiogenesis can also directly regulate immune cell activation through sodium transport (Fig. 2). Recent study has revealed that kidney lymphangiogenesis directly suppresses activation and accumulation of DCs through reducing Na+ retention, consequently relieving hypertension and mitigating the progression of HTN [132]. As Na+ stimulation can activate DCs [137], reduction of Na+ retention through enhanced kidney lymphangiogenesis can directly inhibit DCs activation.

Fig. 2
figure 2

Distinct mechanisms of lymphangiogenesis in certain kidney disease. a. In kidney hypertensive disease, sodium retention induces lymphangiogenesis through a Na +—TonEBP—VEGF-C pathway. Na + directly activates transcription factor of tonicity responsive enhancer-binding protein (TonEBP) in macrophages and dendritic cells (DCs) to promote expression of VEGF-C from macrophages, and then induces lymphangiogenesis. Sodium retention can directly activate DCs to express cytokines for further antigen-specific T cell accumulation and activation. Na + enters dendritic cells, subsequently leading to Ca2 + influx and then activation of protein kinase C, eventually resulting in increased expression of reactive oxygen species (ROS). ROS oxidates fatty acids into isolevuglandins (IsoLGs), which activates dendritic cells to produce proinflammatory cytokines (IL-1β, IL-6, IL-23) and activate T cells to proliferate and express inflammatory cytokines including TNF, IFN-γ and TGF-β. Lymphangiogenesis can reduce sodium retention, therefore inhibits DCs activation and the inflammatory response. b. In diabetic kidney disease, excessive cytokines expressed during the chronic inflammation condition create a specific microenvironment, which significantly induces abnormally-structured lymphangiogenesis

Polycystic kidney disease

Unlike the early stage of kidney inflammation, M2 macrophages are predominant macrophages within PKD, which can promote tissue repair and are ultimately profibrotic [138]. Of note, M2 macrophages distinctly enhance cyst enlargement in PKD [139]. Studies have demonstrated that kidney lymphangiogenesis can remodel vessel structure, expand lymphatics to transport accumulated fluid in cysts and inhibit cyst progression [140]. However, few study concentrates on the transportation of local immune cells or the immune regulatory function of lymphangiogenesis within PKD. In line with other kidney diseases, lymphangiogenesis may alleviate the progression of PKD by transporting inflammatory cells and reducing M2 macrophage infiltration surrounding the cysts. However, available evidence has indicated a protective role of CD8+ T cells in the context of PKD [141]. Further studies are required to investigate the immune regulatory role of kidney lymphangiogenesis and its overall impact on PKD.

Kidney transplantation

After kidney transplantation, the mutual interaction between the allograft and the recipient’s immune system may generate a series of immune responses, resulting in transplant rejection. Immune cells are trafficked between the kidney allograft and the recipient’s original system via blood circulation and newly formed lymphatic vessels [142, 143]. Studies with conflicting results have revealed that lymphangiogenesis can both serve a protective role and a maleficent role in transplant rejection. Kidney allografts with greater density of kidney lymphatic vessels are less likely to generate renal interstitial fibrosis and renal tubule atrophy [144]. Likewise, promoting lymphangiogenesis in the kidney allograft significantly alleviates transplant rejection and extends the mice recipient's survival time [145]. Vigorous neo-synthesized lymphatic vessels transport inflammatory cells, clearing interstitial edema and subsequently protecting the kidney allograft [145,146,147]. In line with acute kidney injury, lymphatic regulation of immune cells via lymphangiogenesis protects recipients from transplant rejection. Kidney lymphangiogenesis can directly suppress local CD8+ T cell's immune response in kidney allograft via PD-L1, which is secreted by LECs [148]. In contrast to AKI, lymphatic migration of Treg cells may suppress allograft rejection [149].

Inversely, several studies have suggested that the expansion extent of lymphatic vessels positively correlates with the severity of transplant rejection [144, 150, 151]. In addition, it is demonstrated that inhibiting lymphangiogenesis can mitigate the injury of allografts [152], while ligation of lymphatic vessels benefits transplant rejection. Neo-synthesized lymphatic vessels have an abundant accumulation of CD 45+ lymphocytes, mainly MHCII+, ED-1, IDO, HIS13, and CD103 antigen-present cells. The increase of these antigen-presenting cells can exacerbate injury of transplant rejection in kidney allografts [153]. Increasing recruitment of antigen-presenting cells in recipient dLNs initiates the alloimmune response, leading to inflammatory cell infiltration in the kidney allograft and further destruction of the structure and function of the allograft [154].

Conclusion

Despite an increased emphasis on the immunological regulatory function of lymphangiogenesis in renal disorders, there are still notable gaps in our understanding. A substantial body of evidence indicates that kidney lymphangiogenesis plays a significant part in immunological regulation, with its effects being either beneficial or detrimental depending on the specific kidney condition under consideration. Recent studies have elucidated several immunological functions of lymphatic veins for specific kidney disorders. Nevertheless, existing research predominantly examines renal inflammation, fibrosis, and a restricted range of prevalent kidney disorders, such as HTN and DKD. Furthermore, these investigations primarily rely on animal models as the primary means of study. Moreover, a more comprehensive examination is required to elucidate the underlying factors contributing to the varied impacts of lymphangiogenesis on distinct renal disorders. There is an urgent need for a more extensive comprehension of the immunological regulatory processes underlying renal lymphangiogenesis within the framework of kidney disease. Examining these complex pathways can yield new perspectives in developing therapeutic interventions that specifically target the beneficial aspects of lymphangiogenesis or reduce its detrimental impact on renal disorders.

Availability of data and materials

Not applicable.

Abbreviations

LEC:

Lymphatic endothelial cell

AKI:

Acute kidney injury

CKD:

Chronic kidney disease

DC:

Dendritic cell

dLNs:

Draining lymph nodes

DKD:

Diabetic kidney disease

ESKD:

End-stage kidney disease

HUS:

Haemolytic uraemic syndrome

VEGF-C:

Vascular endothelial growth factor C

VEGF-D:

Vascular endothelial growth factor D

VEGF-3:

Vascular endothelial growth factor receptor 3

VEGFR3:

Vascular endothelial growth factor receptor 3

NRP2:

Neuropilin 2

TGF-β:

Transforming growth factor β

CTGF:

Connective tissue growth factor

LYVE-1:

Lymphatic vessel endothelial receptor-1

A2HTN:

Angiotensin II-induced HTN

CCR7:

Chemokine (C–C motif) receptor 7

CCL19:

Chemokine (C–C motif) ligand 19

CXCL:

Chemokine (C-X-C motif) ligand

CCL:

Chemokine (C–C motif) ligand

ICAM1-1:

Intercellular adhesion molecule 1

VCAM-1:

Vascular cell adhension molecule

VE-Cadherin:

Vascular endothelial cadherin

IL-1β:

Interleukin-1β

TNF-α:

Tumor necrosis factor-α

IL-10:

Interleukin-10

TonEBP:

Transcription factor tonicity-responsive enhancer-binding protein

HEV:

High endothelial venule

HTN:

Hypertensive nephropathy

IgAN:

IgA nephropathy

IFN-γ:

Interferon γ

LHTN:

L-NAME-induced HTN

PKD:

Polycystic kidney disease

TRCM cells:

Recirculating memory T cells

Treg cell:

Regulatory T cell

SSHTN:

Salt-sensitive HTN

S1P:

Sphingosine-1-phosphate

TCR:

T cell antigen receptor

Th cell:

T helper cell

Th1 cell:

T helper cell 1

Th17 cell:

T helper cell 17

PD-L1:

Programmed death-ligand 1

UUO:

Unilateral ureteral obstruction

ROS:

Reactive oxygen species

DAMPs:

Danger-associated molecule patterns

CD137L:

CD137 ligand

References

  1. Hampton HR, Chtanova T. Lymphatic migration of immune cells. Front Immunol. 2019;10:1168. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2019.01168.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Mou R, Chen K, Zhu P, Xu Q, Ma L. The impact of stem/progenitor cells on lymphangiogenesis in vascular disease. Cells. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells11244056.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Zhang Y, Zhang C, Li L, Liang X, Cheng P, Li Q, et al. Lymphangiogenesis in renal fibrosis arises from macrophages via VEGF-C/VEGFR3-dependent autophagy and polarization. Cell Death Dis. 2021;12(1):109. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-020-03385-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Alitalo K, Carmeliet P. Molecular mechanisms of lymphangiogenesis in health and disease. Cancer Cell. 2002;1(3):219–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s1535-6108(02)00051-x.

    Article  CAS  PubMed  Google Scholar 

  5. Ducoli L, Detmar M. Beyond PROX1: transcriptional, epigenetic, and noncoding RNA regulation of lymphatic identity and function. Dev Cell. 2021;56(4):406–26. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.devcel.2021.01.018.

    Article  CAS  PubMed  Google Scholar 

  6. Liu J, Yu C. Lymphangiogenesis and lymphatic barrier dysfunction in renal fibrosis. Int J Mol Sci. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms23136970.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Maruccia M, Giudice G, Ciudad P, Manrique OJ, Cazzato G, Chen HC, et al. Lymph node transfer and neolymphangiogenesis: from theory to evidence. Plast Reconstr Surg. 2023;152(5):904e–12e. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/PRS.0000000000010434.

    Article  CAS  PubMed  Google Scholar 

  8. Oliver G, Kipnis J, Randolph GJ, Harvey NL. The lymphatic vasculature in the 21(st) century: novel functional roles in homeostasis and disease. Cell. 2020;182(2):270–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2020.06.039.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hu Z, Zhao X, Wu Z, Qu B, Yuan M, Xing Y, et al. Lymphatic vessel: origin, heterogeneity, biological functions, and therapeutic targets. Signal Transduct Target Ther. 2024;9(1):9. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-023-01723-x.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Liao S, von der Weid PY. Lymphatic system: an active pathway for immune protection. Semin Cell Dev Biol. 2015;38:83–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcdb.2014.11.012.

    Article  CAS  PubMed  Google Scholar 

  11. Kataru RP, Jung K, Jang C, Yang H, Schwendener RA, Baik JE, et al. Critical role of CD11b+ macrophages and VEGF in inflammatory lymphangiogenesis, antigen clearance, and inflammation resolution. Blood. 2009;113(22):5650–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2008-09-176776.

    Article  CAS  PubMed  Google Scholar 

  12. Donnan MD, Kenig-Kozlovsky Y, Quaggin SE. The lymphatics in kidney health and disease. Nat Rev Nephrol. 2021;17(10):655–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41581-021-00438-y.

    Article  PubMed  Google Scholar 

  13. Stasi E, Sciascia S, Naretto C, Baldovino S, Roccatello D. Lymphatic system and the kidney: from lymphangiogenesis to renal inflammation and fibrosis development. Int J Mol Sci. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms25052853.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Balasubbramanian D, Lopez Gelston CA, Rutkowski JM, Mitchell BM. Immune cell trafficking, lymphatics and hypertension. Br J Pharmacol. 2019;176(12):1978–88. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bph.14370.

    Article  CAS  PubMed  Google Scholar 

  15. Baker ML, Cantley LG. The lymphatic system in kidney disease. Kidney360. 2023;4(6):e841–50. https://doiorg.publicaciones.saludcastillayleon.es/10.3406/KID.0000000000000120.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Wu J, Pei G, Zeng R, Xu G. Lymphatic vessels enhancing adaptive immunity deteriorates renal inflammation and renal fibrosis. Kidney Dis. 2020;6(3):150–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1159/000506201.

    Article  Google Scholar 

  17. Russell PS, Hong J, Windsor JA, Itkin M, Phillips ARJ. Renal lymphatics: anatomy, physiology, and clinical implications. Front Physiol. 2019;10:251. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphys.2019.00251.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Xu J, Ma X, Yu K, Wang R, Wang S, Liu R, et al. Lactate up-regulates the expression of PD-L1 in kidney and causes immunosuppression in septic acute renal injury. J Microbiol Immunol Infect. 2021;54(3):404–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jmii.2019.10.006.

    Article  CAS  PubMed  Google Scholar 

  19. Wang W, Li X, Ding X, Xiong S, Hu Z, Lu X, et al. Lymphatic endothelial transcription factor Tbx1 promotes an immunosuppressive microenvironment to facilitate post-myocardial infarction repair. Immunity. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.immuni.2023.07.019.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Podgrabinska S, Kamalu O, Mayer L, Shimaoka M, Snoeck H, Randolph GJ, et al. Inflamed lymphatic endothelium suppresses dendritic cell maturation and function via Mac-1/ICAM-1-dependent mechanism. J Immunol. 2009;183(3):1767–79.

    Article  CAS  PubMed  Google Scholar 

  21. Angeli V, Lim HY. Biomechanical control of lymphatic vessel physiology and functions. Cell Mol Immunol. 2023;20(9):1051–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41423-023-01042-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Weavers H, Martin P. The cell biology of inflammation: from common traits to remarkable immunological adaptations. J Cell Biol. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1083/jcb.202004003.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Jalkanen S, Salmi M. Lymphatic endothelial cells of the lymph node. Nat Rev Immunol. 2020;20(9):566–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41577-020-0281-x.

    Article  CAS  PubMed  Google Scholar 

  24. Johnson LA, Jackson DG. Hyaluronan and its receptors: key mediators of immune cell entry and trafficking in the lymphatic system. Cells. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells10082061.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Lämmermann T, Bader BL, Monkley SJ, Worbs T, Wedlich-Söldner R, Hirsch K, et al. Rapid leukocyte migration by integrin-independent flowing and squeezing. Nature. 2008;453(7191):51–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature06887.

    Article  CAS  PubMed  Google Scholar 

  26. Lämmermann T, Sixt M. Mechanical modes of ‘amoeboid’ cell migration. Curr Opin Cell Biol. 2009;21(5):636–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ceb.2009.05.003.

    Article  CAS  PubMed  Google Scholar 

  27. Nourshargh S, Hordijk PL, Sixt M. Breaching multiple barriers: leukocyte motility through venular walls and the interstitium. Nat Rev Mol Cell Biol. 2010;11(5):366–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrm2889.

    Article  CAS  PubMed  Google Scholar 

  28. Kel JM, Girard-Madoux MJ, Reizis B, Clausen BE. TGF-beta is required to maintain the pool of immature langerhans cells in the epidermis. J Immunol. 2010;185(6):3248–55. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.1000981.

    Article  CAS  PubMed  Google Scholar 

  29. Imai K, Minamiya Y, Koyota S, Ito M, Saito H, Sato Y, et al. Inhibition of dendritic cell migration by transforming growth factor-β1 increases tumor-draining lymph node metastasis. J Exp Clin Cancer Res. 2012;31(1):3. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1756-9966-31-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cumberbatch M, Clelland K, Dearman RJ, Kimber I. Impact of cutaneous IL-10 on resident epidermal langerhans’ cells and the development of polarized immune responses. J Immunol. 2005;175(1):43–50. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.175.1.43.

    Article  CAS  PubMed  Google Scholar 

  31. Roozendaal R, Mempel TR, Pitcher LA, Gonzalez SF, Verschoor A, Mebius RE, et al. Conduits mediate transport of low-molecular-weight antigen to lymph node follicles. Immunity. 2009;30(2):264–76. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.immuni.2008.12.014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Gretz JE, Norbury CC, Anderson AO, Proudfoot AE, Shaw S. Lymph-borne chemokines and other low molecular weight molecules reach high endothelial venules via specialized conduits while a functional barrier limits access to the lymphocyte microenvironments in lymph node cortex. J Exp Med. 2000;192(10):1425–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1084/jem.192.10.1425.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sixt M, Kanazawa N, Selg M, Samson T, Roos G, Reinhardt DP, et al. The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity. 2005;22(1):19–29. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.immuni.2004.11.013.

    Article  CAS  PubMed  Google Scholar 

  34. Kähäri L, Fair-Mäkelä R, Auvinen K, Rantakari P, Jalkanen S, Ivaska J, et al. Transcytosis route mediates rapid delivery of intact antibodies to draining lymph nodes. J Clin Invest. 2019;129(8):3086–102. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci125740.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Jakubzick C, Bogunovic M, Bonito AJ, Kuan EL, Merad M, Randolph GJ. Lymph-migrating, tissue-derived dendritic cells are minor constituents within steady-state lymph nodes. J Exp Med. 2008;205(12):2839–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1084/jem.20081430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chebotareva N, Vinogradov A. Different Types of Chronic Kidney Disease (CKD) You Need To Know. Chinese Medicine. 2024.

  37. Ji R-C. The role of lymphangiogenesis in cardiovascular diseases and heart transplantation. Heart Fail Rev. 2022;27(5):1837–56.

    Article  PubMed  Google Scholar 

  38. Liu P, Ding P, Sun C, Chen S, Lowe S, Meng L, et al. Lymphangiogenesis in gastric cancer: function and mechanism. Eur J Med Res. 2023;28(1):405.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Mueller A, Zhao Y, Cicek H, Paust H-J, Sivayoganathan A, Linke A, et al. Transcriptional and clonal characterization of cytotoxic t cells in crescentic glomerulonephritis. J Am Soc Nephrol. 2023;34(6):1003–18.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Reiss AB, Jacob B, Zubair A, Srivastava A, Johnson M, De Leon J. Fibrosis in chronic kidney disease: pathophysiology and therapeutic targets. J Clin Med. 2024;13(7):1881. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/jcm13071881.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Steele MM, Lund AW. Afferent lymphatic transport and peripheral tissue immunity. J Immunol. 2021;206(2):264–72.

    Article  CAS  PubMed  Google Scholar 

  42. Tanabe K, Wada J, Sato Y. Targeting angiogenesis and lymphangiogenesis in kidney disease. Nat Rev Nephrol. 2020;16(5):289–303. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41581-020-0260-2.

    Article  CAS  PubMed  Google Scholar 

  43. Glinton KE, Ma W, Lantz C, Grigoryeva LS, DeBerge M, Liu X, et al. Macrophage-produced VEGFC is induced by efferocytosis to ameliorate cardiac injury and inflammation. J Clin Invest. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci140685.

    Article  PubMed  PubMed Central  Google Scholar 

  44. D’Amore PA, Alcaide P. Macrophage efferocytosis with VEGFC and lymphangiogenesis: rescuing the broken heart. J Clin Invest. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci158703.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Beaini S, Saliba Y, Hajal J, Smayra V, Bakhos JJ, Joubran N, et al. VEGF-C attenuates renal damage in salt-sensitive hypertension. J Cell Physiol. 2019;234(6):9616–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jcp.27648.

    Article  CAS  PubMed  Google Scholar 

  46. Cui T, Feng C, Jiang H, Jin Y, Feng J. Inhibition of PFKFB3 expression stimulates macrophage-mediated lymphangiogenesis post-acute myocardial infarction. Front Biosci. 2023;28(11):277. https://doiorg.publicaciones.saludcastillayleon.es/10.31083/j.fbl2811277.

    Article  CAS  Google Scholar 

  47. Wang C, Yue Y, Huang S, Wang K, Yang X, Chen J, et al. M2b macrophages stimulate lymphangiogenesis to reduce myocardial fibrosis after myocardial ischaemia/reperfusion injury. Pharm Biol. 2022;60(1):384–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/13880209.2022.2033798.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zarjou A, Black LM, Bolisetty S, Traylor AM, Bowhay SA, Zhang MZ, et al. Dynamic signature of lymphangiogenesis during acute kidney injury and chronic kidney disease. Lab Invest. 2019;99(9):1376–88. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41374-019-0259-0.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Kinashi H, Ito Y, Sun T, Katsuno T, Takei Y. Roles of the TGF-β⁻VEGF-C pathway in fibrosis-related lymphangiogenesis. Int J Mol Sci. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms19092487.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Kinashi H, Falke LL, Nguyen TQ, Bovenschen N, Aten J, Leask A, et al. Connective tissue growth factor regulates fibrosis-associated renal lymphangiogenesis. Kidney Int. 2017;92(4):850–63.

    Article  CAS  PubMed  Google Scholar 

  51. Korhonen EA, Murtomaki A, Jha SK, Anisimov A, Pink A, Zhang Y, et al. Lymphangiogenesis requires Ang2/Tie/PI3K signaling for VEGFR3 cell-surface expression. J Clin Invest. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI155478.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Fatima A, Wang Y, Uchida Y, Norden P, Liu T, Culver A, et al. Foxc1 and Foxc2 deletion causes abnormal lymphangiogenesis and correlates with ERK hyperactivation. J Clin Invest. 2016;126(7):2437–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI80465.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Sakamoto I, Ito Y, Mizuno M, Suzuki Y, Sawai A, Tanaka A, et al. Lymphatic vessels develop during tubulointerstitial fibrosis. Kidney Int. 2009;75(8):828–38. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ki.2008.661.

    Article  CAS  PubMed  Google Scholar 

  54. Zhang Y, Lu Y, Ma L, Cao X, Xiao J, Chen J, et al. Activation of vascular endothelial growth factor receptor-3 in macrophages restrains TLR4-NF-κB signaling and protects against endotoxin shock. Immunity. 2014;40(4):501–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.immuni.2014.01.013.

    Article  CAS  PubMed  Google Scholar 

  55. Guo YC, Zhang M, Wang FX, Pei GC, Sun F, Zhang Y, et al. Macrophages regulate unilateral ureteral obstruction-induced renal lymphangiogenesis through C-C motif chemokine receptor 2-dependent phosphatidylinositol 3-kinase-AKT-mechanistic target of rapamycin signaling and hypoxia-inducible factor-1α/vascular endothelial growth factor-C expression. Am J Pathol. 2017;187(8):1736–49. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ajpath.2017.04.007.

    Article  CAS  PubMed  Google Scholar 

  56. Pei G, Yao Y, Yang Q, Wang M, Wang Y, Wu J, et al. Lymphangiogenesis in kidney and lymph node mediates renal inflammation and fibrosis. Sci Adv. 2019;5(6):5075. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.aaw5075.

    Article  CAS  Google Scholar 

  57. Tang PM, Nikolic-Paterson DJ, Lan HY. Macrophages: versatile players in renal inflammation and fibrosis. Nat Rev Nephrol. 2019;15(3):144–58. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41581-019-0110-2.

    Article  PubMed  Google Scholar 

  58. Kerjaschki D, Huttary N, Raab I, Regele H, Bojarski-Nagy K, Bartel G, et al. Lymphatic endothelial progenitor cells contribute to de novo lymphangiogenesis in human renal transplants. Nat Med. 2006;12(2):230–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nm1340.

    Article  CAS  PubMed  Google Scholar 

  59. Kasinath V, Yilmam OA, Uehara M, Jiang L, Ordikhani F, Li X, et al. Activation of fibroblastic reticular cells in kidney lymph node during crescentic glomerulonephritis. Kidney Int. 2019;95(2):310–20. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.kint.2018.08.040.

    Article  CAS  PubMed  Google Scholar 

  60. Kinashi H, Falke LL, Nguyen TQ, Bovenschen N, Aten J, Leask A, et al. Connective tissue growth factor regulates fibrosis-associated renal lymphangiogenesis. Kidney Int. 2017;92(4):850–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.kint.2017.03.029.

    Article  CAS  PubMed  Google Scholar 

  61. Randolph GJ, Ivanov S, Zinselmeyer BH, Scallan JP. The lymphatic system: integral roles in immunity. Annu Rev Immunol. 2017;35:31–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev-immunol-041015-055354.

    Article  CAS  PubMed  Google Scholar 

  62. Creed HA, Rutkowski JM. Emerging roles for lymphatics in acute kidney injury: beneficial or maleficent? Exp Biol Med. 2021;246(7):845–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/1535370220983235.

    Article  CAS  Google Scholar 

  63. Kinsey GR, Okusa MD. Expanding role of T cells in acute kidney injury. Curr Opin Nephrol Hypertens. 2014;23(1):9–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/01.mnh.0000436695.29173.de.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Maisel K, Sasso MS, Potin L, Swartz MA. Exploiting lymphatic vessels for immunomodulation: rationale, opportunities, and challenges. Adv Drug Deliv Rev. 2017;114:43–59. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.addr.2017.07.005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Maeda Y, Seki N, Sato N, Sugahara K, Chiba K. Sphingosine 1-phosphate receptor type 1 regulates egress of mature T cells from mouse bone marrow. Int Immunol. 2010;22(6):515–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/intimm/dxq036.

    Article  CAS  PubMed  Google Scholar 

  66. Platt AM, Randolph GJ. Dendritic cell migration through the lymphatic vasculature to lymph nodes. Adv Immunol. 2013;120:51–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/b978-0-12-417028-5.00002-8.

    Article  CAS  PubMed  Google Scholar 

  67. Collett JA, Ortiz-Soriano V, Li X, Flannery AH, Toto RD, Moe OW, et al. Serum IL-17 levels are higher in critically ill patients with AKI and associated with worse outcomes. Crit Care. 2022;26(1):107. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13054-022-03976-4.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Pindjakova J, Hanley SA, Duffy MM, Sutton CE, Weidhofer GA, Miller MN, et al. Interleukin-1 accounts for intrarenal Th17 cell activation during ureteral obstruction. Kidney Int. 2012;81(4):379–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ki.2011.348.

    Article  CAS  PubMed  Google Scholar 

  69. Mehrotra P, Patel JB, Ivancic CM, Collett JA, Basile DP. Th-17 cell activation in response to high salt following acute kidney injury is associated with progressive fibrosis and attenuated by AT-1R antagonism. Kidney Int. 2015;88(4):776–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ki.2015.200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Lane RS, Femel J, Breazeale AP, Loo CP, Thibault G, Kaempf A, et al. IFNγ-activated dermal lymphatic vessels inhibit cytotoxic T cells in melanoma and inflamed skin. J Exp Med. 2018;215(12):3057–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1084/jem.20180654.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Jaworska K, Ratajczak J, Huang L, Whalen K, Yang M, Stevens BK, et al. Both PD-1 ligands protect the kidney from ischemia reperfusion injury. J Immunol. 2015;194(1):325–33. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.1400497.

    Article  CAS  PubMed  Google Scholar 

  72. Wang N, Jiang L, Zhu B, Wen Y, Xi XM. Fluid balance and mortality in critically ill patients with acute kidney injury: a multicenter prospective epidemiological study. Crit Care. 2015;19:371. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13054-015-1085-4.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Pei G, Yao Y, Yang Q, Wang M, Wang Y, Wu J, et al. Lymphangiogenesis in kidney and lymph node mediates renal inflammation and fibrosis. Sci Adv. 2019;5(6):5075. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.aaw5075.

    Article  CAS  Google Scholar 

  74. Snelgrove SL, Lo C, Hall P, Lo CY, Alikhan MA, Coates PT, et al. Activated renal dendritic cells cross present intrarenal antigens after ischemia-reperfusion injury. Transplantation. 2017;101(5):1013–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/tp.0000000000001427.

    Article  CAS  PubMed  Google Scholar 

  75. Menning A, Höpken UE, Siegmund K, Lipp M, Hamann A, Huehn J. Distinctive role of CCR7 in migration and functional activity of naive- and effector/memory-like treg subsets. Eur J Immunol. 2007;37(6):1575–83. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/eji.200737201.

    Article  CAS  PubMed  Google Scholar 

  76. Yazdani S, Hijmans RS, Poosti F, Dam W, Navis G, van Goor H, et al. Targeting tubulointerstitial remodeling in proteinuric nephropathy in rats. Dis Model Mech. 2015;8(8):919–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1242/dmm.018580.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Angeli V, Ginhoux F, Llodrà J, Quemeneur L, Frenette PS, Skobe M, et al. B cell-driven lymphangiogenesis in inflamed lymph nodes enhances dendritic cell mobilization. Immunity. 2006;24(2):203–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.immuni.2006.01.003.

    Article  CAS  PubMed  Google Scholar 

  78. Hirakawa S, Brown LF, Kodama S, Paavonen K, Alitalo K, Detmar M. VEGF-C-induced lymphangiogenesis in sentinel lymph nodes promotes tumor metastasis to distant sites. Blood. 2007;109(3):1010–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2006-05-021758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Harrell MI, Iritani BM, Ruddell A. Tumor-induced sentinel lymph node lymphangiogenesis and increased lymph flow precede melanoma metastasis. Am J Pathol. 2007;170(2):774–86. https://doiorg.publicaciones.saludcastillayleon.es/10.2353/ajpath.2007.060761.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Black LM, Winfree S, Khochare SD, Kamocka MM, Traylor AM, Esman SK, et al. Quantitative 3-dimensional imaging and tissue cytometry reveals lymphatic expansion in acute kidney injury. Lab Invest. 2021;101(9):1186–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41374-021-00609-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Michael M, Bagga A, Sartain SE, Smith RJH. Haemolytic uraemic syndrome. Lancet. 2022;400(10364):1722–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0140-6736(22)01202-8.

    Article  CAS  PubMed  Google Scholar 

  82. Kröller S, Wissuwa B, Dennhardt S, Krieg N, Thiemermann C, Daniel C, et al. Bruton’s tyrosine kinase inhibition attenuates disease progression by reducing renal immune cell invasion in mice with hemolytic-uremic syndrome. Front Immunol. 2023;14:1105181. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1105181.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Stenson EK, Kendrick J, Dixon B, Thurman JM. The complement system in pediatric acute kidney injury. Pediatr Nephrol. 2023;38(5):1411–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00467-022-05755-3.

    Article  PubMed  Google Scholar 

  84. Chen IR, Huang CC, Tu SJ, Wang GJ, Lai PC, Lee YT, et al. Dysregulation of immune cell subpopulations in atypical hemolytic uremic syndrome. Int J Mol Sci. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms241210007.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Purvis GSD, Collino M, Aranda-Tavio H, Chiazza F, O’Riordan CE, Zeboudj L, et al. Inhibition of Bruton’s TK regulates macrophage NF-κB and NLRP3 inflammasome activation in metabolic inflammation. Br J Pharmacol. 2020;177(19):4416–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bph.15182.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Meng XM, Nikolic-Paterson DJ, Lan HY. Inflammatory processes in renal fibrosis. Nat Rev Nephrol. 2014;10(9):493–503. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrneph.2014.114.

    Article  CAS  PubMed  Google Scholar 

  87. Hasegawa S, Nakano T, Torisu K, Tsuchimoto A, Eriguchi M, Haruyama N, et al. Vascular endothelial growth factor-C ameliorates renal interstitial fibrosis through lymphangiogenesis in mouse unilateral ureteral obstruction. Lab Invest. 2017;97(12):1439–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/labinvest.2017.77.

    Article  CAS  PubMed  Google Scholar 

  88. Zhang T, Guan G, Liu G, Sun J, Chen B, Li X, et al. Disturbance of lymph circulation develops renal fibrosis in rats with or without contralateral nephrectomy. Nephrology. 2008;13(2):128–38. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1440-1797.2007.00851.x.

    Article  CAS  PubMed  Google Scholar 

  89. 'Renal recruitment of B lymphocytes exacerbates tubulointerstitial fibrosis by promoting monocyte mobilization and infiltration after unilateral ureteral obstruction'. J Pathol. 2019;248(2):253. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/path.5272.

  90. Bar-Or A, Fawaz L, Fan B, Darlington PJ, Rieger A, Ghorayeb C, et al. Abnormal B-cell cytokine responses a trigger of T-cell-mediated disease in MS? Ann Neurol. 2010;67(4):452–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ana.21939.

    Article  CAS  PubMed  Google Scholar 

  91. Iwata S, Saito K, Tokunaga M, Yamaoka K, Nawata M, Yukawa S, et al. Phenotypic changes of lymphocytes in patients with systemic lupus erythematosus who are in longterm remission after B cell depletion therapy with rituximab. J Rheumatol. 2011;38(4):633–41. https://doiorg.publicaciones.saludcastillayleon.es/10.3899/jrheum.100729.

    Article  CAS  PubMed  Google Scholar 

  92. do Valle Duraes F, Lafont A, Beibel M, Martin K, Darribat K, Cuttat R, et al. Immune cell landscaping reveals a protective role for regulatory T cells during kidney injury and fibrosis. JCI Insight. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci.insight.130651.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Liu L, Kou P, Zeng Q, Pei G, Li Y, Liang H, et al. CD4+ T Lymphocytes, especially Th2 cells, contribute to the progress of renal fibrosis. Am J Nephrol. 2012;36(4):386–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1159/000343283.

    Article  CAS  PubMed  Google Scholar 

  94. Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 cells. Annu Rev Immunol. 2009;27:485–517. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev.immunol.021908.132710.

    Article  CAS  PubMed  Google Scholar 

  95. Gao M, Wang J, Zang J, An Y, Dong Y. The mechanism of CD8(+) T cells for reducing myofibroblasts accumulation during renal fibrosis. Biomolecules. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biom11070990.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Wang H, Wang J, Bai Y, Li J, Li L, Dong Y. CD11c⁺ CD8⁺ T cells reduce renal fibrosis following ureteric obstruction by inducing fibroblast apoptosis. Int J Mol Sci. 2016. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms18010001.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Kajiya K, Hirakawa S, Detmar M. Vascular endothelial growth factor-A mediates ultraviolet B-induced impairment of lymphatic vessel function. Am J Pathol. 2006;169(4):1496–503. https://doiorg.publicaciones.saludcastillayleon.es/10.2353/ajpath.2006.060197.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Rutledge JC, Ng KF, Aung HH, Wilson DW. Role of triglyceride-rich lipoproteins in diabetic nephropathy. Nat Rev Nephrol. 2010;6(6):361–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrneph.2010.59.

    Article  CAS  PubMed  Google Scholar 

  99. Kim Y, Hwang SD, Lim JH, Kim MY, Kim EN, Choi BS, et al. Attenuated lymphatic proliferation ameliorates diabetic nephropathy and high-fat diet-induced renal lipotoxicity. Sci Rep. 2019;9(1):1994. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-018-38250-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Suzuki Y, Ito Y, Mizuno M, Kinashi H, Sawai A, Noda Y, et al. Transforming growth factor-β induces vascular endothelial growth factor-C expression leading to lymphangiogenesis in rat unilateral ureteral obstruction. Kidney Int. 2012;81(9):865–79. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ki.2011.464.

    Article  CAS  PubMed  Google Scholar 

  101. Hwang SD, Song JH, Kim Y, Lim JH, Kim MY, Kim EN, et al. Inhibition of lymphatic proliferation by the selective VEGFR-3 inhibitor SAR131675 ameliorates diabetic nephropathy in db/db mice. Cell Death Dis. 2019;10(3):219. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-019-1436-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Zheng Z, Zheng F. Immune cells and inflammation in diabetic nephropathy. J Diabetes Res. 2016;2016:1841690. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2016/1841690.

    Article  CAS  PubMed  Google Scholar 

  103. Avihingsanon Y, Benjachat T, Tassanarong A, Sodsai P, Kittikovit V, Hirankarn N. Decreased renal expression of vascular endothelial growth factor in lupus nephritis is associated with worse prognosis. Kidney Int. 2009;75(12):1340–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ki.2009.75.

    Article  CAS  PubMed  Google Scholar 

  104. Sato W, Kosugi T, Zhang L, Roncal CA, Heinig M, Campbell-Thompson M, et al. The pivotal role of VEGF on glomerular macrophage infiltration in advanced diabetic nephropathy. Lab Invest. 2008;88(9):949–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/labinvest.2008.60.

    Article  CAS  PubMed  Google Scholar 

  105. Keir LS, Firth R, Aponik L, Feitelberg D, Sakimoto S, Aguilar E, et al. VEGF regulates local inhibitory complement proteins in the eye and kidney. J Clin Invest. 2017;127(1):199–214. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci86418.

    Article  PubMed  Google Scholar 

  106. Rodas L, Barnadas E, Pereira A, Castrejon N, Saurina A, Calls J, et al. The density of renal lymphatics correlates with clinical outcomes in IgA nephropathy. Kidney Int Rep. 2022;7(4):823–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ekir.2021.12.029.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Hu D, Wang Z, Wang S, Li Y, Pei G, Zeng R, et al. Lymphatic vessels in patients with crescentic glomerulonephritis: association with renal pathology and prognosis. J Nephrol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40620-024-01903-0.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Lee HW, Qin YX, Kim YM, Park EY, Hwang JS, Huo GH, et al. Expression of lymphatic endothelium-specific hyaluronan receptor LYVE-1 in the developing mouse kidney. Cell Tissue Res. 2011;343(2):429–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00441-010-1098-x.

    Article  CAS  PubMed  Google Scholar 

  109. Kolovou K, Laskari K, Roumelioti M, Tektonidou MG, Panayiotidis P, Boletis JN, et al. B-cell oligoclonal expansions in renal tissue of patients with immune-mediated glomerular disease. Clin Immunol. 2020;217:108488. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.clim.2020.108488.

    Article  CAS  PubMed  Google Scholar 

  110. Cai Y, Chen MX, Deng YJ, Liu LL, Lin XP, Lu PF, et al. Clinical and pathological implications of increases in tonsillar CD19(+)CD5(+) B cells, CD208(+) dendritic cells, and IgA1-positive cells of immunoglobulin a nephropathy. Curr Med Sci. 2022;42(1):93–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11596-022-2532-5.

    Article  CAS  PubMed  Google Scholar 

  111. Qing J, Li C, Hu X, Song W, Tirichen H, Yaigoub H, et al. Differentiation of T helper 17 cells may mediate the abnormal humoral immunity in IgA nephropathy and inflammatory bowel disease based on shared genetic effects. Front Immunol. 2022;13:916934. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.916934.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Du W, Gao CY, You X, Li L, Zhao ZB, Fang M, et al. Increased proportion of follicular helper T cells is associated with B cell activation and disease severity in IgA nephropathy. Front Immunol. 2022;13:901465. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.901465.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Suzuki H, Suzuki Y, Aizawa M, Yamanaka T, Kihara M, Pang H, et al. Th1 polarization in murine IgA nephropathy directed by bone marrow-derived cells. Kidney Int. 2007;72(3):319–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.ki.5002300.

    Article  CAS  PubMed  Google Scholar 

  114. Ebihara I, Hirayama K, Yamamoto S, Muro K, Yamagata K, Koyama A. Th2 predominance at the single-cell level in patients with IgA nephropathy. Nephrol Dial Transplant. 2001;16(9):1783–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/ndt/16.9.1783.

    Article  CAS  PubMed  Google Scholar 

  115. Lu G, Zhang X, Shen L, Qiao Q, Li Y, Sun J, et al. CCL20 secreted from IgA1-stimulated human mesangial cells recruits inflammatory Th17 cells in IgA nephropathy. PLoS ONE. 2017;12(5):e0178352. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0178352.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Wei H, Chen L, Li Q, Liang X, Wang K, Zhang Y, et al. CD137L-macrophage induce lymphatic endothelial cells autophagy to promote lymphangiogenesis in renal fibrosis. Int J Biol Sci. 2022;18(3):1171–87. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/ijbs.66781.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Watanabe H, Mamelak AJ, Weiss E, Wang B, Freed I, Brice AK, et al. Anti-vascular endothelial growth factor receptor-2 antibody accelerates renal disease in the NZB/W F1 murine systemic lupus erythematosus model. Clin Cancer Res. 2005;11(1):407–9.

    Article  CAS  PubMed  Google Scholar 

  118. Tang Y, Zhang Y, Li X, Xu R, Ji Y, Liu J, et al. Immune landscape and the key role of APOE+ monocytes of lupus nephritis under the single-cell and spatial transcriptional vista. Clin Transl Med. 2023;13(4):e1237. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ctm2.1237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Machnik A, Neuhofer W, Jantsch J, Dahlmann A, Tammela T, Machura K, et al. Macrophages regulate salt-dependent volume and blood pressure by a vascular endothelial growth factor-C-dependent buffering mechanism. Nat Med. 2009;15(5):545–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nm.1960.

    Article  CAS  PubMed  Google Scholar 

  120. Lopez Gelston CA, 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. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/circresaha.118.312765.

    Article  CAS  PubMed  Google Scholar 

  121. De Miguel C, Lund H, Mattson DL. High dietary protein exacerbates hypertension and renal damage in Dahl SS rats by increasing infiltrating immune cells in the kidney. Hypertension. 2011;57(2):269–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/hypertensionaha.110.154302.

    Article  PubMed  Google Scholar 

  122. Rodriguez-Iturbe B. Renal infiltration of immunocompetent cells: cause and effect of sodium-sensitive hypertension. Clin Exp Nephrol. 2010;14(2):105–11. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10157-010-0268-1.

    Article  PubMed  Google Scholar 

  123. Johnson RJ, Herrera-Acosta J, Schreiner GF, Rodriguez-Iturbe B. Subtle acquired renal injury as a mechanism of salt-sensitive hypertension. N Engl J Med. 2002;346(12):913–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMra011078.

    Article  CAS  PubMed  Google Scholar 

  124. De Miguel C, Das S, Lund H, Mattson DL. T lymphocytes mediate hypertension and kidney damage in Dahl salt-sensitive rats. Am J Physiol Regul Integr Comp Physiol. 2010;298(4):R1136-42. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/ajpregu.00298.2009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Rodríguez-Iturbe B, Franco M, Tapia E, Quiroz Y, Johnson RJ. Renal inflammation, autoimmunity and salt-sensitive hypertension. Clin Exp Pharmacol Physiol. 2012;39(1):96–103. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1440-1681.2011.05482.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Franco M, Tapia E, Bautista R, Pacheco U, Santamaria J, Quiroz Y, et al. Impaired pressure natriuresis resulting in salt-sensitive hypertension is caused by tubulointerstitial immune cell infiltration in the kidney. Am J Physiol Renal Physiol. 2013;304(7):F982-90. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/ajprenal.00463.2012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Justin Rucker A, Crowley SD. The role of macrophages in hypertension and its complications. Pflugers Arch. 2017;469(3–4):419–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00424-017-1950-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Balasubbramanian D, Baranwal G, Clark MC, 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. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/hjh.0000000000002349.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. 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 Renal Physiol. 2017;312(5):F861-f9. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/ajprenal.00679.2016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. 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. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/ajh/hpz139.

    Article  CAS  PubMed  Google Scholar 

  131. Wilcox BK, Henley MR, Navaneethabalakrishnan S, Martinez KA, Pournouri A, Goodlett BL, et al. Hypertensive stimuli indirectly stimulate lymphangiogenesis through immune cell secreted factors. Cells. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells11142139.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Goodlett BL, Balasubbramanian D, Navaneethabalakrishnan S, Love SE, Luera EM, Konatham S, et al. Genetically inducing renal lymphangiogenesis attenuates hypertension in mice. Clin Sci. 2022;136(23):1759–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1042/cs20220547.

    Article  CAS  Google Scholar 

  133. 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. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/pharmaceutics14010084.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Basile DP, Abais-Battad JM, Mattson DL. Contribution of Th17 cells to tissue injury in hypertension. Curr Opin Nephrol Hypertens. 2021;30(2):151–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/mnh.0000000000000680.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Donnan MD. Kidney lymphatics: new insights in development and disease. Curr Opin Nephrol Hypertens. 2021;30(4):450–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/mnh.0000000000000717.

    Article  CAS  PubMed  Google Scholar 

  136. Mikolajczyk TP, Guzik TJ. Adaptive immunity in hypertension. Curr Hypertens Rep. 2019;21(9):68. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11906-019-0971-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Barbaro NR, Foss JD, Kryshtal DO, Tsyba N, Kumaresan S, Xiao L, et al. Dendritic cell amiloride-sensitive channels mediate sodium-induced inflammation and hypertension. Cell Rep. 2017;21(4):1009–20. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2017.10.002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Weimbs T. Are cyst-associated macrophages in polycystic kidney disease the equivalent to TAMs in cancer? J Am Soc Nephrol: JASN. 2018;29(10):2447–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1681/asn.2018080846.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Swenson-Fields KI, Vivian CJ, Salah SM, Peda JD, Davis BM, van Rooijen N, et al. Macrophages promote polycystic kidney disease progression. Kidney Int. 2013;83(5):855–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ki.2012.446.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. 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. https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.48183.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Kleczko EK, Marsh KH, Tyler LC, Furgeson SB, Bullock BL, Altmann CJ, et al. CD8(+) T cells modulate autosomal dominant polycystic kidney disease progression. Kidney Int. 2018;94(6):1127–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.kint.2018.06.025.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Aschen SZ, Farias-Eisner G, Cuzzone DA, Albano NJ, Ghanta S, Weitman ES, et al. Lymph node transplantation results in spontaneous lymphatic reconnection and restoration of lymphatic flow. Plast Reconstr Surg. 2014;133(2):301–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/01.prs.0000436840.69752.7e.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Jackson DG. Lymphatic regulation of cellular trafficking. J Clin Cell Immunol. 2014. https://doiorg.publicaciones.saludcastillayleon.es/10.4172/2155-9899.1000258.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Kerjaschki D, Regele HM, Moosberger I, Nagy-Bojarski K, Watschinger B, Soleiman A, et al. Lymphatic neoangiogenesis in human kidney transplants is associated with immunologically active lymphocytic infiltrates. J Am Soc Nephrol. 2004;15(3):603–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/01.asn.0000113316.52371.2e.

    Article  CAS  PubMed  Google Scholar 

  145. Pedersen MS, Müller M, Rülicke T, Leitner N, Kain R, Regele H, et al. Lymphangiogenesis in a mouse model of renal transplant rejection extends life span of the recipients. Kidney Int. 2020;97(1):89–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.kint.2019.07.027.

    Article  CAS  PubMed  Google Scholar 

  146. Mehlhorn U, Davis KL, Burke EJ, Adams D, Laine GA, Allen SJ. Impact of cardiopulmonary bypass and cardioplegic arrest on myocardial lymphatic function. Am J Physiol. 1995;268(1 Pt 2):H178–83. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/ajpheart.1995.268.1.H178.

    Article  CAS  PubMed  Google Scholar 

  147. Kong XQ, Wang LX, Kong DG. Cardiac lymphatic interruption is a major cause for allograft failure after cardiac transplantation. Lymphat Res Biol. 2007;5(1):45–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/lrb.2007.5108.

    Article  PubMed  Google Scholar 

  148. Rouhani SJ, Eccles JD, Tewalt EF, Engelhard VH. Regulation of T-cell tolerance by lymphatic endothelial cells. J Clin Cell Immunol. 2014. https://doiorg.publicaciones.saludcastillayleon.es/10.4172/2155-9899.1000242.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Zhang N, Schröppel B, Lal G, Jakubzick C, Mao X, Chen D, et al. Regulatory T cells sequentially migrate from inflamed tissues to draining lymph nodes to suppress the alloimmune response. Immunity. 2009;30(3):458–69. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.immuni.2008.12.022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Stuht S, Gwinner W, Franz I, Schwarz A, Jonigk D, Kreipe H, et al. Lymphatic neoangiogenesis in human renal allografts: results from sequential protocol biopsies. Am J Transplant. 2007;7(2):377–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1600-6143.2006.01638.x.

    Article  CAS  PubMed  Google Scholar 

  151. Phillips S, Kapp M, Crowe D, Garces J, Fogo AB, Giannico GA. Endothelial activation, lymphangiogenesis, and humoral rejection of kidney transplants. Hum Pathol. 2016;51:86–95. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.humpath.2015.12.020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Palin NK, Savikko J, Koskinen PK. Sirolimus inhibits lymphangiogenesis in rat renal allografts, a novel mechanism to prevent chronic kidney allograft injury. Transpl Int. 2013;26(2):195–205. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/tri.12005.

    Article  CAS  PubMed  Google Scholar 

  153. Talsma DT, Katta K, Boersema M, Adepu S, Naggi A, Torri G, et al. Increased migration of antigen presenting cells to newly-formed lymphatic vessels in transplanted kidneys by glycol-split heparin. PLoS ONE. 2017;12(6):e0180206. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0180206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Espinosa JR, Samy KP, Kirk AD. Memory T cells in organ transplantation: progress and challenges. Nat Rev Nephrol. 2016;12(6):339–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrneph.2016.9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Chow F, Ozols E, Nikolic-Paterson DJ, Atkins RC, Tesch GH. Macrophages in mouse type 2 diabetic nephropathy: Correlation with diabetic state and progressive renal injury. Kidney Int. 2004;65(1):116–28. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1523-1755.2004.00367.x.

    Article  CAS  PubMed  Google Scholar 

  156. Chow FY, Nikolic-Paterson NJ, Atkins RC, Tesch GH. Macrophages in streptozotocin-induced diabetic nephropathy: potential role in renal fibrosis. Nephrol Dial Transplant. 2004;19(12):2987–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/ndt/gfh441.

    Article  CAS  PubMed  Google Scholar 

  157. Cucak H, Nielsen Fink H, Højgaard Pedersen M, Rosendahl A. Enalapril treatment increases T cell number and promotes polarization towards M1-like macrophages locally in diabetic nephropathy. Int Immunopharmacol. 2015;25(1):30–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.intimp.2015.01.003.

    Article  CAS  PubMed  Google Scholar 

  158. Zhu J. T helper 2 (Th2) cell differentiation type 2 innate lymphoid cell (ILC2) development and regulation of interleukin-4 (IL-4) and IL-13 production. Cytokine. 2015;75(1):14–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cyto.2015.05.010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Huang H, Peng Y, Long X-D, Liu Z, Wen X, Jia M, Liang Y, Huang A. Tonsillar CD4+CD25+ regulatory T cells from IgA nephropathy patients have decreased immunosuppressive activity in experimental IgA nephropathy rats. Am J Nephrol. 2013;37(5):472–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1159/000350533.

    Article  CAS  PubMed  Google Scholar 

  160. Huang D-L, He Y-R, Liu Y-J, He H-Y, Gu Z-Y, Liu Y-M, Liu W-J, Luo Z, Ju M-J. The immunomodulation role of Th17 and Treg in renal transplantation. Front Immunol. 2023;14:1113560. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1113560.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Kolovou K, Laskari K, Roumelioti M, Tektonidou MG, Panayiotidis P, Boletis JN, Marinaki S, Sfikakis PP. B-cell oligoclonal expansions in renal tissue of patients with immune-mediated glomerular disease. Clin Immunol. 2020;217:108488. 10.1016/j.clim.2020.108488

  162. Barnett N, Dorling A, Mamode N. B cells in renal transplantation: pathological aspects and therapeutic interventions. Nephrol Dial Transplant. 2011;26(3):767–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/ndt/gfq716.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The work was supported by discipline construction fund of Sichuan Provincial People's Hospital; Supported by Sichuan Science and Technology Program 2022YFS0331; Sichuan Provincial Health Commission 21PJ083. The National Natural Science Foundation of China (82200810); and the Natural Science Foundation of Sichuan (2023NSFSC1526).The project of 2020 High-level Overseas Chinese Talent Returning Funding; Foundation of Applied Basic Research Project of Sichuan Provincial Science and Technology (2020YJ0179); Foundation for Young Talent Fund of Sichuan Provincial People’s Hospital (2022QN02); Discipline construction fund of Sichuan Provincial People’s Hospital.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: Xiangheng Lu and Chi Liu; investigation original draft preparation: Kuai Ma, Junyi Ren, Haoyu Peng and Jia Wang; review and editing: Moussa Ide Nasser and Xiaoxiao Wang; funding acquisition: Chi Liu.

Corresponding authors

Correspondence to Xiaoxiao Wang, Moussa Ide Nasser or Chi Liu.

Ethics declarations

Ethics approval and consent to participate

There is no ethical issue for all authors.

Consent for publication

All authors consent for publication.

Competing interests

No competition of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lu, X., Ma, K., Ren, J. et al. The immune regulatory role of lymphangiogenesis in kidney disease. J Transl Med 22, 1053 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05859-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05859-4

Keywords