Abstract
The rate of arteriovenous fistula (AVF) maturation failure remains unacceptably high despite continuous efforts on technique improvement and careful pre-surgery planning. In fact, half of all newly created AVFs are unable to be used for hemodialysis (HD) without a salvage procedure. While vascular stenosis in the venous limb of the access is the culprit, the underlying factors leading to vascular narrowing and AVF maturation failure are yet to be determined. We have recently demonstrated that AVF non-maturation is associated with post-operative medial fibrosis and fibrotic stenosis, and post-operative intimal hyperplasia (IH) exacerbates the situation. Multiple pathological processes and signaling pathways are underlying the stenotic remodeling of the AVF. Our group has recently indicated that a pro-inflammatory cytokine platelet factor 4 (PF4/CXCL4) is upregulated in veins that fail to mature after AVF creation. Platelet factor 4 is a fibrosis marker and can be detected in vascular stenosis tissue, suggesting that it may contribute to AVF maturation failure through stimulation of fibrosis and development of fibrotic stenosis. Here, we present an overview of the how PF4-mediated fibrosis determines AVF maturation failure.
Keywords: AV fistula, dialysis access, platelet factor 4, arteriovenous fistula maturation, arteriovenous fistula failure, intimal hyperplasia
Introduction
Since Brescia and his colleagues1 created the first arteriovenous fistula (AVF) in New York in 1965, this type of vascular access has been used to deliver hemodialysis (HD) in patients suffering from various types of kidney failure, including the most severe one: end stage renal disease (ESRD).2 In fact, AVF is considered the best amongst all three vascular access methods to deliver HD.3–5 Compared to the other two methods, which are arteriovenous grafts (AVGs) and central vein catheters (CVCs), AVFs have lower complication rates, thrombosis and infection rates, fewer access related hospitalization, better long-term patency, and lower health care related cost burden.6–11 Patients with AVFs also have shown substantially lower mortality rates and increased life expectancy.6 Therefore, the AVF has become the preferred option in ESRD patients12,13 and recommended by the Kidney Disease Outcome Quality Initiative (KDOQI) vascular access guidelines.14 However, the frequency of AVF primary failure has remained high for the last five decades.15,16 Studies have shown that up to 23%–50% of newly created AVFs fail to mature and cannot be used for HD without undergoing a salvage procedure,8–21 and a 60% failure to mature rate amongst total AVFs has been reported in the US.22–24 This causes a significant negative impact on morbidity, mortality, and life quality of patients who require HD for survival, as well as increased medical costs.5
The role of outward remodeling in AVF non-maturation has been recently demonstrated,25 Fibrosis has been previously detected in stenotic AVFs,26 and our recent study demonstrated a close association between AVF maturation failure and post-operative fibrotic stenosis.27 At the molecular level, the pathology behind AVF maturation failure has not been fully illustrated, but several proteins are thought to be critical during this process, such as heme oxygenase−1 and −2 (HO-1 and HO-2), monocyte chemoattractant protein-1 (MCP-1), Kruppel-like factor 2 (KLF2), transforming growth factor β1 (TGFβ1), and inflammatory markers.28–30 The last one, such as matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs) have also been considered as potential predictors of AVF maturation.28–30 Platelet factor 4 (PF4/CXCL4) is a pro-inflammatory cytokine that has previously been associated with fibrosis,31–33 and we recently found it is upregulated in veins that fail to mature after AVF creation.34 These discoveries suggested that PF4 plays in AVF maturation failure. Therefore, in this review, we summarize the recent findings that link fibrosis with AVF maturation failure, as well as the involvement of PF4 in fibrosis and explore the potential role of PF4 in AVF maturation failure.
Fibrosis and AVF maturation failure
Fibrosis
Fibrosis is an outcome of the dysregulated tissue repair process in response to various types of tissue injury.35 It is characterized by excessive accumulation/deposition of extracellular matrix (ECM) components, such as collagens and fibronectins.35–38 During tissue injuries endothelial cells are damaged, and the basement membrane is thus exposed, which causes a clotting process and platelets are recruited. This results in releasing of factors such as platelet-derived growth factors (PDGFs) and transforming growth factor β1 (TGFβ1) that lead to smooth muscle cell (SMC) and fibroblasts activation. Activated fibroblasts have increased contractility and able to secrete inflammatory mediators and synthesize ECM components.35,39 Fibrosis can affect normal tissue and organ functions in a pathological state,40,41 and may lead to kidney, cardiac, liver failure, vessel dysfunction, or even life threatening conditions, such as idiopathic pulmonary fibrosis in extreme cases.42–45 In the vasculature, fibrosis is featured by reduced liminal diameter and arterial wall thickening caused by excessive deposition of ECM.46,47 Vascular fibrosis involves proliferation of vascular smooth muscle cells (VSMCs), accumulation of ECM particularly collagen and fibronectin in the vascular wall, and inhibition of matrix degradation.46,48,49
Fibrosis can result from many causes, inflammation is the most notably one in general.35,50 Fibrosis can be triggered by both acute and chronic inflammation,37 with inflammatory monocytes and tissue-resident macrophages acting as key regulators of tissue fibrosis.51–53 Inflammation leads to injury of epithelial and often endothelial cells, which in turn enhances the release of inflammatory mediators and the recruitment of inflammatory cells, which activates effector cells and drive the fibrogenic process.54 Fibroblasts, SMC, and myofibroblasts are all effector cells and key fibrosis mediators in many organs. They are responsible for the synthesis of ECM proteins,37,55,56 mostly collagen I, III and fibronectin,37,57 in all types of fibrosis.58–60 Fibroblasts can also produce collagen IV in some instances.61,62 Interestingly, SMC proliferation can be stimulated by monomeric/soluble collagen I, while soluble collagen IV stimulates SMC migration in vitro.63 This suggests that there may be a feed forward signaling loop between SMC and collagens that contributes to fibrosis progression. Profibrotic macrophages interact with fibroblasts,64 which also contributes to ECM deposition during fibrosis.65–70 Fibroblasts can undergo phenotype switching into myofibroblasts during fibrosis,71–74 and myofibroblasts that are contractile express α-smooth muscle actin (αSMA) as a biomarker.75 Apart from fibroblasts and myofibroblasts, collagens can also be produced by hematopoietic, epithelial, and endothelial cells.76
Hypertension is another important cause of fibrosis, particularly in cardiovascular fibrosis. Prohypertensive factors, including activation of the renin-angiotensinaldosterone system (RAAS), inflammation, oxidative stress, salt intake, etc. have all been shown to result in excessive arterial fibrosis and ECM deposition.77 Vascular changes associated with hypertension are featured by decrease of nitric oxide (NO) level and increase of reactive oxygen species (ROS) production, proinflammatory and profibrotic pathway activation, suppression of ECM degradation including decrease of collagen turnover, and ECM remodeling especially the increase of fibrillar type I collagen.77–79 All these effects combine to increase the possibility of fibrosis occurrence. In fact, ECM accumulation that resulted from increase of collagen synthesis and reduction of collagen degradation is the major reason of fibrosis development in response to chronic hypertension,80 especially when considering that collagens I and III are the predominant components in vascular ECM.81 Apart from collagen, elastin also plays a role in vascular fibrosis. Deficiency of elastin may exacerbate vascular fibrosis and increase the stiffness of vessels that is caused by collagen excess.81–83 Furthermore, imbalanced elastin/collagen ratio has been shown to lead to the deposition of excessive ECM proteins, particularly collagen and fibronectin.81
Other cardiovascular disease-associated risk factors, such as hyperglycemia, dyslipidemia (hypercholesterolemia), and hyperhomocysteinemia (HHcy) are also associated with vascular fibrosis initiation and progression.46 Increase of collagens, fibronectin and altered matrix degradation have been shown involved in interstitial fibrogenesis in hypercholesterolemic rats.84,85 Circulating oxidized low-density lipoproteins (ox-LDL) can also promote fibrosis through stimulating the synthesis and expression of TGFβ in hypercholesterolemia patients.86–88 Several of the above features of vascular fibrosis, such as excessive ECM deposition and imbalanced elastin/collagen proportions have all been shown in HHcy.89 The increase of ECM deposition in HHcy may be due to alteration of ECM metabolism that are caused by activation of MMPs,90 while the elastin/collagen imbalance may be due to increase intracellular Ca2+ signaling.91
Several physiological and pathological medical conditions, such as aging, diabetes, and obesity are known risk factors that would contribute to vascular fibrosis. Aging is one of the main causes of hypertension,77 while diabetes would cause expansion of ECM with the occurrence of fibrosis in many tissues including the vasculature.92 High glucose has been shown to increase angiotensin II (Ang II) (will be discussed below) generation which will result in promoting fibrosis via stimulation of TGFβ secretion.93 Vascular remodeling and stiffness, on the other hand, are shown to be associated with obesity94 and insulin resistance.95
Besides the vasculature, fibrosis also occurs in various anatomic locations due to different diseases and/or pathologies. Progressive forms of chronic kidney disease (CKD) are associated with excessive deposition of ECM, leading to ESRD,96–98 and hypertension and diabetes are the two leading causes of renal fibrosis.37 A lot of diseases can lead to pulmonary fibrosis, among which idiopathic pulmonary fibrosis is a progressive one without substantial inflammation.99 Finally, some genetic mutations have been found to be associated with fibrosis. For example, mutations in telomerase reverse transcriptase (TERT) and mucin 5B (MUC5B) are associated with pulmonary fibrosis,100–103 while mutations in the myh7 gene is associated with cardiac fibrosis,104 and mutation in dystrophin (DMD) is associated with Duchenne muscular dystrophy-associated skeletal muscle fibrosis.105
Molecules and molecular pathways associated with fibrosis
At the molecular level, fibrosis is the result of a complex crosstalk between various signaling pathways that are involved in injury response, such as TGFβ, Wingless and Int-1 (Wnt), Notch, Yes-associated protein/transcription co-activator with PDZ-binding motif (YAP/TAZ), AMP-activated protein kinase (AMPK), Ras, etc.,106–109 and TGFβ pathway is considered to be the primary factor driving fibrosis.108 TGFβ is expressed in many cell types, including endothelial cells, VSMCs, myofibroblasts, and adventitial macrophages,77,110 while fibroblasts are the major targets of this factor,111 although TGFβ also stimulates a fibrogenic phenotype in epithelial cells.112 Intriguingly, some other cell types, such as endothelial cells and mural cells may convert to fibroblasts in response to TGFβ.111 TGFβ reduces collagenase production while increases TIMPs and results in excessive collagen/ECM accumulation.46,113,114
In the canonical TGFβ pathway, binding of ligand to TGFβ receptor II (TGFβRII) recruits and phosphorylates TGFβ receptor Is (TGFβRIs). These phosphorylated/activated TGFβRIs will then phosphorylate the effector protein Smads, which then migrate to the nucleus to function as transcription co-activators of many fibrosis associated genes.37,115 The signaling of TGFβ can be enhanced by binding of TGFβ receptor III (TGFβRIII) or endoglin.116 TGFβ is expressed and stored in the ECM as a large latent complex (LLC), a molecule structure that comprises of an active TGFβ, a latency-associated protein (LAP), and a latent TGFβ binding protein (LTBP).117 Active TGFβ can be obtained through many mechanisms, including MMPs and other proteases,115,118–125 integrins such as αvβ6,126–132 matrix proteins including the ED-A fibronectin isoform, fibulin-1c, and thrombospondin-1,133–136 and physical and chemical conditions such as low pH, heat, radiation, shear stress, and ROS.137 It is worth mentioning that besides activating TGFβ, some MMPs themselves are involved in fibrosis. For example, it has been shown that MMPs are necessary for SMC and fibroblasts migration that leads to fibrosis.56,138 MMPs also degrades ECM proteins that result in ECM modification which leads to cardiovascular remodeling and fibrosis,77,139,140 and MMP9 activation potentiates fibrosis.141
Expression of TGFβ has been found to correlate with progression of liver, lung, kidney, skin, and cardiac fibrosis, while reduction of fibrosis development upon inhibition of TGFβ pathway has been reported in experimental models.89,142 TGFβ affects inflammation,142 enhances infiltration and/or proliferation of fibroblasts, induces proliferation and activation of intrinsic fibroblasts into collagen producing myofibroblasts, increases ECM synthesis, and inhibits degradation of collagen.143–145 Studies have shown that activation of vascular TGFβ1 increases the synthesis of ECM proteins, such as fibronectin, collagens, and plasminogen activator inhibitor-1 (PAI-1),146–151 and active TGFβ1 also promotes fibroblast differentiation into myofibroblasts,152 featured by initiation of αSMA expression in fibroblasts.147,153 Upregulation of PAI-1, a downstream effector of TGFβ, would result in ECM accumulation via prevention of collagen degradation.77 Concurrently, connective tissue growth factor (CTGF), another downstream effector of TGFβ, is implicated in fibroblast proliferation and a key mediator of ECM production during fibrosis.133,154
As mentioned above, the TGFβ pathway induces fibrosis as a participant of a larger complex crosstalk, and TGFβR can mediate signals via mitogen-activated protein kinase-extracellular signal-regulated kinase (MAPKERK) and ERK pathway activation, which facilitates fibroblast survival and promotes fibrosis.155 Meanwhile, TGFβR also signals through c-Jun N-terminal kinase (JNK), p38-MAPK, nuclear factor κ B (NF-κB), phosphoinositide 3-kinase-protein kinase B-mammalian target of rapamycin (PI3K-Akt-mTOR), Rho-associated coiled-coil forming protein kinase (ROCK),108,156–163 and cooperates with Wnt & Notch signaling cascade.164 In addition, TGFβ1 is able to induce several miRNA species that have fibrotic effects.108 In fact, a series of miRNAs, such as miR-192, miR-19b, miR-21, miR-29, miR-30, and miR-133 are all involved in fibrosis.165–168
Interestingly, besides being the end product of fibrosis, collagens are associated with fibrosis onset. ECM fragments are important drivers of fibrosis by stimulating chemokine and proinflammatory cytokine production in inflammatory monocytes and macrophages, including the aforementioned activation of TGFβ.134,136,169 The collagen fragment ac-PGP produced by MMPs indirectly induces inflammation by attracting leukocytes through chemotaxis.170,171 This molecule also drives inflammation and ECM remodeling by enhancing MMP9 as well as neutrophil elastase secretion and activation in a pulmonary inflammation model.172
Vascular fibrosis, different from fibrosis in other tissues, can be further promoted by vasoactive agents, such as Ang II, aldosterone, and endothelin-1 (ET-1). Ang II, other than being a blood pressure regulator, is also responsible for vascular cell growth/apoptosis, VSMC migration, inflammation responses, and ECM remodeling.173 It promotes inflammation46 and ECM synthesis which leads to vascular fibrosis through TGFβ, CTGF, interleukin-6 (IL6), tumor necrosis factor α (TNFα), and MCP-1.174,175 Overexpression of miR-122 exacerbates Ang II-mediated ECM deposition.176,177 TGFβ1 activation during aging and hypertension is mediated by Ang II178,179 and ET-1,180 as well as mechanical stress149,181 and ROS.182 Ang II acts through its receptor angiotensin II receptor type 1 (AT-1R) and mineralocorticoid receptor (MR) to increase TGFβ signaling and promote ECM synthesis.140,183–186 Antagonizing or knocking out of AT-1R decrease fibrosis, reduce intima-medial thickness, and attenuate intima hyperplasia.187–190 Similarly, conversion of Ang II into Ang-(1–7) by the angiotensin converting enzyme (ACE) homolog ACE2 has been shown to prevent cellular proliferation and vascular fibrosis.191 Loss of ACE2 will lead to increase of Ang II-mediated expression of TGFβ, CTGF, and collagens,192,193 while overexpression of ACE2 prevents the Ang II-induced increase of proinflammatory reaction.194–196 Inhibition of ACE with H2S can control the vascular fibrosis progression characteristic of chronic hypertension.197–199 In contrast, decrease of H2S in HHcy ameliorates its ability to inhibit ACE, which results in increase of Ang II-dependent profibrotic stimulation.200 Ang II can also activate Smad in an TGFβ-independent manner,180,201 and it can induce activation of p38 MAPK which is associated with the development and progression of fibrosis.202–204 Other than that, Ang II has been reported to activate CTGF73 and MMP2 and 9.77
Aldosterone has been shown to promote collagen I synthesis in rat.205,206 It also stimulates PAI-1 expression and induces CTGF expression via p38 MAPK cascade and MR.207,208 Inhibiting MR with galectin-3 attenuates hypertension, inflammation, and vascular fibrosis in an aldosterone-salt challenge rat model.209 ET-1, in turn, can stimulate fibroblast-induced collagen synthesis.210,211
In patients suffering from chronic hypertension, the production of advanced glycation end products (AGEs) has been accelerated.92,212 This molecule stimulates the release of TGFβ.213–215 It is also notoriously known to halt matrix degradation by both irreversibly crosslinking with ECM macromolecules including collagen backbones216 and reducing the activity of MMPs.217,218
Apart from AGEs, tissue transglutaminase (TG2) is also a crosslinker, which promotes vascular stiffness by increasing cell surface-ECM crosslink.219 ECM proteins such as fibronectin, collagen, and laminin have all been reported as substrates of TG2.77 Under fibrotic condition, TG2/ECM protein crosslinks are increased,77 while inhibition of TG2 has been shown to attenuate fibrosis in a mouse model.220 Interestingly, TG2 itself also activates TGFβ/Smad signaling and promotes collagen synthesis and fibrosis.221
In lymphatic collecting vessels, the C-C chemokine receptor 7 (CCR7) has been implicated in preventing fibrosis. Increased collagen deposition has been shown in the adventitia of lymphatic collecting vessels of CCR7−/− mice and fibroblasts can be observed within the excess adventitial collagen. This fibrosis is reversed by re-expression of CCR7 in dendritic cells (DCs).222
There are also many other signal pathways and molecules important to fibrosis. The Wnt pathway, besides cross-talking with TGFβ, is also important to fibrosis by itself. Activation of canonical Wnt pathway has been reported in kidney, liver, skin, and cardiac fibrosis,223,224 while bleomycin-induced pulmonary fibrosis has been shown to be highly dependent on the Wnt-1 pathway.225 Wang et al.226 have shown that Wnt-1-inducible signaling protein 1 (WISP-1) can mediate TGFβ1-driven renal fibrosis in a mouse model, and induction of TGFβ and collagen expression in fibroblasts by Wnt3a has been shown by Poon et al.227 Interleukins (ILs), such as IL-17A, type 2-associated IL-4, and IL-13 have been shown to induce fibrosis.35,228–233 The transcription factor SPI1 (PU.1) polarizes resting fibroblasts and repolarizes ECM degrading inflammatory fibroblasts to an ECM-producing fibrotic phenotype,35,234 and inactivation of SPI1 leads to regression of fibrosis.234 The glycoprotein OX40-OX40 ligand (OX40/OX40L) axis promotes collagen synthesis/fibrosis and regulates cytokine balance toward a proinflammatory and profibrotic profile.235 Blockade of this protein prevents inflammation driven fibrosis in skin, lung, vessels, etc., and anti-OX40L antibody can reduce the severity of bleomycin-induced fibrosis.235 Finally, fibrosis can also be induced by PDGF, integrins, and the microbiome, among other factors.236–239
Fibrosis and AVF
Fibrosis had not been considered a major factor in AVF maturation failure until recently.27 It has been suggested that fibrosis has a causative association with irregular shapes, disrupted membranes, pseudopodia-like projections, and intracellular collagen-containing vacuoles of VSMCs populating the media of pre-access veins.240 This indicates that vein fibrosis may result from and/or contribute to the transformation of differentiated VSMCs into the myofibroblastic and migratory phenotype that contribute to intimal hyperplasia (IH) and AVF failure.241 This is supported by our recent findings using lineage tracing techniques that demonstrate differentiated venous VSMCs ultimately contribute to both medial wall thickening and IH in a mouse jugular branch end to carotid side anastomosis model.242 In another study, we have found that one downstream target of the TGFβ/Smad pathway, tetraspanin 2 (TSPAN2), is downregulated in both an in vitro VSMC de-differentiation model and in failed human AVF samples.243 This further links vascular fibrosis with AVF maturation failure. In a different mouse model, Mishra et al.244 have found that an aorta-to-vena cava AVF would result in fibrosis around the capillaries as well as interstitial regions at week 6 post-surgery, and treatment with H2S at this point will result in attenuation of pathological condition by week 8–10. Medial fibrosis is common in CKD patients,245 and increased vascular stiffness caused by medial fibrosis has been proposed as a limiting factor for arterial dilation and AVF maturation in patients with CKD.241
Recently, we performed a study using venous samples pairs collected from patients who underwent two-stage upper-arm AVF surgeries at a single center, including pre-access veins from the first-stage access creation and AVF cross-sections from the second-stage transposition. Analysis of these samples showed that post-operative medial fibrosis and circumferential alignment of fibers around the lumen are closely associated with AVF maturation failure.27 Meanwhile, we also noticed that non-matured AVFs presented higher level of collagen deposition in the media layer.27 Interestingly, although not associated with stenosis development and AVF maturation failure by itself,246 post-operative IH showed an association with AVF maturation failure in patients with high post-operative medial fibrosis.27 Consistently, we have also observed that AVFs with co-existing IH and medial fibrosis had a lower increase in venous diameter during remodeling.27 A subsequent study further demonstrated increased collagen crosslinking in AVFs that failed and higher vascular expression of the collagen crosslinking enzyme lysyl oxidase (LOX).247
Our discovery of the association between fibrosis and AVF maturation failure is consistent with recent findings by Cai et al. Using RNA-sequence profiling of murine VSMCs, this group has shown that lower AVF patency rate is associated with higher expression of fibrotic genes.248 At 28 days after AVF creation, the patency rate, venous diameter, average neointima/media + adventitia area ratio, and areas and average cell density in neointima are all lower in female mice compared to male. In contrast, increased TGFβ1, TGFβRI, and collagen I are shown in VSMCs of the AVF from female mice, together with higher levels of α-SMA and pSmad3 and reduced bone morphogenetic protein 7 (BMP7) and interleukin 17 receptor b (IL-17Rb) expression. Most pSmad3 have been found to locate in the media layer and neointima, which promotes constrictive remodeling and venous fibrosis in the AVF from female mice.248 Furthermore, MR, being one of the receptors of Ang II which is critical to vascular fibrosis and IH,187 has been found upregulated in dysfunctional AVF.187
The knowledge regarding molecular cues and mechanisms responsible for the development of fibrosis in AVF is still limited. However, several studies have indicated that TGFβ may play an important role in AVF maturation failure,249,250 while others have shown that TGFβ is heavily involved in vascular remodeling.251,252 By performing bulk RNA-seq analysis of human pre-access veins, our group recently identified nine transcripts (false discovery rate [FDR] ≤ 0.05) encoding for fibrosis related proteins upregulated in veins that failed versus veins that had successful AVF maturation.34 Among these nine genes, PF4/CXCL4 has been previously linked to the development of fibrosis.32,33
PF4, fibrosis, and AVF maturation failure
PF4 and fibrosis
PF4 is a very abundant chemokine that is released during platelet activation.253 It is mainly expressed by megakaryocytes, but can also be secreted from monocytes, macrophages, and DCs.254–257 PF4 is involved in thrombosis, hemostasis, and innate immunity, including effects on monocyte and neutrophil chemotaxis,258,259 as well as promoting host inflammatory response in many ways. PF4 activates granulocytes by causing release of lysosomal enzymes from neutrophils,260 stimulates CD4+CD25+ T cells proliferation,261 prevents the apoptosis of monocytes and promotes their differentiation into macrophages,262 and stimulates natural killer (NK) cells to produce IL8 and migrate in inflamed environments.263,264
The functions of PF4 are mediated by its interaction with glycosaminoglycans, growth factors, lipoprotein receptor-related proteins, chemokine and growth factor receptors, integrins, or with several of those ligands at the same time.31 PF4 binds to C-X-C chemokine receptor 3B (CXCR3B) in microvascular endothelial cells to reduce DNA synthesis and promote apoptosis.265 It also induces intracellular Ca2+ release and the migration of activated human T lymphocytes via CXCR3A.266 PF4 drives the chemotaxis of monocytic THP-1 cells by binding to CCR1,267 and the interaction between PF4 and LDL-R leads to retention of LDL on fibroblasts surface, which affects LDL metabolism.268 Signaling through LDL receptor-related protein 1 (LRP1), PF4 promotes VSMCs to transform into a synthetic inflammatory phenotype characterized by a decline in differentiation markers, increased cytokine production, and cell proliferation.269 LRP1 acts as a repressor of the canonical Wnt pathway,270 and PF4 signaling through this receptor would contribute to Wnt pathway activation and β-catenin overexpression. It is believed that β-catenin could enhance TGFβ/Smads activity and increase cell differentiation as well as inflammatory and pro-fibrotic gene transcription.108,227,271,272 By interacting with integrins,31,273,274 PF4 has been shown to interfere with the cell cycle in Chinese hamster ovary (CHO) cells31,273 and cell migration in leukocytes.274
Interestingly, PF4 has also been reported to bind to one type of TGFβRI, activin receptor-like kinase 5 (ALK5), on NRK 49F fibroblasts and thus blocks it from binding to TGFβ1.275 The blockade of TGFβ signaling through ALK5 may have important implications for vascular remodeling upon tissue injury such as anastomosis creation in AVFs, because TGFβ signaling could be diverted through the ALK1 receptor instead. TGFβ signaling through ALK5 has been implicated with normal outward remodeling in AVF under laminar unidirectional blood flow, while signaling through ALK1 has been related to inward remodeling and stenosis when oscillating/disturbed blood flow is presented.29 It is known that both pathways, ALK1-Smad1/5 and ALK5-Smad2/3, are activated and upregulated during vascular remodeling and fibrosis.276 Moreover, the interaction and balance between these two branches, and the presence of other factors and signals are all capable of modulating TGFβ pathway.277–282 The role of PF4 on the regulation of this balance between branches has yet to be studied and would be an interesting topic to be investigated further.
PF4 has been previously shown as a fibrosis marker. Higher mRNA levels of PF4 can be seen in mice with liver fibrosis, while PF4 knockout mice have significantly reduce liver damage.33 Activated platelets contribute to fibrosis through the release of PDGF, PF4, and TGFβ.283–285 Increase of PF4 has been reported upon treatment of bleomycin, a drug that causes fibrosis, in lung tissue.286 In recent years, more evidence in line with this initial study have been reported. PF4 induces a pro-inflammatory and pro-fibrotic phenotype in monocyte-derived DCs, increasing ECM production, and inducing myofibroblast differentiation.287 Differential expression of hematopoietic PF4 marks the progression of bone marrow fibrosis, as the absence of PF4 decreases the activation of fibrotic pathways in megakaryocytes and inflammation in fibrosisdriving cells in three primary myelofibrosis models.288 PF4 is the marker of systemic sclerosis (SSc), a disease characterized by skin and internal organ fibrosis.289,290 In patients who are suffering from this disease, higher levels of PF4 are produced in plasmacytoid dendritic cells (pDCs) in response to toll-like receptor 8 (TLR8),291–293 and PF4 complexes with DNA/RNA to induce interferon α (IFN-α) activity in pDCs and stimulates the production of anti-PF4 antibodies from B-cells.290 Both increased levels of PF4 in circulation and anti-PF4 activity correlate with lung fibrosis.290,293,294 At the same time, the presence of pDCs in the skin correlates with PF4 levels, supporting a mechanistic link for skin fibrosis in SSc patients.291
On the other hand, Zhang et al.295 have shown that PF4 treatment decreases interstitial inflammation and fibrosis after renal transplantation at day 56. To our knowledge, this is the only report that indicates an inhibitory effect of PF4 on fibrosis.
PF4 in AVF maturation failure
As mentioned above, we have shown that PF4 is upregulated in veins that fail maturation after AVF creation,34 and recently our group found localization of PF4 predominantly in the adventitia (Figure 1). The adventitial layer of veins consists of fibroblasts and may be important for the generation of fibrotic stenosis in failed AVF. Previous studies by our group and others have indicated that the vast majority of cells that comprise the hyperplastic intima are myofibroblasts.296,297 A pig AVG model demonstrated that fibroblasts in the adventitia are able to differentiate into myofibroblasts by expressing αSMA,298 and these myofibroblasts are capable of migrating inward to form IH.298,299 Meanwhile, PF4 can be detected from vascular stenosis tissue derived from chronic thromboembolic pulmonary hypertension (CTEPH) patients together with extracellular collagen I.300 All these results suggest that PF4 may play an important role in adventitial fibroblasts differentiation and stenosis generation. This is in line with our most recent discovery that PF4 treatment of isolated human primary vein fibroblasts could stimulate upregulation of αSMA and ECM components at both RNA and protein levels (Figure 2).
Figure 1.

Immunofluorescent staining of veins with PF4 (red). Stronger PF4 staining are shown in the adventitia (a, d and g). DAPI staining and the merged images are shown in (b), (e), (h), and (c), (f), (i), respectively. Each scale bar represents 10 μm.
Figure 2.

PF4 induces αSMA (ACTA2) and collagen I (Col1A1). The representative western blot is shown in (c). The statistics of the qRT-PCR of ACTA2 and Col1A1, and that of the western blot of ACTA2 and Col1A1 are shown in (a, b, d and e) respectively (n = 4).
Note: Part (c) is a representative figure, the quantification and significance for “*” of part (c) is showing in parts (d) and (e).
Very recently, we have found that the intracellular expression of PF4 and itgb6, the gene encoding for integrin β6 subunit, are associated to each other, and overexpressing PF4 will lead to upregulation of itgb6 at the protein level, while knocking down of PF4 results in downregulation of itgb6 (unpublished data). As mentioned above, integrin αvβ6 is required for the release of the active TGFβ from the LAP-TGFβ complex,129 and TGFβ can directly stimulate the expression of ECM proteins301,302 and is considered the most important factor for fibrosis.107,108
Conclusion and future direction
The molecular cause of AVF maturation failure may be multifactorial but a common manifestation is overt vascular fibrosis combined with IH in the AVF.27 Therefore, finding the mechanisms that promote fibrosis and myofibroblasts activation during post-operative AVF remodeling is crucial to the design of anti-stenotic therapies. There is emerging evidence that reveals a potential involvement of PF4 in fibrosis, including the finding of upregulation of PF4 in veins that fail.34 Hypothetical mechanisms that link upregulation of PF4 with fibrosis onset and AVF maturation failure are summarized in Figure 3. Briefly, increase of PF4 in extracellular space may directly induce upregulation of αSMA and ECM proteins. Molecular features of fibrosis, such as fibroblasts differentiation into myofibroblasts, that is, expression of αSMA and ECM proteins, may be stimulated by PF4 via interaction with its various receptors. In contrast, increase of intracellular PF4 may lead to upregulation of itgb6 and thus elevates the level of αvβ6, which in turn promotes the activation of the TGFβ pathway and subsequently induces fibrosis.
Figure 3.

Potential mechanisms of PF4 involvement in fibrosis. Extracellular PF4 may directly induce upregulation of αSMA and ECM proteins (left-hand-side). In contrast, upregulation of intracellular PF4 may lead to increase of ITGB6 and membrane αvβ6, which may promote TGFβ pathway activation, and subsequently stimulate fibrosis (right-hand-side). All these mechanisms may contribute to fibrosis remodeling associated with AVF maturation failure.
Considering the multiple biological processes in which PF4 is involved including hemostasis, PF4 itself may not be a good therapeutic target to improve AVF maturation.258,259 However, the downstream receptors of this molecule, especially those responsible for fibrosis onset, αSMA, and ECM protein expression, may serve as good candidates for developing therapies to reduce the rate of AVF maturation failure. Therefore, further studies that focus on PF4 and its downstream receptors should be performed to illustrate the molecular mechanisms behind the involvement of PF4 in fibrosis. This may shed light on our goal of improving AVF patency, HD delivery, and the quality of life of ESRD patients.
Funding
The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This manuscript and related studies are funded by Albany Medical College, Albany, New York and Roach Family Fund at The Community Foundation, Albany, New York.
Footnotes
Declaration of conflicting interests
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
References
- 1.Brescia MJ, Cimino JE, Appel K, et al. Chronic hemodialysis using venipuncture and a surgically created arteriovenous fistula. New Engl J Med 1966; 275(20): 1089–1092. [DOI] [PubMed] [Google Scholar]
- 2.USRDS. USRDS annual data report: epidemiology of kidney disease in the United States. Bethesda, MD: National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, 2020. [Google Scholar]
- 3.Foundation NK. 2006 updates clinical practice guidelines and recommendations. New York, NY: National Kidney Foundation, 2006. [Google Scholar]
- 4.Polkinghorne KR, Chin GK, MacGinley RJ, et al. KHACARI Guideline: vascular access – central venous catheters, arteriovenous fistulae and arteriovenous grafts. Nephrology 2013; 18(11): 701–705. [DOI] [PubMed] [Google Scholar]
- 5.Stolic R. Most important chronic complications of arteriovenous fistulas for hemodialysis. Med Princ Pract 2013; 22(3): 220–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hicks CW, Canner JK, Arhuidese I, et al. Mortality benefits of different hemodialysis access types are age dependent. J. Vasc. Surg 2015; 61(2): 449–456. [DOI] [PubMed] [Google Scholar]
- 7.Leermakers JJ, Bode AS, Vaidya A, et al. Cost-effectiveness of vascular access for haemodialysis: arteriovenous fistulas versus arteriovenous grafts. Eur J Vasc Endovasc Surg 2013; 45(1): 84–92. [DOI] [PubMed] [Google Scholar]
- 8.Malas MB, Canner JK, Hicks CW, et al. Trends in incident hemodialysis access and mortality. JAMA Surg 2015; 150(5): 441–448. [DOI] [PubMed] [Google Scholar]
- 9.Nassar GM and Ayus JC. Infectious complications of the hemodialysis access. Kidney Int 2001; 60(1): 1–13. [DOI] [PubMed] [Google Scholar]
- 10.Ramanathan V, Chiu EJ, Thomas JT, et al. Healthcare costs associated with hemodialysis catheter-related infections: a single-center experience. Infect. Control Hosp. Epidemiol 2007; 28(5): 606–609. [DOI] [PubMed] [Google Scholar]
- 11.Woo K, Doros G, Ng T, et al. Comparison of the efficacy of upper arm transposed arteriovenous fistulae and upper arm prosthetic grafts. J Vasc Surg 2009; 50(6): 1405–11.e1. [DOI] [PubMed] [Google Scholar]
- 12.Almasri J, Alsawas M, Mainou M, et al. Outcomes of vascular access for hemodialysis: A systematic review and meta-analysis. J. Vasc. Surg 2016; 64(1): 236–243. [DOI] [PubMed] [Google Scholar]
- 13.Murad MH, Elamin MB, Sidawy AN, et al. Autogenous versus prosthetic vascular access for hemodialysis: a systematic review and meta-analysis. J. Vasc. Surg 2008; 48(5 Suppl): 34S–47S. [DOI] [PubMed] [Google Scholar]
- 14.Lok CE, Huber TS, Lee T, et al. KDOQI clinical practice guideline for vascular access: 2019 update. Am J Kidney Dis 2020; 75(4 Suppl 2): S1–S164. [DOI] [PubMed] [Google Scholar]
- 15.Al-Jaishi AA, Lok CE, Garg AX, et al. Vascular access creation before hemodialysis initiation and use: a populationbased cohort study. Clin J Am Soc Nephrol 2015; 10(3): 418–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Schinstock CA, Albright RC, Williams AW, et al. Outcomes of arteriovenous fistula creation after the fistula first initiative. Clin J Am Soc Nephrol 2011; 6(8): 1996–2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Al-Jaishi AA, Oliver MJ, Thomas SM, et al. Patency rates of the arteriovenous fistula for hemodialysis: a systematic review and meta-analysis. Am J Kidney Dis 2014; 63(3): 464–478. [DOI] [PubMed] [Google Scholar]
- 18.Maya ID and Allon M. Vascular access: core curriculum 2008. Am J Kidney Dis 2008; 51(4): 702–708. [DOI] [PubMed] [Google Scholar]
- 19.Maya ID, O–Neal JC, Young CJ, et al. Outcomes of brachiocephalic fistulas, transposed brachiobasilic fistulas, and upper arm grafts. Clin J Am Soc Nephrol 2009; 4(1): 86–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Pisoni RL, Zepel L, Port FK, et al. Trends in US vascular access use, patient preferences, and related practices: an update from the US DOPPS practice monitor with international comparisons. Am J Kidney Dis 2015; 65(6): 905–915. [DOI] [PubMed] [Google Scholar]
- 21.Ravani P, Spergel LM, Asif A, et al. Clinical epidemiology of arteriovenous fistula in 2007. J. Nephrol 2007; 20(2): 141–149. [PubMed] [Google Scholar]
- 22.Dember LM, Beck GJ, Allon M, et al. Effect of clopidogrel on early failure of arteriovenous fistulas for hemodialysis: a randomized controlled trial. JAMA 2008; 299(18): 2164–2171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lok CE, Allon M, Moist L, et al. Risk equation determining unsuccessful cannulation events and failure to maturation in arteriovenous fistulas (REDUCE FTM I). J Am Soc Nephrol 2006; 17(11): 3204–3212. [DOI] [PubMed] [Google Scholar]
- 24.Tordoir JHM, Zonnebeld N, van Loon MM, et al. Surgical and endovascular intervention for dialysis access maturation failure during and after arteriovenous fistula surgery: review of the evidence. Eur J Vasc Endovasc Surg 2018; 55(2): 240–248. [DOI] [PubMed] [Google Scholar]
- 25.Allon M, Greene T, Dember LM, et al. Association between preoperative vascular function and postoperative arteriovenous fistula development. J Am Soc Nephrol 2016; 27(12): 3788–3795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Simone S, Loverre A, Cariello M, et al. Arteriovenous fistula stenosis in hemodialysis patients is characterized by an increased adventitial fibrosis. J Nephrol 2014; 27(5): 555–562. [DOI] [PubMed] [Google Scholar]
- 27.Martinez L, Duque JC, Tabbara M, et al. Fibrotic venous remodeling and nonmaturation of arteriovenous fistulas. J Am Soc Nephrol 2018; 29(3): 1030–1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Berceli SA, Jiang Z, Klingman NV, et al. Early differential MMP-2 and −9 dynamics during flow-induced arterial and vein graft adaptations. J. Surg. Res 2006; 134(2): 327–334. [DOI] [PubMed] [Google Scholar]
- 29.Brahmbhatt A, Remuzzi A, Franzoni M, et al. The molecular mechanisms of hemodialysis vascular access failure. Kidney Int 2016; 89(2): 303–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lee ES, Shen Q, Pitts RL, et al. Serum metalloproteinases MMP-2, MMP-9, and metalloproteinase tissue inhibitors in patients are associated with arteriovenous fistula maturation. J Vasc Surg 2011; 54(2): 454–460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Aidoudi S and Bikfalvi A. Interaction of PF4 (CXCL4) with the vasculature: a role in atherosclerosis and angiogenesis. Thromb. Haemost 2010; 104(5): 941–948. [DOI] [PubMed] [Google Scholar]
- 32.Schwarz KB, Rosensweig J, Sharma S, et al. Plasma markers of platelet activation in cystic fibrosis liver and lung disease. J. Pediatr. Gastroenterol. Nutr 2003; 37(2): 187–191. [DOI] [PubMed] [Google Scholar]
- 33.Zaldivar MM, Pauels K, von Hundelshausen P, et al. CXC chemokine ligand 4 (Cxcl4) is a platelet-derived mediator of experimental liver fibrosis. Hepatology 2010; 51(4): 1345–1353. [DOI] [PubMed] [Google Scholar]
- 34.Martinez L, Tabbara M, Duque JC, et al. Transcriptomics of human arteriovenous fistula failure: genes associated with nonmaturation. Am J Kidney Dis 2019; 74(1): 73–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Henderson NC, Rieder F and Wynn TA. Fibrosis: from mechanisms to medicines. Nature 2020; 587(7835): 555–566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bataller R and Brenner DA. Liver fibrosis. J. Clin. Investig 2005; 115(2): 209–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Rockey DC, Bell PD and Hill JA. Fibrosis: a common pathway to organ injury and failure. New Engl J Med 2015; 372(12): 1138–1149. [DOI] [PubMed] [Google Scholar]
- 38.Wynn TA. Integrating mechanisms of pulmonary fibrosis. J Exp. Med 2011; 208(7): 1339–1350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Karsdal MA, Nielsen SH, Leeming DJ, et al. The good and the bad collagens of fibrosis: their role in signaling and organ function. Adv Drug Deliv Rev 2017; 121:43–56. [DOI] [PubMed] [Google Scholar]
- 40.Urban ML, Manenti L and Vaglio A. Fibrosis: a common pathway to organ injury and failure. New Engl J Med 2015; 373(1): 95–96. [DOI] [PubMed] [Google Scholar]
- 41.Wynn TA and Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med 2012; 18(7): 1028–1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Duffield JS. Cellular and molecular mechanisms in kidney fibrosis. J. Clin. Investig 2014; 124(6): 2299–2306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Elmufdi F, Henke CA, Perlman DM, et al. Novel mechanisms and treatment of idiopathic pulmonary fibrosis. Discov. Med 2015; 20(109): 145–153. [PubMed] [Google Scholar]
- 44.Mallat A and Lotersztajn S. Cellular mechanisms of tissue fibrosis. 5. Novel insights into liver fibrosis. Am. J. Physiol. Cell Physiol 2013; 305(8): C789–C799. [DOI] [PubMed] [Google Scholar]
- 45.Murtha LA, Schuliga MJ, Mabotuwana NS, et al. The processes and mechanisms of cardiac and pulmonary fibrosis. Front Physiol 2017; 8:777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lan TH, Huang XQ and Tan HM. Vascular fibrosis in atherosclerosis. Cardiovasc Pathol 2013; 22(5): 401–407. [DOI] [PubMed] [Google Scholar]
- 47.Shirwany NA and Zou MH. Arterial stiffness: a brief review. Acta Pharmacol. Sin 2010; 31(10): 1267–1276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ponticos M and Smith BD. Extracellular matrix synthesis in vascular disease: hypertension, and atherosclerosis. J Biomed Res 2014; 28(1): 25–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Touyz RM. Intracellular mechanisms involved in vascular remodelling of resistance arteries in hypertension: role of angiotensin II. Exp. Physiol 2005; 90(4): 449–455. [DOI] [PubMed] [Google Scholar]
- 50.Eming SA, Martin P and Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci. Transl. Med 2014; 6(265): 265sr6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Krenkel O and Tacke F. Liver macrophages in tissue homeostasis and disease. Nat. Rev. Immunol 2017; 17(5): 306–321. [DOI] [PubMed] [Google Scholar]
- 52.Vannella KM and Wynn TA. Mechanisms of organ injury and repair by macrophages. Annu Rev Physiol 2017; 79:593–617. [DOI] [PubMed] [Google Scholar]
- 53.Wynn TA and Vannella KM. Macrophages in tissue repair, regeneration, and fibrosis. Immunity 2016; 44(3): 450–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wynn TA. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat Rev Immunol 2004; 4(8): 583–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hinz B, Phan SH, Thannickal VJ, et al. The myofibroblast: one function, multiple origins. Am. J. Pathol 2007; 170(6): 1807–1816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Mohindra R, Agrawal DK and Thankam FG. Altered vascular extracellular matrix in the pathogenesis of Atherosclerosis. J Cardiovasc Transl Res 2021; 14(4): 647–660. [DOI] [PubMed] [Google Scholar]
- 57.Hardie WD, Glasser SW and Hagood JS. Emerging concepts in the pathogenesis of lung fibrosis. Am J Pathol 2009; 175(1): 3–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Jelaska A and Korn JH. Role of apoptosis and transforming growth factor beta1 in fibroblast selection and activation in systemic sclerosis. Arthritis Rheum 2000; 43(10): 2230–2239. [DOI] [PubMed] [Google Scholar]
- 59.Pechkovsky DV, Hackett TL, An SS, et al. Human lung parenchyma but not proximal bronchi produces fibroblasts with enhanced TGF-β signaling and α-SMA expression. Am J Respir Cell Mol Biol 2010; 43(6): 641–651. [DOI] [PubMed] [Google Scholar]
- 60.Popov Y and Schuppan D. Targeting liver fibrosis: strategies for development and validation of antifibrotic therapies. Hepatology 2009; 50(4): 1294–1306. [DOI] [PubMed] [Google Scholar]
- 61.Cassiman D, Libbrecht L, Desmet V, et al. Hepatic stellate cell/myofibroblast subpopulations in fibrotic human and rat livers. J. Hepatol 2002; 36(2): 200–209. [DOI] [PubMed] [Google Scholar]
- 62.Urushiyama H, Terasaki Y, Nagasaka S, et al. Role of α1 and α2 chains of type IV collagen in early fibrotic lesions of idiopathic interstitial pneumonias and migration of lung fibroblasts. Lab Invest 2015; 95(8): 872–885. [DOI] [PubMed] [Google Scholar]
- 63.Liu B, Itoh H, Louie O, et al. The role of phospholipase C and phosphatidylinositol 3-kinase in vascular smooth muscle cell migration and proliferation. J. Surg. Res 2004; 120(2): 256–265. [DOI] [PubMed] [Google Scholar]
- 64.Adler M, Mayo A, Zhou X, et al. Principles of cell circuits for tissue repair and fibrosis. iScience 2020; 23(2): 100841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Henderson NC, Mackinnon AC, Farnworth SL, et al. Galectin-3 expression and secretion links macrophages to the promotion of renal fibrosis. Am. J. Pathol 2008; 172(2): 288–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lodyga M, Cambridge E, Karvonen HM, et al. Cadherin11-mediated adhesion of macrophages to myofibroblasts establishes a profibrotic niche of active TGF-β. Sci Signal 2019; 12(564): eaao3496. [DOI] [PubMed] [Google Scholar]
- 67.Pakshir P and Hinz B. The big five in fibrosis: macrophages, myofibroblasts, matrix, mechanics, and miscommunication. Matrix Biol 2018; 68–69:81–93. [DOI] [PubMed] [Google Scholar]
- 68.Ramachandran P, Dobie R, Wilson-Kanamori JR, et al. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 2019; 575(7783): 512–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Shook BA, Wasko RR, Rivera-Gonzalez GC, et al. Myofibroblast proliferation and heterogeneity are supported by macrophages during skin repair. Science 2018; 362(6417): eaar2971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Zhou X, Franklin RA, Adler M, et al. Circuit design features of a stable two-cell system. Cell 2018; 172(4): 744–757.e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.El Agha E, Kramann R, Schneider RK, et al. Mesenchymal stem cells in fibrotic disease. Cell Stem Cell 2017; 21(2): 166–177. [DOI] [PubMed] [Google Scholar]
- 72.Kisseleva T, Cong M, Paik Y, et al. Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc Natl Acad Sci 2012; 109(24): 9448–9453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Nattel S. Molecular and cellular mechanisms of atrial fibrosis in atrial fibrillation. JACC Clin Electrophysiol 2017; 3(5): 425–435. [DOI] [PubMed] [Google Scholar]
- 74.Troeger JS, Mederacke I, Gwak GY, et al. Deactivation of hepatic stellate cells during liver fibrosis resolution in mice. Gastroenterology 2012; 143(4): 1073–83.e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Rockey DC, Housset CN and Friedman SL. Activationdependent contractility of rat hepatic lipocytes in culture and in vivo. J. Clin. Investig 1993; 92(4): 1795–1804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Mack M and Yanagita M. Origin of myofibroblasts and cellular events triggering fibrosis. Kidney Int 2015; 87(2): 297–307. [DOI] [PubMed] [Google Scholar]
- 77.Harvey A, Montezano AC, Lopes RA, et al. Vascular fibrosis in aging and hypertension: molecular mechanisms and clinical implications. Can J Cardiol 2016; 32(5): 659–668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Bashey RI, Cox R, McCann J, et al. Changes in collagen biosynthesis, types, and mechanics of aorta in hypertensive rats. J. Lab. Clin. Med 1989; 113(5): 604–611. [PubMed] [Google Scholar]
- 79.Laviades C, Varo N, Fernández J, et al. Abnormalities of the extracellular degradation of collagen type I in essential hypertension. Circulation 1998; 98(6): 535–540. [DOI] [PubMed] [Google Scholar]
- 80.Ban CR and Twigg SM. Fibrosis in diabetes complications: pathogenic mechanisms and circulating and urinary markers. Vasc. Health Risk Manag 2008; 4(3): 575–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lakatta EG. The reality of aging viewed from the arterial wall. Artery Res 2013; 7(2): 73–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Arribas SM, Hinek A and González MC. Elastic fibres and vascular structure in hypertension. Pharmacol. Ther 2006; 111(3): 771–791. [DOI] [PubMed] [Google Scholar]
- 83.Lakatta EG and Levy D. Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises. Part I. Aging arteries: a “set up” for vascular disease. Circulation 2003; 107(1): 139–146. [DOI] [PubMed] [Google Scholar]
- 84.Eddy AA, Liu E and McCulloch L. Interstitial inflammation and fibrosis in rats with diet-induced hypercholesterolemia. Kidney Int 1996; 50(4): 1139–1149. [DOI] [PubMed] [Google Scholar]
- 85.Eddy AA. Interstitial fibrosis in hypercholesterolemic rats: role of oxidation, matrix synthesis, and proteolytic cascades. Kidney Int 1998; 53(5): 1182–1189. [DOI] [PubMed] [Google Scholar]
- 86.Chade AR, Mushin OP, Zhu X, et al. Pathways of renal fibrosis and modulation of matrix turnover in experimental hypercholesterolemia. Hypertension 2005; 46(4): 772–779. [DOI] [PubMed] [Google Scholar]
- 87.Ding G, van Goor H, Ricardo SD, et al. Oxidized LDL stimulates the expression of TGF-beta and fibronectin in human glomerular epithelial cells. Kidney Int 1997; 51(1): 147–154. [DOI] [PubMed] [Google Scholar]
- 88.Song CY, Kim BC, Hong HK, et al. Oxidized LDL activates PAI-1 transcription through autocrine activation of TGF-beta signaling in mesangial cells. Kidney Int 2005; 67(5): 1743–1752. [DOI] [PubMed] [Google Scholar]
- 89.Testai L, Brancaleone V, Flori L, et al. Modulation of EndMT by hydrogen sulfide in the prevention of cardiovascular fibrosis. Antioxidants 2021; 10(6): 910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Tyagi SC, et al. Reduction-oxidation (redox) and vascular tissue level of homocyst(e)ine in human coronary atherosclerotic lesions and role in extracellular matrix remodeling and vascular tone. Mol Cell Biochem 1998; 181(1–2): 107–116. [DOI] [PubMed] [Google Scholar]
- 91.Mujumdar VS, Hayden MR and Tyagi SC. Homocyst(e)ine induces calcium second messenger in vascular smooth muscle cells. J Cell Physiol 2000; 183(1): 28–36. [DOI] [PubMed] [Google Scholar]
- 92.Andreea SI, Marieta C and Anca D. AGEs and glucose levels modulate type I and III procollagen mRNA synthesis in dermal fibroblasts cells culture. Exp. Diabetes Res 2008; 2008:473603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Leehey DJ, Singh AK, Alavi N, et al. Role of angiotensin II in diabetic nephropathy. Kidney Int Suppl 2000; 77:S93–S98. [DOI] [PubMed] [Google Scholar]
- 94.Martínez-Martínez E, Miana M, Jurado-López R, et al. The potential role of leptin in the vascular remodeling associated with obesity. Int. J. Obes 2014; 38(12): 1565–1572. [DOI] [PubMed] [Google Scholar]
- 95.Jia G and Sowers JR. Endothelial dysfunction potentially interacts with impaired glucose metabolism to increase cardiovascular risk. Hypertension 2014; 64(6): 1192–1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Böttinger EP and Bitzer M. TGF-beta signaling in renal disease. J Am Soc Nephrol 2002; 13(10): 2600–2610. [DOI] [PubMed] [Google Scholar]
- 97.Eddy AA. Overview of the cellular and molecular basis of kidney fibrosis. Kidney Int Suppl 2014; 4(1): 2–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Eddy AA and Neilson EG. Chronic kidney disease progression. J Am Soc Nephrol 2006; 17(11): 2964–2966. [DOI] [PubMed] [Google Scholar]
- 99.Thannickal VJ, Toews GB, White ES, et al. Martinez FJ. Mechanisms of pulmonary fibrosis. Annu Rev Med 2004; 55:395–417. [DOI] [PubMed] [Google Scholar]
- 100.Allen RJ, et al. Genetic variants associated with susceptibility to idiopathic pulmonary fibrosis in people of European ancestry: a genome-wide association study. Lancet Respir Med 2017; 5(11): 869–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Fingerlin TE, Murphy E, Zhang W, et al. Genome-wide association study identifies multiple susceptibility loci for pulmonary fibrosis. Nat Genet 2013; 45(6): 613–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Gansner JM, Rosas IO and Ebert BL. Pulmonary fibrosis, bone marrow failure, and telomerase mutation. New Engl. J. Med 2012; 366(16): 1551–1553. [DOI] [PubMed] [Google Scholar]
- 103.Hunninghake GM, Hatabu H, Okajima Y, et al. MUC5B promoter polymorphism and interstitial lung abnormalities. New Engl J Med 2013; 368(23):2192–2200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kim HY, Park JE, Lee SC, et al. Genotype-related clinical characteristics and myocardial fibrosis and their association with prognosis in hypertrophic cardiomyopathy. J Clin Med 2020; 9(6): 1671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Young CNJ, Gosselin MRF, Rumney R, et al. Total absence of dystrophin expression exacerbates ectopic myofiber calcification and fibrosis and alters macrophage infiltration patterns. Am J Pathol 2020; 190(1): 190–205. [DOI] [PubMed] [Google Scholar]
- 106.Biernacka A, Dobaczewski M and Frangogiannis NG. TGF-β signaling in fibrosis. Growth Factors 2011; 29(5): 196–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Leask A and Abraham DJ. TGF-beta signaling and the fibrotic response. FASEB J 2004; 18(7): 816–827. [DOI] [PubMed] [Google Scholar]
- 108.Meng XM, Nikolic-Paterson DJ and Lan HY. TGF-β: the master regulator of fibrosis. Nat Rev Nephrol 2016; 12(6): 325–338. [DOI] [PubMed] [Google Scholar]
- 109.Piersma B, Bank RA and Boersema M. Signaling in fibrosis: TGF-β, WNT, and YAP/TAZ converge. Front Med 2015; 2:59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Chen B, Huang S, Su Y, et al. Macrophage Smad3 protects the infarcted heart, stimulating phagocytosis and regulating inflammation. Circ Res 2019; 125(1): 55–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Frangogiannis N Transforming growth factor-β in tissue fibrosis. J Exp Med 2020; 217(3): e20190103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Zeisberg M, Hanai JI, Sugimoto H, et al. BMP-7 counteracts TGF-β1–induced epithelial-to-mesenchymal transition and reverses chronic renal injury. Nat Med 2003; 9(7): 964–968. [DOI] [PubMed] [Google Scholar]
- 113.Duncan MR, Frazier KS, Abramson S, et al. Connective tissue growth factor mediates transforming growth factor betainduced collagen synthesis: down-regulation by cAMP. FASEB J 1999; 13(13): 1774–1786. [PubMed] [Google Scholar]
- 114.Yamanaka M, Shegogue D, Pei H, et al. Sphingosine kinase 1 (SPHK1) is induced by transforming growth factor-beta and mediates TIMP-1 up-regulation. J Biol Chem 2004; 279(52): 53994–54001. [DOI] [PubMed] [Google Scholar]
- 115.Lodyga M and Hinz B. TGF-β1: a truly transforming growth factor in fibrosis and immunity. Semin Cell Dev Biol 2020; 101:123–139. [DOI] [PubMed] [Google Scholar]
- 116.Heldin CH and Moustakas A. Signaling receptors for TGF-β family members. Cold Spring Harb Perspect Biol 2016; 8(8): a022053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Horiguchi M, Ota M and Rifkin DB. Matrix control of transforming growth factor-β function. J Biochem 2012; 152(4): 321–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Annes JP, Munger JS and Rifkin DB. Making sense of latent TGFbeta activation. J. Cell Sci 2003; 116(Pt 2): 217–224. [DOI] [PubMed] [Google Scholar]
- 119.Bourd-Boittin K, Bonnier D, Leyme A, et al. Protease profiling of liver fibrosis reveals the ADAM metallopeptidase with thrombospondin type 1 motif, 1 as a central activator of transforming growth factor beta. Hepatology 2011; 54(6): 2173–2184. [DOI] [PubMed] [Google Scholar]
- 120.Briassouli P, Rifkin D, Clancy RM, et al. Binding of antiSSA antibodies to apoptotic fetal cardiocytes stimulates urokinase plasminogen activator (uPA)/uPA receptordependent activation of TGF-β and potentiates fibrosis. J Immunol 2011; 187(10): 5392–5401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Edgtton KL, Gow RM, Kelly DJ, et al. Plasmin is not protective in experimental renal interstitial fibrosis. Kidney Int 2004; 66(1): 68–76. [DOI] [PubMed] [Google Scholar]
- 122.Khalil N, Corne S, Whitman C, et al. Plasmin regulates the activation of cell-associated latent TGF-beta 1 secreted by rat alveolar macrophages after in vivo bleomycin injury. Am J Respir Cell Mol Biol 1996; 15(2): 252–259. [DOI] [PubMed] [Google Scholar]
- 123.Okuno M, Akita K, Moriwaki H, et al. Prevention of rat hepatic fibrosis by the protease inhibitor, camostat mesilate, via reduced generation of active TGF-beta. Gastroenterology 2001; 120(7): 1784–1800. [DOI] [PubMed] [Google Scholar]
- 124.Shea BS, Probst CK, Brazee PL, et al. Uncoupling of the profibrotic and hemostatic effects of thrombin in lung fibrosis. JCI Insight 2017; 2(9): e86608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Yao Y, Hu C, Song Q, et al. ADAMTS16 activates latent TGF-β, accentuating fibrosis and dysfunction of the pressure-overloaded heart. Cardiovasc Res 2020; 116(5): 956–969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Asano Y, Ihn H, Yamane K, et al. Increased expression of integrin αvβ3 contributes to the establishment of autocrine TGF-β signaling in scleroderma fibroblasts. J Immunol 2005; 175(11): 7708–7718. [DOI] [PubMed] [Google Scholar]
- 127.Asano Y, Ihn H, Yamane K, et al. Involvement of alphavbeta5 integrin-mediated activation of latent transforming growth factor beta1 in autocrine transforming growth factor beta signaling in systemic sclerosis fibroblasts. Arthritis Rheum 2005; 52(9): 2897–2905. [DOI] [PubMed] [Google Scholar]
- 128.Häkkinen L, Koivisto L, Gardner H, et al. Increased expression of beta6-integrin in skin leads to spontaneous development of chronic wounds. Am J Pathol 2004; 164(1): 229–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Munger JS, Huang X, Kawakatsu H, et al. The integrin alpha v beta 6 binds and activates latent TGF beta 1: a mechanism for regulating pulmonary inflammation and fibrosis. Cell 1999; 96(3): 319–328. [DOI] [PubMed] [Google Scholar]
- 130.Sarrazy V, Koehler A, Chow ML, et al. Integrins αvβ5 and αvβ3 promote latent TGF-β1 activation by human cardiac fibroblast contraction. Cardiovasc Res 2014; 102(3): 407–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Scotton CJ, Krupiczojc MA, Königshoff M, et al. Increased local expression of coagulation factor X contributes to the fibrotic response in human and murine lung injury. J Clin Investig 2009; 119(9): 2550–2563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Tatler AL, John AE, Jolly L, et al. Integrin αvβ5-mediated TGF-β activation by airway smooth muscle cells in asthma. Immunol 2011; 187(11): 6094–6107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Frangogiannis NG. Matricellular proteins in cardiac adaptation and disease. Physiol. Rev 2012; 92(2): 635–688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Klingberg F, Chau G, Walraven M, et al. The ED-A domain enhances the capacity of fibronectin to store latent TGF-β binding protein-1 in the fibroblast matrix. J Cell Sci 2018; 131(5): jcs201293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Liu G, Cooley MA, Jarnicki AG, et al. Fibulin-1c regulates transforming growth factor-β activation in pulmonary tissue fibrosis. JCI Insight 2019; 5(16): e124529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Serini G, Bochaton-Piallat ML, Ropraz P, et al. The fibronectin domain ED-A is crucial for myofibroblastic phenotype induction by transforming growth factor-beta1. J. Cell Biol 1998; 142(3): 873–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Robertson IB and Rifkin DB. Regulation of the bioavailability of TGF-β and TGF-β-related proteins. Cold Spring Harb Perspect Biol 2016; 8(6): a021907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Amălinei C, Căruntu ID, Giuşcă SE, et al. Matrix metalloproteinases involvement in pathologic conditions. Rom J Morphol Embryol 2010; 51(2): 215–228. [PubMed] [Google Scholar]
- 139.Giannandrea M and Parks WC. Diverse functions of matrix metalloproteinases during fibrosis. Dis. Model Mech 2014; 7(2): 193–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Jia G, Hill MA and Sowers JR. Diabetic cardiomyopathy: an update of mechanisms contributing to this clinical entity. Circ Res 2018; 122(4): 624–638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Prakobwong S, Yongvanit P, Hiraku Y, et al. Involvement of MMP-9 in peribiliary fibrosis and cholangiocarcinogenesis via Rac1-dependent DNA damage in a hamster model. Int J Cancer 2010; 127(11): 2576–2587. [DOI] [PubMed] [Google Scholar]
- 142.Verrecchia F and Mauviel A. Transforming growth factor-beta and fibrosis. World J Gastroenterol 2007; 13(22): 3056–3062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Mishra R, Cool BL, Laderoute KR, et al. AMP-activated protein kinase inhibits transforming growth factor-betainduced Smad3-dependent transcription and myofibroblast transdifferentiation. J Biol Chem 2008; 283(16): 10461–10469. [DOI] [PubMed] [Google Scholar]
- 144.Morikawa M, Derynck R and Miyazono K. TGF-β and the TGF-β family: context-dependent roles in cell and tissue physiology. Cold Spring Harb Perspect Biol 2016; 8(5): a021873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Schnaper HW, Hayashida T, Hubchak SC, et al. TGF-beta signal transduction and mesangial cell fibrogenesis. Am J Physiol Renal Physiol 2003; 284(2): F243–F252. [DOI] [PubMed] [Google Scholar]
- 146.Datta PK, Blake MC and Moses HL. Regulation of plasminogen activator inhibitor-1 expression by transforming growth factor-beta -induced physical and functional interactions between Smads and Sp1. J Biol Chem 2000; 275(51): 40014–40019. [DOI] [PubMed] [Google Scholar]
- 147.Desmoulière A, Geinoz A, Gabbiani F, et al. Transforming growth factor-beta 1 induces alpha-smooth muscle actin expression in granulation tissue myofibroblasts and in quiescent and growing cultured fibroblasts. J. Cell Biol 1993; 122(1): 103–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Douillet CD, Velarde V, Christopher JT, et al. Mechanisms by which bradykinin promotes fibrosis in vascular smooth muscle cells: role of TGF-beta and MAPK. Am J Physiol Heart Circ Physiol 2000; 279(6): H2829–H2837. [DOI] [PubMed] [Google Scholar]
- 149.O’Callaghan CJ and Williams B. Mechanical strain-induced extracellular matrix production by human vascular smooth muscle cells: role of TGF-beta(1). Hypertension 2000; 36(3): 319–324. [DOI] [PubMed] [Google Scholar]
- 150.Roberts AB, Sporn MB, Assoian RK, et al. Transforming growth factor type beta: rapid induction of fibrosis and angiogenesis in vivo and stimulation of collagen formation in vitro. Proc Natl Acad Sci USA 1986; 83(12): 4167–4171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Samarakoon R, Higgins SP, Higgins CE, et al. The TGF-β1/p53/PAI-1 signaling axis in vascular senescence: role of caveolin-1. Biomolecules 2019; 9(8): 341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Hinz B, Mastrangelo D, Iselin CE, et al. Mechanical tension controls granulation tissue contractile activity and myofibroblast differentiation. Am. J. Pathol 2001; 159(3): 1009–1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Hinz B, Celetta G, Tomasek JJ, et al. Alpha-smooth muscle actin expression upregulates fibroblast contractile activity. Mol. Biol. Cell 2001; 12(9): 2730–2741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Rupérez M, Lorenzo O, Blanco-Colio LM, et al. Connective tissue growth factor is a mediator of angiotensin II-induced fibrosis. Circulation 2003; 108(12): 1499–1505. [DOI] [PubMed] [Google Scholar]
- 155.Thum T, Gross C, Fiedler J, et al. MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts. Nature 2008; 456(7224): 980–984. [DOI] [PubMed] [Google Scholar]
- 156.Funaba M, Zimmerman CM and Mathews LS. Modulation of Smad2-mediated signaling by extracellular signal-regulated kinase. J Biol Chem 2002; 277(44): 41361–41368. [DOI] [PubMed] [Google Scholar]
- 157.Furukawa F, Matsuzaki K, Mori S, et al. p38 MAPK mediates fibrogenic signal through Smad3 phosphorylation in rat myofibroblasts. Hepatology 2003; 38(4): 879–889. [DOI] [PubMed] [Google Scholar]
- 158.Kretzschmar M, Doody J, Timokhina I, et al. A mechanism of repression of TGFbeta/Smad signaling by oncogenic Ras. Genes Dev 1999; 13(7): 804–816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Lee MK, Pardoux C, Hall MC, et al. TGF-beta activates Erk MAP kinase signalling through direct phosphorylation of ShcA. EMBO J 2007; 26(17): 3957–3967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Seay U, Sedding D, Krick S, et al. Transforming growth factor-beta-dependent growth inhibition in primary vascular smooth muscle cells is p38-dependent. J Pharmacol Exp Ther 2005; 315(3): 1005–1012. [DOI] [PubMed] [Google Scholar]
- 161.Yoshida K, Matsuzaki K, Mori S, et al. Transforming growth factor-beta and platelet-derived growth factor signal via c-Jun N-terminal kinase-dependent Smad2/3 phosphorylation in rat hepatic stellate cells after acute liver injury. Am J Pathol 2005; 166(4): 1029–1039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Yu B, Sladojevic N, Blair JE, et al. Targeting Rho-associated coiled-coil forming protein kinase (ROCK) in cardiovascular fibrosis and stiffening. Expert Opin Ther Targets 2020; 24(1): 47–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Zhang YE. Non-Smad signaling pathways of the TGF-β family. Cold Spring Harb Perspect Biol 2017; 9(2): a022129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Derynck R and Zhang YE. Smad-dependent and Smadindependent pathways in TGF-beta family signalling. Nature 2003; 425(6958): 577–584. [DOI] [PubMed] [Google Scholar]
- 165.Dawson K, Wakili R, Ordög B, et al. MicroRNA29: a mechanistic contributor and potential biomarker in atrial fibrillation. Circulation 2013; 127(14): 1466–1475e1. [DOI] [PubMed] [Google Scholar]
- 166.Lakner AM, Steuerwald NM, Walling TL, et al. Inhibitory effects of microRNA 19b in hepatic stellate cell-mediated fibrogenesis. Hepatology 2012; 56(1): 300–310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Putta S, Lanting L, Sun G, et al. Inhibiting microRNA-192 ameliorates renal fibrosis in diabetic nephropathy. J Am Soc Nephrol 2012; 23(3): 458–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.van Rooij E and Olson EN. Searching for miR-acles in cardiac fibrosis. Circ Res 2009; 104(2): 138–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Li Y, Jiang D, Liang J, et al. Severe lung fibrosis requires an invasive fibroblast phenotype regulated by hyaluronan and CD44. J Exp Med 2011; 208(7): 1459–1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Koelink PJ, Overbeek SA, Braber S, et al. Collagen degradation and neutrophilic infiltration: a vicious circle in inflammatory bowel disease. Gut 2014; 63(4): 578–587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Rodríguez D, Morrison CJ and Overall CM. Matrix metalloproteinases: what do they not do? New substrates and biological roles identified by murine models and proteomics. Biochim Biophys Acta 2010; 1803(1): 39–54. [DOI] [PubMed] [Google Scholar]
- 172.Akthar S, Patel DF, Beale RC, et al. Matrikines are key regulators in modulating the amplitude of lung inflammation in acute pulmonary infection. Nat Commun 2015; 6:8423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Ruiz-Ortega M, Lorenzo O, Rupérez M, et al. Role of the renin-angiotensin system in vascular diseases: expanding the field. Hypertension 2001; 38(6): 1382–1387. [DOI] [PubMed] [Google Scholar]
- 174.Border WA and Noble NA. Interactions of transforming growth factor-beta and angiotensin II in renal fibrosis. Hypertension 1998; 31(1 Pt 2): 181–188. [DOI] [PubMed] [Google Scholar]
- 175.Wolf G Renal injury due to renin-angiotensin-aldosterone system activation of the transforming growth factor-beta pathway. Kidney Int 2006; 70(11): 1914–1919. [DOI] [PubMed] [Google Scholar]
- 176.Song JJ, Yang M, Liu Y, et al. MicroRNA-122 aggravates angiotensin II-mediated apoptosis and autophagy imbalance in rat aortic adventitial fibroblasts via the modulation of SIRT6-elabela-ACE2 signaling. Eur. J. Pharmacol 2020; 883:173374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Xu R, Zhang ZZ, Chen LJ, et al. Ascending aortic adventitial remodeling and fibrosis are ameliorated with Apelin-13 in rats after TAC via suppression of the miRNA-122 and LGR4-β-catenin signaling. Peptides 2016; 86:85–94. [DOI] [PubMed] [Google Scholar]
- 178.Gibbons GH, Pratt RE and Dzau VJ. Vascular smooth muscle cell hypertrophy vs. hyperplasia. Autocrine transforming growth factor-beta 1 expression determines growth response to angiotensin II. J Clin Investig 1992; 90(2): 456–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Itoh H, Mukoyama M, Pratt RE, et al. Multiple autocrine growth factors modulate vascular smooth muscle cell growth response to angiotensin II. J. Clin. Investig 1993; 91(5): 2268–2274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Rodríguez-Vita J, Sánchez-López E, Esteban V, et al. Angiotensin II activates the Smad pathway in vascular smooth muscle cells by a transforming growth factor-betaindependent mechanism. Circulation 2005; 111(19): 2509–2517. [DOI] [PubMed] [Google Scholar]
- 181.Sucosky P, Balachandran K, Elhammali A, et al. Altered shear stress stimulates upregulation of endothelial VCAM-1 and ICAM-1 in a BMP-4- and TGF-β1-dependent pathway. Arterioscler Thromb Vasc Biol 2008; 29(2): 254–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Rhyu DY, Yang Y, Ha H, et al. Role of reactive oxygen species in TGF-beta1-induced mitogen-activated protein kinase activation and epithelial-mesenchymal transition in renal tubular epithelial cells. J Am Soc Nephrol 2005; 16(3): 667–675. [DOI] [PubMed] [Google Scholar]
- 183.Carver KA, Smith TL, Gallagher PE, et al. Angiotensin-(1–7) prevents angiotensin II-induced fibrosis in cremaster microvessels. Microcirculation 2015; 22(1): 19–27. [DOI] [PubMed] [Google Scholar]
- 184.Montezano AC, Burger D, Paravicini TM, et al. Nicotinamide adenine dinucleotide phosphate reduced oxidase 5 (Nox5) regulation by angiotensin II and endothelin-1 is mediated via calcium/calmodulin-dependent, rac-1-independent pathways in human endothelial cells. Circ Res 2010; 106(8): 1363–1373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Qi G, Jia L, Li Y, et al. Angiotensin II infusion-induced inflammation, monocytic fibroblast precursor infiltration, and cardiac fibrosis are pressure dependent. Cardiovasc. Toxicol 2011; 11(2): 157–167. [DOI] [PubMed] [Google Scholar]
- 186.Weigert C, Brodbeck K, Klopfer K, et al. Angiotensin II induces human TGF-beta 1 promoter activation: similarity to hyperglycaemia. Diabetologia 2002; 45(6): 890–898. [DOI] [PubMed] [Google Scholar]
- 187.Chen B, Wang P, Brem A, et al. Mineralocorticoid receptor: a hidden culprit for hemodialysis vascular access dysfunction. EBioMedicine 2019; 39:621–627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Ehsan A, McGraw AP, Aronovitz MJ, et al. Mineralocorticoid receptor antagonism inhibits vein graft remodeling in mice. J Thorac Cardiovasc Surg 2013; 145(6): 1642–1649.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Ishidoya S, Morrissey J, McCracken R, et al. Angiotensin II receptor antagonist ameliorates renal tubulointerstitial fibrosis caused by unilateral ureteral obstruction. Kidney Int 1995; 47(5): 1285–1294. [DOI] [PubMed] [Google Scholar]
- 190.Ruiz-Ortega M, González S, Serón D, et al. ACE inhibition reduces proteinuria, glomerular lesions and extracellular matrix production in a normotensive rat model of immune complex nephritis. Kidney Int 1995; 48(6): 1778–1791. [DOI] [PubMed] [Google Scholar]
- 191.Zhang Z, Chen L, Zhong J, et al. ACE2/Ang-(1–7) signaling and vascular remodeling. Sci. China Life Sci 2014; 57(8): 802–808. [DOI] [PubMed] [Google Scholar]
- 192.Song B, Zhang ZZ, Zhong JC, et al. Loss of angiotensinconverting enzyme 2 exacerbates myocardial injury via activation of the CTGF-fractalkine signaling pathway. Circ J 2013; 77(12): 2997–3006. [DOI] [PubMed] [Google Scholar]
- 193.Zhong J, Basu R, Guo D, et al. Angiotensin-converting enzyme 2 suppresses pathological hypertrophy, myocardial fibrosis, and cardiac dysfunction. Circulation 2010; 122(7): 717–728. [DOI] [PubMed] [Google Scholar]
- 194.Sampaio WO, Souza dos Santos RA, Faria-Silva R, et al. Angiotensin-(1–7) through receptor Mas mediates endothelial nitric oxide synthase activation via Akt-dependent pathways. Hypertension 2007; 49(1): 185–192. [DOI] [PubMed] [Google Scholar]
- 195.Song B, Jin H, Yu X, et al. Angiotensin-converting enzyme 2 attenuates oxidative stress and VSMC proliferation via the JAK2/STAT3/SOCS3 and profilin-1/MAPK signaling pathways. Regul Pept 2013; 185:44–51. [DOI] [PubMed] [Google Scholar]
- 196.Tallant EA, Ferrario CM and Gallagher PE. Angiotensin-(1–7) inhibits growth of cardiac myocytes through activation of the mas receptor. Am J Physiol Heart Circ Physiol 2005; 289(4): H1560–H1566. [DOI] [PubMed] [Google Scholar]
- 197.Laggner H, Hermann M, Esterbauer H, et al. The novel gaseous vasorelaxant hydrogen sulfide inhibits angiotensinconverting enzyme activity of endothelial cells. J. Hypertens 2007; 25(10): 2100–2104. [DOI] [PubMed] [Google Scholar]
- 198.Meng G, Ma Y, Xie L, et al. Emerging role of hydrogen sulfide in hypertension and related cardiovascular diseases. Br J Pharmacol 2015; 172(23): 5501–5511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Snijder PM, Frenay AR, de Boer RA, et al. Exogenous administration of thiosulfate, a donor of hydrogen sulfide, attenuates angiotensin II-induced hypertensive heart disease in rats. Br J Pharmacol 2015; 172(6): 1494–1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Sen U, Mishra PK, Tyagi N, et al. Homocysteine to hydrogen sulfide or hypertension. Cell Biochem. Biophys 2010; 57(2–3): 49–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Russo I and Frangogiannis NG. Diabetes-associated cardiac fibrosis: cellular effectors, molecular mechanisms and therapeutic opportunities. J Mol Cell Cardiol 2016; 90:84–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Hsieh CC and Papaconstantinou J. The effect of aging on p38 signaling pathway activity in the mouse liver and in response to ROS generated by 3-nitropropionic acid. Mech. Ageing Dev 2002; 123(11): 1423–1435. [DOI] [PubMed] [Google Scholar]
- 203.Li Z, Li J, Bu X, et al. Age-induced augmentation of p38 MAPK phosphorylation in mouse lung. Exp Gerontol 2011; 46(8): 694–702. [DOI] [PubMed] [Google Scholar]
- 204.Wu Z, Yu Y, Liu C, et al. Role of p38 mitogen-activated protein kinase in vascular endothelial aging: interaction with Arginase-II and S6K1 signaling pathway. Aging 2015; 7(1): 70–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Briones AM, Nguyen Dinh Cat A, Callera GE, et al. Adipocytes produce aldosterone through calcineurindependent signaling pathways: implications in diabetes mellitus-associated obesity and vascular dysfunction. Hypertension 2012; 59(5): 1069–1078. [DOI] [PubMed] [Google Scholar]
- 206.Savoia C, Touyz RM, Amiri F, et al. Selective mineralocorticoid receptor blocker eplerenone reduces resistance artery stiffness in hypertensive patients. Hypertension 2008; 51(2): 432–439. [DOI] [PubMed] [Google Scholar]
- 207.Brown NJ, Nakamura S, Ma L, et al. Aldosterone modulates plasminogen activator inhibitor-1 and glomerulosclerosis in vivo. Kidney Int 2000; 58(3): 1219–1227. [DOI] [PubMed] [Google Scholar]
- 208.Lee YS, Kim JA, Kim KL, et al. Aldosterone upregulates connective tissue growth factor gene expression via p38 MAPK pathway and mineralocorticoid receptor in ventricular myocytes. J Korean Med Sci 2004; 19(6): 805–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Calvier L, Miana M, Reboul P, et al. Galectin-3 mediates aldosterone-induced vascular fibrosis. Arterioscler Thromb Vasc Biol 2013; 33(1): 67–75. [DOI] [PubMed] [Google Scholar]
- 210.Hafizi S, Wharton J, Chester AH, et al. Profibrotic effects of endothelin-1 via the ETA receptor in cultured human cardiac fibroblasts. Cell Physiol Biochem 2004; 14(4–6): 285–292. [DOI] [PubMed] [Google Scholar]
- 211.Horstmeyer A, Licht C, Scherr G, et al. Signalling and regulation of collagen I synthesis by ET-1 and TGF-beta1. FEBS J 2005; 272(24): 6297–6309. [DOI] [PubMed] [Google Scholar]
- 212.Makita Z, Bucala R, Rayfield EJ, et al. Reactive glycosylation endproducts in diabetic uraemia and treatment of renal failure. Lancet 1994; 343(8912): 1519–1522. [DOI] [PubMed] [Google Scholar]
- 213.Cooper ME. Importance of advanced glycation end products in diabetes-associated cardiovascular and renal disease. Am. J. Hypertens 2004; 17(12 Pt 2): 31S–38S. [DOI] [PubMed] [Google Scholar]
- 214.Reddy GK. AGE-related cross-linking of collagen is associated with aortic wall matrix stiffness in the pathogenesis of drug-induced diabetes in rats. Microvasc. Res 2004; 68(2): 132–142. [DOI] [PubMed] [Google Scholar]
- 215.Throckmorton DC, Brogden AP, Min B, et al. PDGF and TGF-beta mediate collagen production by mesangial cells exposed to advanced glycosylation end products. Kidney Int 1995; 48(1): 111–117. [DOI] [PubMed] [Google Scholar]
- 216.Torjesen AA, Sigurðsson S, Westenberg JJ, et al. Pulse pressure relation to aortic and left ventricular structure in the age, gene/environment susceptibility (AGES)-Reykjavik study. Hypertension 2014; 64(4): 756–761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Galis ZS and Khatri JJ. Matrix metalloproteinases in vascular remodeling and atherogenesis: the good, the bad, and the ugly. Circ Res 2002; 90(3): 251–262. [PubMed] [Google Scholar]
- 218.Newby AC. Matrix metalloproteinases regulate migration, proliferation, and death of vascular smooth muscle cells by degrading matrix and non-matrix substrates. Cardiovasc. Res 2006; 69(3): 614–624. [DOI] [PubMed] [Google Scholar]
- 219.Deenadayalu V, Puttabyatappa Y, Liu AT, et al. Testosterone-induced relaxation of coronary arteries: activation of BKCa channels via the cgmp-dependent protein kinase. Am J Physiol Heart Circ Physiol 2012; 302(1): H115–H123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Shinde AV, Su Y, Palanski BA, et al. Pharmacologic inhibition of the enzymatic effects of tissue transglutaminase reduces cardiac fibrosis and attenuates cardiomyocyte hypertrophy following pressure overload. J Mol Cell Cardiol 2018; 117:36–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Steppan J, Bergman Y, Viegas K, et al. Tissue transglutaminase modulates vascular stiffness and function through crosslinking-dependent and crosslinking-independent functions. J Am Heart Assoc 2017; 6(2): e004161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Ivanov S, Scallan JP, Kim KW, et al. CCR7 and IRF4dependent dendritic cells regulate lymphatic collecting vessel permeability. J Clin Investig 2016; 126(4): 1581–1591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Surendran K, Schiavi S and Hruska KA. Wnt-dependent beta-catenin signaling is activated after unilateral ureteral obstruction, and recombinant secreted frizzled-related protein 4 alters the progression of renal fibrosis. J Am Soc Nephrol 2005; 16(8): 2373–2384. [DOI] [PubMed] [Google Scholar]
- 224.Wei J, Melichian D, Komura K, et al. Canonical Wnt signaling induces skin fibrosis and subcutaneous lipoatrophy: a novel mouse model for scleroderma? Arthritis Rheum 2011; 63(6): 1707–1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Königshoff M, Kramer M, Balsara N, et al. WNT1-inducible signaling protein-1 mediates pulmonary fibrosis in mice and is upregulated in humans with idiopathic pulmonary fibrosis. J Clin Investig 2009; 119(4): 772–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Wang D, Dai C, Li Y, et al. Canonical Wnt/β-catenin signaling mediates transforming growth factor-β1-driven podocyte injury and proteinuria. Kidney Int 2011; 80(11): 1159–1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Poon R, Nik SA, Ahn J, et al. Beta-catenin and transforming growth factor beta have distinct roles regulating fibroblast cell motility and the induction of collagen lattice contraction. BMC Cell Biol 2009; 10:38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Feng W, Li W, Liu W, et al. IL-17 induces myocardial fibrosis and enhances RANKL/OPG and MMP/TIMP signaling in isoproterenol-induced heart failure. Exp Mol Pathol 2009; 87(3): 212–218. [DOI] [PubMed] [Google Scholar]
- 229.Meng F, Wang K, Aoyama T, et al. Interleukin-17 signaling in inflammatory, Kupffer cells, and hepatic stellate cells exacerbates liver fibrosis in mice. Gastroenterology 2012; 143(3): 765–776.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Park MJ, Moon SJ, Lee EJ, et al. IL-1-IL-17 signaling axis contributes to fibrosis and inflammation in two different murine models of systemic sclerosis. Front Immunol 2018; 9:1611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Sun B, Wang H, Zhang L, et al. Role of interleukin 17 in TGF-β signaling-mediated renal interstitial fibrosis. Cytokine 2018; 106:80–88. [DOI] [PubMed] [Google Scholar]
- 232.Wang BZ, Wang LP, Han H, et al. Interleukin-17A antagonist attenuates radiation-induced lung injuries in mice. Exp Lung Res 2014; 40(2): 77–85. [DOI] [PubMed] [Google Scholar]
- 233.Wilson MS, Madala SK, Ramalingam TR, et al. Bleomycin and IL-1beta-mediated pulmonary fibrosis is IL-17A dependent. J Exp Med 2010; 207(3): 535–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Wohlfahrt T, Rauber S, Uebe S, et al. PU.1 controls fibroblast polarization and tissue fibrosis. Nature 2019; 566(7744): 344–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Elhai M, Avouac J, Hoffmann-Vold AM, et al. OX40L blockade protects against inflammation-driven fibrosis. Proc Natl Acad Sci USA 2016; 113(27): E3901–E3910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Abreu MT, Taylor KD, Lin YC, et al. Mutations in NOD2 are associated with fibrostenosing disease in patients with Crohn’s disease. Gastroenterology 2002; 123(3): 679–688. [DOI] [PubMed] [Google Scholar]
- 237.Henderson NC, Arnold TD, Katamura Y, et al. Targeting of αv integrin identifies a core molecular pathway that regulates fibrosis in several organs. Nat Med 2013; 19(12): 1617–1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Levine D, Rockey DC, Milner TA, et al. Expression of the integrin alpha8beta1 during pulmonary and hepatic fibrosis. Am J Pathol 2000; 156(6): 1927–1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Wynn TA. Common and unique mechanisms regulate fibrosis in various fibroproliferative diseases. J Clin Investig 2007; 117(3): 524–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Wali MA, Eid RA and Al-Homrany MA. Smooth muscle changes in the cephalic vein of renal failure patients before use as an arteriovenous fistula (AVF). J Smooth Muscle Res 2002; 38(3): 75–85. [DOI] [PubMed] [Google Scholar]
- 241.Vazquez-Padron RI and Allon M. New insights into dialysis vascular access: impact of preexisting arterial and venous pathology on AVF and AVG outcomes. Clin J Am Soc Nephrol 2016; 11(8): 1495–1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Zhao J, Jourd’heuil FL, Xue M, et al. Dual function for mature vascular smooth muscle cells during arteriovenous fistula remodeling. J Am Heart Assoc 2017; 6(4): e004891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Zhao J, Wu W, Zhang W, et al. Selective expression of TSPAN2 in vascular smooth muscle is independently regulated by TGF-β1/SMAD and myocardin/serum response factor. FASEB J 2017; 31(6): 2576–2591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Mishra PK, Tyagi N, Sen U, et al. H2S ameliorates oxidative and proteolytic stresses and protects the heart against adverse remodeling in chronic heart failure. Am J Physiol Heart Circ Physiol 2010; 298(2): H451–H456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Allon M, Litovsky S, Young CJ, et al. Correlation of preexisting vascular pathology with arteriovenous graft outcomes in hemodialysis patients. Am J Kidney Dis 2013; 62(6): 1122–1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Duque JC, Tabbara M, Martinez L, et al. Similar degree of intimal hyperplasia in surgically detected stenotic and nonstenotic arteriovenous fistula segments: a preliminary report. Surgery 2018; 163(4): 866–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Hernandez DR, Applewhite B, Martinez L, et al. Inhibition of lysyl oxidase with β-aminopropionitrile improves venous adaptation after arteriovenous fistula creation. Kidney360 2021; 2(2): 270–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Cai C, Kilari S, Singh AK, et al. Differences in transforming growth Factor-β1/BMP7 signaling and venous fibrosis contribute to female Sex differences in arteriovenous fistulas. J Am Heart Assoc 2020; 9(16): e017420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Heine GH, Ulrich C, Sester U, et al. Transforming growth factor beta1 genotype polymorphisms determine AV fistula patency in hemodialysis patients. Kidney Int 2003; 64(3): 1101–1107. [DOI] [PubMed] [Google Scholar]
- 250.Stracke S, Konner K, Köstlin I, et al. Increased expression of TGF-beta1 and IGF-I in inflammatory stenotic lesions of hemodialysis fistulas. Kidney Int 2002; 61(3): 1011–1019. [DOI] [PubMed] [Google Scholar]
- 251.Gao F, Chambon P, Tellides G, et al. Disruption of TGF-β signaling in smooth muscle cell prevents flow-induced vascular remodeling. Biochem Biophys Res Commun 2014; 454(1): 245–250. [DOI] [PubMed] [Google Scholar]
- 252.Serralheiro P, Soares A, Costa Almeida CM, et al. TGF-β1 in vascular wall pathology: unraveling chronic venous insufficiency pathophysiology. Int J Mol Sci 2017; 18(12): 2534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Yun SH, Sim EH, Goh RY, et al. Platelet activation: the mechanisms and potential biomarkers. Biomed Res Int 2016; 2016:9060143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Furuya M, Tanaka R, Miyagi E, et al. Impaired CXCL4 expression in tumor-associated macrophages (TAMs) of ovarian cancers arising in endometriosis. Cancer Biol. Ther 2012; 13(8): 671–680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Maier M, Geiger EV, Henrich D, et al. Platelet factor 4 is highly upregulated in dendritic cells after severe trauma. Mol. Med 2009; 15(11–12): 384–391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Ravid K, Beeler DL, Rabin MS, et al. Selective targeting of gene products with the megakaryocyte platelet factor 4 promoter. Proc Natl Acad Sci USA 1991; 88(4): 1521–1525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Schaffner A, Rhyn P, Schoedon G, et al. Regulated expression of platelet factor 4 in human monocytes: role of PARs as a quantitatively important monocyte activation pathway. J. Leukoc. Biol 2005; 78(1): 202–209. [DOI] [PubMed] [Google Scholar]
- 258.Deuel TF, Senior RM, Chang D, et al. Platelet factor 4 is chemotactic for neutrophils and monocytes. Proc Natl Acad Sci USA 1981; 78(7): 4584–4587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Thomas MR and Storey RF. The role of platelets in inflammation. Thromb. Haemost 2015; 114(3): 449–458. [DOI] [PubMed] [Google Scholar]
- 260.Bebawy ST, Gorka J, Hyers TM, et al. In vitro effects of platelet factor 4 on normal human neutrophil functions. J Leukoc Biol 1986; 39(4): 423–434. [DOI] [PubMed] [Google Scholar]
- 261.Liu CY, Battaglia M, Lee SH, et al. Platelet factor 4 differentially modulates CD4+CD25+ (regulatory) versus CD4+CD25− (nonregulatory) T cells. J Immunol 2005; 174(5): 2680–2686. [DOI] [PubMed] [Google Scholar]
- 262.Scheuerer B, Ernst M, Dürrbaum-Landmann I, et al. The CXC-chemokine platelet factor 4 promotes monocyte survival and induces monocyte differentiation into macrophages. Blood 2000; 95(4): 1158–1166. [PubMed] [Google Scholar]
- 263.Martí F, Bertran E, Llucià M, et al. Platelet factor 4 induces human natural killer cells to synthesize and release interleukin-8. J Leukoc Biol 2002; 72(3): 590–597. [PubMed] [Google Scholar]
- 264.Struyf S, Salogni L, Burdick MD, et al. Angiostatic and chemotactic activities of the CXC chemokine CXCL4L1 (platelet factor-4 variant) are mediated by CXCR3. Blood 2011; 117(2): 480–488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Lasagni L, Francalanci M, Annunziato F, et al. An alternatively spliced variant of CXCR3 mediates the inhibition of endothelial cell growth induced by IP-10, mig, and I-TAC, and acts as functional receptor for platelet factor 4. J Exp Med 2003; 197(11): 1537–1549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Mueller A, Meiser A, McDonagh EM, et al. CXCL4induced migration of activated T lymphocytes is mediated by the chemokine receptor CXCR3. J Leukoc Biol 2008; 83(4): 875–882. [DOI] [PubMed] [Google Scholar]
- 267.Fox JM, Kausar F, Day A, et al. CXCL4/Platelet factor 4 is an agonist of CCR1 and drives human monocyte migration. Sci Rep 2018; 8(1): 9466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Sachais BS, Kuo A, Nassar T, et al. Platelet factor 4 binds to low-density lipoprotein receptors and disrupts the endocytic machinery, resulting in retention of low-density lipoprotein on the cell surface. Blood 2002; 99(10): 3613–3622. [DOI] [PubMed] [Google Scholar]
- 269.Shi G, Field DJ, Long X, et al. Platelet factor 4 mediates vascular smooth muscle cell injury responses. Blood 2013; 121(21): 4417–4427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Zilberberg A, Yaniv A and Gazit A. The low density lipoprotein receptor-1, LRP1, interacts with the human frizzled-1 (HFz1) and down-regulates the canonical Wnt signaling pathway. J Biol Chem 2004; 279(17): 17535–17542. [DOI] [PubMed] [Google Scholar]
- 271.Attisano L and Wrana JL. Signal transduction by the TGF-beta superfamily. Science 2002; 296(5573): 1646–1647. [DOI] [PubMed] [Google Scholar]
- 272.Guo X and Wang XF. Signaling cross-talk between TGFbeta/BMP and other pathways. Cell Res 2009; 19(1): 71–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Aidoudi S, Bujakowska K, Kieffer N, et al. The CXC-chemokine CXCL4 interacts with integrins implicated in angiogenesis. PLoS One 2008; 3(7): e2657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Lishko VK, Yakubenko VP, Ugarova TP, et al. Leukocyte integrin Mac-1 (CD11b/CD18, αMβ2, CR3) acts as a functional receptor for platelet factor 4. J Biol Chem 2018; 293(18): 6869–6882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Whitson RH, Wong WL and Itakura K. Platelet factor 4 selectively inhibits binding of TGF-β1 to the type I TGF-β1 receptor. J Cell Biochem 1991; 47(1): 31–42. [DOI] [PubMed] [Google Scholar]
- 276.Nakerakanti S and Trojanowska M. The role of TGF-β receptors in fibrosis. Open Rheumatol J 2012; 6:156–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Baker AB, Ettenson DS, Jonas M, et al. Endothelial cells provide feedback control for vascular remodeling through a mechanosensitive autocrine TGF-beta signaling pathway. Circ. Res 2008; 103(3): 289–297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Egorova AD, Van der Heiden K, Van de Pas S, et al. Tgfβ/Alk5 signaling is required for shear stress induced klf2 expression in embryonic endothelial cells. Dev Dyn 2011; 240(7): 1670–1680. [DOI] [PubMed] [Google Scholar]
- 279.González-Núñez M, Muñoz-Félix JM and López-Novoa JM. The ALK-1/Smad1 pathway in cardiovascular physiopathology: a new target for therapy? Biochim Biophys Acta 2013; 1832(10): 1492–1510. [DOI] [PubMed] [Google Scholar]
- 280.Goumans MJ, Valdimarsdottir G, Itoh S, et al. Balancing the activation state of the endothelium via two distinct TGF-beta type I receptors. EMBO J 2002; 21(7): 1743–1753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Tang Y, Yang X, Friesel RE, et al. Mechanisms of TGF-β-induced differentiation in human vascular smooth muscle cells. J Vasc Res 2011; 48(6): 485–494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Walshe TE, dela Paz NG and D’Amore PA. The role of shear-induced transforming growth factor-β signaling in the endothelium. Arterioscler Thromb Vasc Biol 2013; 33(11): 2608–2617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Bhatt DL, Stone GW, Mahaffey KW, et al. Effect of platelet inhibition with cangrelor during PCI on ischemic events. New Engl J Med 2013; 368(14): 1303–1313. [DOI] [PubMed] [Google Scholar]
- 284.Murata S, Maruyama T, Nowatari T, et al. Signal transduction of platelet-induced liver regeneration and decrease of liver fibrosis. Int J Mol Sci 2014; 15(4): 5412–5425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Rahman M, Gustafsson D, Wang Y, et al. Ticagrelor reduces neutrophil recruitment and lung damage in abdominal sepsis. Platelets 2014; 25(4): 257–263. [DOI] [PubMed] [Google Scholar]
- 286.Zhan T, Wei T, Dong L, et al. Cangrelor alleviates bleomycin-induced pulmonary fibrosis by inhibiting platelet activation in mice. Mol Immunol 2020; 120:83–92. [DOI] [PubMed] [Google Scholar]
- 287.Silva-Cardoso SC, Tao W, Angiolilli C, et al. CXCL4 links inflammation and fibrosis by reprogramming monocyte-derived dendritic cells in vitro. Front Immunol 2020; 11:2149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Gleitz HFE, Dugourd AJF, Leimkühler NB, et al. Increased CXCL4 expression in hematopoietic cells links inflammation and progression of bone marrow fibrosis in MPN. Blood 2020; 136(18): 2051–2064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Gabrielli A, Avvedimento EV and Krieg T. Scleroderma. New Engl J Med 2009; 360(19): 1989–2003. [DOI] [PubMed] [Google Scholar]
- 290.Lande R, Mennella A, Palazzo R, et al. Anti-CXCL4 antibody reactivity is present in systemic sclerosis (SSc) and correlates with the SSc type I interferon signature. Int J Mol Sci 2020; 21(14): 5102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.Ah Kioon MD, Tripodo C, Fernandez D, et al. Plasmacytoid dendritic cells promote systemic sclerosis with a key role for TLR8. Sci Transl Med 2018; 10(423): eaam8458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Lande R, Lee EY, Palazzo R, et al. CXCL4 assembles DNA into liquid crystalline complexes to amplify TLR9-mediated interferon-α production in systemic sclerosis. Nat Commun 2019; 10(1): 1731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.van Bon L, Affandi AJ, Broen J, et al. Proteome-wide analysis and CXCL4 as a biomarker in systemic sclerosis. New Engl J Med 2014; 370(5): 433–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Volkmann ER, Tashkin DP, Roth MD, et al. Changes in plasma CXCL4 levels are associated with improvements in lung function in patients receiving immunosuppressive therapy for systemic sclerosis-related interstitial lung disease. Arthritis Res Ther 2016; 18(1): 305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Zhang L, Zhu Y, Zhang D, et al. Platelet factor 4 protects kidney allograft in a rat kidney transplantation model. Inflammation 2015; 38(2): 520–526. [DOI] [PubMed] [Google Scholar]
- 296.Tabbara M, Duque JC, Martinez L, et al. Pre-existing and postoperative intimal hyperplasia and arteriovenous fistula outcomes. Am J Kidney Dis 2016; 68(3): 455–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Wang Y, Krishnamoorthy M, Banerjee R, et al. Venous stenosis in a pig arteriovenous fistula model: anatomy, mechanisms and cellular phenotypes. Nephrol Dial Transplant 2007; 23(2): 525–533. [DOI] [PubMed] [Google Scholar]
- 298.Li L, Terry CM, Blumenthal DK, et al. Cellular and morphological changes during neointimal hyperplasia development in a porcine arteriovenous graft model. Nephrol Dial Transplant 2007; 22(11): 3139–3146. [DOI] [PubMed] [Google Scholar]
- 299.Misra S, Doherty MG, Woodrum D, et al. Adventitial remodeling with increased matrix metalloproteinase-2 activity in a porcine arteriovenous polytetrafluoroethylene grafts. Kidney Int 2005; 68(6): 2890–2900. [DOI] [PubMed] [Google Scholar]
- 300.Zabini D, Nagaraj C, Stacher E, et al. Angiostatic factors in the pulmonary endarterectomy material from chronic thromboembolic pulmonary hypertension patients cause endothelial dysfunction. PLoS One 2012; 7(8): e43793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Border WA, Okuda S, Languino LR, et al. Transforming growth factor-beta regulates production of proteoglycans by mesangial cells. Kidney Int 1990; 37(2): 689–695. [DOI] [PubMed] [Google Scholar]
- 302.Haberstroh U, Zahner G, Disser M, et al. TGF-beta stimulates rat mesangial cell proliferation in culture: role of PDGF beta-receptor expression. Am J Physiol 1993; 264(2 Pt 2): F199–F205. [DOI] [PubMed] [Google Scholar]
