Review Open Access
Copyright ©The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jan 21, 2017; 23(3): 382-405
Published online Jan 21, 2017. doi: 10.3748/wjg.v23.i3.382
Pancreatic stellate cell: Pandora's box for pancreatic disease biology
Ratnakar R Bynigeri, Aparna Jakkampudi, Ramaiah Jangala, Chivukula Subramanyam, Mitnala Sasikala, G Venkat Rao, D Nageshwar Reddy, Rupjyoti Talukdar, Institute of Basic Sciences, Asian Healthcare Foundation, Hyderabad 500082, India
D Nageshwar Reddy, Rupjyoti Talukdar, Department of Medical Gastroenterology, Asian Institute of Gastroenterology, Hyderabad 500082, India
G Venkat Rao, Department of Surgical Gastroenterology, Asian Institute of Gastroenterology, Hyderabad 500082, India
Author contributions: Bynigeri RR, Jakkampudi A and Jangala R conducted literature search, compiled references and drafted the paper; Subramanyam C, Sasikala M, Rao GV and Reddy DN contributed to manuscript drafting and provided intellectual input; Talukdar R conceived and designed the manuscript, verified content, contributed to manuscript drafting and approved the final manuscript.
Conflict-of-interest statement: The authors declare no conflict of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Rupjyoti Talukdar, MD, Clinical Pancreatologist, Asian Institute of Gastroenterology, Clinician Scientist (Wellcome-DBT Fellow), Asian Healthcare Foundation, 6-3-661 Somajiguda, Hyderabad 500082, Telangana, India. rup_talukdar@yahoo.com
Telephone: +91-40-23378888 Fax: +91-40-23324255
Received: August 30, 2016
Peer-review started: August 31, 2016
First decision: October 10, 2016
Revised: November 9, 2016
Accepted: December 16, 2016
Article in press: December 19, 2016
Published online: January 21, 2017
Processing time: 137 Days and 8.2 Hours

Abstract

Pancreatic stellate cells (PSCs) were identified in the early 1980s, but received much attention after 1998 when the methods to isolate and culture them from murine and human sources were developed. PSCs contribute to a small proportion of all pancreatic cells under physiological condition, but are essential for maintaining the normal pancreatic architecture. Quiescent PSCs are characterized by the presence of vitamin A laden lipid droplets. Upon PSC activation, these perinuclear lipid droplets disappear from the cytosol, attain a myofibroblast like phenotype and expresses the activation marker, alpha smooth muscle actin. PSCs maintain their activated phenotype via an autocrine loop involving different cytokines and contribute to progressive fibrosis in chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC). Several pathways (e.g., JAK-STAT, Smad, Wnt signaling, Hedgehog etc.), transcription factors and miRNAs have been implicated in the inflammatory and profibrogenic function of PSCs. The role of PSCs goes much beyond fibrosis/desmoplasia in PDAC. It is now shown that PSCs are involved in significant crosstalk between the pancreatic cancer cells and the cancer stroma. These interactions result in tumour progression, metastasis, tumour hypoxia, immune evasion and drug resistance. This is the rationale for therapeutic preclinical and clinical trials that have targeted PSCs and the cancer stroma.

Key Words: Pancreatic stellate cells; Pancreatic fibrosis; Pancreatic cancer stroma; Physiological functions; Pancreatic stellate cells-cancer-stromal interactions; Therapeutic targets

Core tip: Pancreatic stellate cells (PSCs) have emerged as one of the major effector cells in chronic pancreatitis and pancreatic ductal adenocarcinoma. In this review, we discuss the physiological function of PSCs and the profibrogenic mechanisms. We also discuss various pathways, transcription factors and miRNAs implicated in the inflammatory and profibrogenic functions mediated by PSCs. We further discuss the crosstalk among PSCs, pancreatic cancer cells and pancreatic cancer stroma and mechanisms that lead to cancer progression, metastasis, tumour hypoxia, immune evasion and drug resistance. We conclude with recent preclinical and clinical studies that have targeted PSCs and cancer stroma.



HISTORICAL PERSPECTIVES

Stellate cells were described for the first time in the perisinusoidal spaces of the liver by Karl Wilhelm Von Kupffer in 1876 and were called "Sternzellen" (meaning star shaped cells). Later in 1951, Ito described the presence of lipid droplet containing cells in the perisinusoidal spaces of the liver and named them "Ito cells"[1]. The Ito cells were shown to emit blue-green fluorescence due to the presence of vitamin A in the lipid droplets[2]. Later in 1971, the usage of multiple techniques provided unequivocal evidence that the "sternzellen" reported by Kupffer and "Ito cells" identified by Ito were the same cell type: the hepatic stellate cells (HSCs)[3,4]. In 1982, a cell type carrying vitamin A containing lipid droplets and exhibiting a transient blue-green fluorescence were described in mouse pancreas[5]. In 1991, the cells exhibiting the vitamin A autofluorescence were identified in the healthy pancreatic sections from humans and rats[6]. These cells are now identified as pancreatic stellate cells (PSCs), which localize the periacinar regions, with long cytoplasmic projections extending towards the basolateral aspects of the acinar cells. Later in 1998, the development of in vitro tools to isolate and culture the PSCs laid a strong foundation to characterize their basic biology[7,8]. These cells also surround the perivascular and periductal regions. Sustained PSC cultures have helped to decipher the crucial factors that act in the inflammatory mechanisms and their mechanistic role in the pancreatic fibrosis in chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC). However, in view of the challenges of limited viability of the PSCs in primary cultures, there had been several attempts to modify isolation and culture techniques. In this regard, techniques were developed to immortalize the normal and tumour associated PSCs. However, further validation studies will be required prior to their routine use in PSC research[9-12]. Interestingly, even though PSCs were associated primarily with the exocrine pancreas, a recent study has reported isolation of PSCs from rat and human pancreatic islets too. These cells demonstrated certain morphologic and functional differences from the conventional PSCs in terms of fewer lipid droplets, lower rates of proliferation, migration and easier activation[13,14].

BASIC BIOLOGY OF PANCREATIC STELLATE CELLS
Origin

The origin of PSCs is still being debated. Till date no direct studies have been executed to identify the origin of PSCs. However, the studies on the origin of HSCs have helped in gaining some insight into this aspect. Even though initially a neuroectodermal origin of PSCs was proposed, it was eventually negated in genetic cell lineage mapping studies[15]. A recent study forwarded refreshing evidence supporting a mesodermal origin of HSCs by using the conditional lineage analysis approach[16,17]. Since most of the characteristic features and functions that sketched the biology of PSCs are similar to HSCs, it is believed that even PSCs might have evolved from a mesodermal origin. Employing such similar tracer techniques might help in ascertaining the origin of PSCs.

In the context of CP and PDAC, even though most of the proliferating PSCs are derived from the resident PSCs within the pancreas, a proportion of PSCs are thought to originate in the bone marrow. This was proposed in a novel sex mismatched study, which evidenced that even bone marrow (BM) derived cells may also contribute to PSC population in CP and PDAC apart from the resident cells of pancreas[18,19]. The speculation that bone marrow is another potential source of PSC was further supported by a recent study involving dibutylin chloride induced CP wherein a model of stable hematopoietic chimerism by grafting enhanced green fluorescence protein (eGFP)-expressing BM cells was used. In this study, 18% of the PSCs in the pancreas was found to originate in the bone marrow[20]. A recent study that used enhanced green fluorescent protein (EGFP)(+)CD45(-) cells transplanted from EGFP-transgenic mice in a carbon tetrachloride (CCL4) model suggested that infiltrating monocytes could also differentiate into stellate cells within the pancreas and liver under the influence of monocyte chemoattractant protein-1 (MCP-1)[21].

Morphologic characteristics

Most of the characteristic features exhibited by quiescent as well as activated PSCs have been determined based on in vitro studies using rat and human PSC isolates. Cultured PSCs display prominent vitamin A containing lipid droplets with perinuclear localization in the cytoplasm. These lipid droplets elicit a fugacious blue-green autofluorescence when exposed to UV light at 328 nm or 350 nm wavelength. The expression of glial fibrillary acidic protein (GFAP) is specific to PSCs in the pancreas and presence of lipid droplets in the cytoplasm define the quiescent phenotype of PSCs[5-8]. The underlying mechanisms involved in the accumulation and disappearance of lipid droplets are still not elaborately elucidated. It was demonstrated in a few studies that albumin colocalizes with the lipid droplets within quiescent PSCs. Activated PSCs, which are characterized by disappearance of lipid droplets, re-developed the lipid droplets and showed resistance against the activating effects of transforming growth factor-β (TGF-β) when transfected with the plasmids expressing albumin, thereby confirming the contribution of albumin in lipid droplet formation. The albumin was reported to be a downstream effector of peroxisome proliferator activated receptor-γ (PPAR-γ), a nuclear receptor that is known to inhibit PSC activation[22,23]. The presence of lipid droplets together with expression of GFAP, desmin, nestin and vimentin is used to differentiate the PSCs from pancreatic fibroblasts[24]. Using GFAP-LacZ transgenic mice model, it was proven that GFAP promoter activity was unique to PSCs alone in the pancreas[25].

Autotransformation of quiescent PSCs to activated phenotype is observed in vitro. The basic phenotypic differences that were observed when the PSCs switch to activated phenotype include the disappearance of lipid droplets and transformation into a myofibroblast-like phenotype. The expression of α-smooth muscle actin (α-SMA) marks the transdifferentiation of the quiescent PSC to an activated phenotype. Figure 1 shows the morphology of PSCs in culture at different time points.

Figure 1
Figure 1 Morphological changes observed in cultured rat pancreatic stellate cells at different time points after isolation. A: Quiescent pancreatic stellate cells (PSCs) in culture exhibiting a flattened shape with lipid droplets, 6 h after isolation (× 20); B, C: PSCs showing flattened angular appearance and exhibiting cytoplasmic extensions with lipid droplets after 24 and 48 h respectively in cultures (× 20); D: PSCs exhibiting dense lipid droplets (lipid droplets are indicated with black arrows) in the cytoplasm (× 40); E: Activated PSCs showing long cytoplasmic processes with no lipid droplets in the cytoplasm after 72 h in cultures (× 20); F: Passage 2 rat PSCs in culture, immunostained for α-smooth muscle actin (α-SMA), a cytoskeletal marker for activated PSCs. Green striations indicate α-SMA and blue spots indicate nuclei, stained with DAPI (× 20).
PSC functions

The physiological and pathological functions of PSCs have been summarized in Table 1. Under physiological conditions, PSCs are believed to contribute to the exocrine cell structure and function via maintenance of the normal basement membrane[26,27] and carry out normal ductal and vascular regulation by virtue of their localization[28]. Quiescent PSCs have a low mitotic index and bears the capability to synthesize matrix proteins and maintain the physiological extracellular matrix. The expression of matrix metalloproteinases (MMPs) and tissue inhibitors of matrix metalloproteinases (TIMPs) are complementary to each other and is a prerequisite to poise the ECM turnover. Increased production of the ECM proteins fibronectin, periostin, MMPs and TIMPs are the most common features exhibited by the activated PSC phenotype[29] and hence described as the effector cells contributing to the stroma associated with CP and PDAC. Besides laying and maintaining the ECM, PSCs have also been demonstrated to secrete acetylcholine that might function as an intermediate regulator for cholecystokinin mediated pancreatic exocrine secretion[30].

Table 1 Function of pancreatic stellate cells in the quiescent state and after activation.
Physiological functions
Store fat and retinoids in their perinuclear droplets, expressing GFAP, desmin and vimentin
Secrete MMPs and TIMPs
Maintains ECM turnover
Involved in maintenance of pancreatic tissue architecture
No or limited secretion of cytokines, chemokines and growth factors
Function as an immune, progenitor and intermediary cell
Possible role in exocrine and endocrine secretions
Pathological functions
Exhibit cell proliferation and migration
Deranged ECM turnover due to loss of balance between MMPs and TIMPs
Secrete various cytokines, chemokines and growth factors and thereby contribute to inflammatory milieu
Stimulate cancer cell proliferation and migration and inhibit their apoptosis
Mediate invasion and metastasis of carcinoma cells
Mediate chemoresistance and radioresistance thereby promoting cancer cell survival
Contribute to the hypovascular and hypoxic tumour microenvironment
Promote angiogenesis, neural invasion and epithelial-mesenchymal transition

Until recently, much attention was paid to unveil the functions of activated PSCs as a multiple cytokine producing profibrogenic cell type, which promotes self-proliferation, migration and fibrogenesis. However, recent advances have even demonstrated certain non-fibrogenic functions of PSCs, which projected PSCs as immune cells[31], intermediary cell[30,32] and also as a progenitor cell[33-35]. An earlier study showed that PSCs could phagocytize senescent neutrophils in experimental acute pancreatitis (AP) and this was reduced by the presence of cytokines while augmented by presence of PPAR-γ ligand[31]. The same group subsequently demonstrated that PSCs could also phagocytize necrotic acinar cells and themselves undergo cell death. No change in TGF-β concentration was detected in the PSC media and medium with PSC and acinar cells, thereby indicating that the death of PSCs could result in inhibition of fibrogenesis in the setting of AP[36]. This role in innate immunity was further supported by the capacity of PSCs to recognize pathogen-associated molecular patterns via Toll-like receptors (TLRs) that are expressed on their surface[37].

Studies have now also proposed a regenerative role especially in the context of AP, where the interaction between extracellular matrix laid by PSCs and acinar surface integrin receptors could result in a scaffold for acinar regeneration. Excess matrix deposition could also potentially be ameliorated by matrix degrading enzymes and apoptosis/cytolysis of activated PSCs[38].

In addition to the above-mentioned functions of PSCs, it is now becoming more evident that these multifunctional cells also affects endocrine secretion in CP. This speculation surfaced from experiments that demonstrated increased numbers of PSCs in rat pancreas in a Type 2 diabetic model[39]. Extension of this study in vitro showed that PSCs could reduce insulin secretion and induce β-cell apoptosis[40-42]. On the contrary, another study showed that PSCs increase insulin secretion from mouse islets[43]. Interestingly, INS-1 cell culture supernatants reduced the secretion of proinflammatory cytokines (that mediate β-cell dysfunction) and ECM proteins from PSCs[44]. Moreover, the expression of regenerating islet-derived protein-1 was high in islet stellate cells (ISCs) isolated from the diabetic mice, which inhibited the viability, migration, synthesis and secretion of ECM proteins in ISCs in vitro[45]. As the in vitro results are more divergent, meticulous studies need to be designed and executed to understand the precise role played by these cells during their reciprocal interaction.

Fate of PSCs

The fate of activated PSCs is an important question that remains unresolved. Figure 2 depicts a schematic representation of the fate of PSCs. One of the two possible explanations that were proposed is that sustained inflammation may perpetuate PSC activation, leading to fibrosis; while the other explanation proposed that the activated PSCs may undergo apoptosis or may revert back to the native phenotype if the inflammation or injury is ceased. Recently, Fitzner et al[46] proposed that activated PSCs could undergo senescence as evidenced by increased senescence-associated β-galactosidase, higher expression of CDKN1A/p21, mdm2 and interleukin (IL)-6. On the contrary, there was lower expression of α-smooth muscle actin. The authors also observed that senescence increased the susceptibility of PSCs to cytolysis and concluded that inflammation, PSC activation and cellular senescence were coupled processes that took place in the same inflammatory microenvironment of CP[46]. In the setting of AP, PSCs could undergo death after phagocytizing necrotic acinar cells[36].

Figure 2
Figure 2 Fate of activated pancreatic stellate cells. The fate of activated pancreatic stellate cells (PSCs) could be potentially two pronged-sustained inflammation and/or autocrine mode of PSC activation may perpetuate its activated phenotype, even in the absence of its paracrine triggers, resulting in the development of pancreatic fibrosis or PSCs might undergo either reversion to quiescent phenotype or apoptosis or may become senescent and further cleared by lymphocytes. In the latter situation, there should not be fibrosis.
PSCS AND FIBROSIS

A pathological hallmark of CP and PDAC is progressive fibrosis that is mediated by the PSCs. One of the earliest cellular events at the initiation of fibrosis is activation of PSCs, which can be mediated concomitantly by a variety of factors, such as oxidative stress, cytokines, growth factors, activin-A, angiotensin, hyperglycemia and pressure, to name a few. Interestingly, activation of PSCs can occur by both autocrine and paracrine mechanisms, which imply that the effects of PSC activation, primarily inflammation and resultant fibrosis can progress, even after removing the primary source. The distinctive sources of exogenous factors that actuate the PSC include activated macrophages, monocytes, pancreatic acinar cells, endothelial cells, pancreatic cancer cells and platelets in inflamed pancreas[47-50]. Figure 3 depicts the autocrine and paracrine mechanisms of PSC activation and the resulting fibrosis.

Figure 3
Figure 3 Autocrine and paracrine factors mediating pancreatic stellate cell activation. Cytokines and growth factors secreted by injured acinar cells, immune cells and cancer cells activate the pancreatic stellate cells (PSCs) in a paracrine fashion and stimulate them to secrete various factors. These factors secreted by PSCs in turn acts in a paracrine fashion and sustains its activation. This autocrine and paracrine signal cycles may help PSCs to retain its activated phenotype, resulting in excess ECM deposition, culminating to pancreatic fibrosis. ROS: Reactive oxygen species; PC: Pancreatic cancer; FGF: Fibroblast growth factor; PDGF: Platelet derived growth factor; TGF-β: Transforming growth factor-β; VEGF: Vascular endothelial growth factor; TNF-α: tumor necrosis factor-α; MMP: Matrix metalloproteinase; CP: Chronic Pancreatitis; TIMPs: Tissue inhibitors of matrix metalloproteinases; CTGF: Connective tissue growth factor; IL: Interleukin.
Alcohol, smoking and PSC activation

Alcohol and smoking are now recognized as independent risk factors for the development of CP. It is known that pancreatic acinar cells can metabolize alcohol to form toxic metabolites that results in oxidative stress. This results in inflammation and PSC activation[51-53]. Furthermore, PSCs themselves can metabolize ethanol to acetaldehyde and generate oxidative stress, thus promoting their own activation and lipid peroxidation. The above findings have been confirmed by immunostaining for 4-hydroxy-nonenal (4-HNE), a reactive product of lipid peroxidation, that demonstrated localization of 4-HNE stained PSCs in fibrotic areas adjacent to acinar cells[54-56]. Ethanol activated PSCs showed increased proliferation by enhancing the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase system stimulated by platelet derived growth factor (PDGF)[57]. Also shown was expression of connective tissue growth factor (CCN2/CTGF) that was attributed to production of acetaldehyde and oxidant stress in ethanol stimulated PSCs, which rendered the properties of cell adhesion, migration and collagen synthesis when stimulated with profibrogenic molecules[58]. Recently, CCN2 was also shown to increase miR-21 expression that in turn enhanced collagen α1 expression in a murine alcoholic CP model. CCN2 and miR-21 were shown to be colocalized in PSC derived exosomes that were positive for cluster of differentiation (CD) 9. In vitro studies revealed that these exosomes serve as molecular cargos to activate and transfer fibrogenic signals to the adjacent PSCs[59].

Lee et al[60] has recently demonstrated that PSCs express nicotinic acetylcholine nAChRs (isoforms α3, α7, β, ε). Furthermore, nicotine and nicotine-derived nitrosamine ketone and cigarette smoke extracts were shown to activate PSCs both in the presence and absence of alcohol. This reiterates the clinical observation of role of smoking as an independent risk factor in the initiation and progression of CP[61].

Pressure and PSC activation

Ductal hypertension resulting from obstructing pancreatic ductal calculi or stricture has been long believed to be a major contributor of pain in CP. This formed the rationale for ductal clearance of stone/stricture by endotherapy and/or surgery in an attempt to ameliorate pain in CP. Experimental evidence to support this concept came forward from studies by Asaumi et al[62] where externally applied pressure of 80 mmHg induced activation of PSCs and generation of reactive oxygen species (ROS) within the activated PSCs[63]. ROS generation was observed as early as 30 min after application of pressure and reached peak by 1 h.

Hyperglycemia and PSC activation

In a study by Ko et al[64], exposure of PSCs to high glucose concentration resulted in stimulation of α-SMA expression, proliferation and expression of extracellular matrix proteins such as CTGF and collagen type IV. PSC activation by hyperglycemia was also confirmed by subsequent studies by Nomiyama et al[65] and Hong et al[39] and the latter study also suggested an additive effect of hyperglycemia and hyperinsulinemia in inducing PSC activation and islet fibrosis in the context of Type 2 diabetes. Observations from these studies have provided an insight into the role of hyperglycemia in preserving the activated phenotype and also in the context of secondary diabetes in patients with CP. A more recent study has indicated that hyperglycemia could result in induction of Cysteine-X-Cysteine ligand (CXCL) 12 production by the PSCs and its receptor, CXCR4 on cancer cells[66].

Cytokines and other activation factors that mediate proinflammatory function of PSCs

Fibrous tissue in CP and PDAC abounds in type I collagen. Among the cytokines that can cause PSC activation, TGF-β stands among the most important. Studies have shown increased collagen synthesis and upregulation of MMP1 in PSCs that were stimulated with TGF-β and TGF-α[67,68]. Other activators of PSCs include interleukin-8 (IL-8), MCP-1 and RANTES (Regulated on Activation, Normal T-cell Expressed and Secreted), which promote PSC activation via autocrine pathways[69]. Activin-A and angiotensin II have also been identified as the autocrine activators of PSCs, contributing to further TGF-β1 expression and PSC proliferation[70,71]. Expression of TGF-β1 and collagen secretion, has also been shown to result from application of external pressure and with hyperglycemia[63-65].

Migration and proliferation of PSCs are other important properties that go parallel along with the proinflammatory and profibrotic cascade. Proliferation and migration of PSCs is mediated by PDGF[57,72,73] (which is expressed after TGF-β mediated activation) and endothelin-1[74,75]. A proinflammatory chemokine, CX3CL1 (fractalkine), reported to circulate in the serum of patients with alcoholic chronic pancreatitis, was demonstrated as an activation and proliferation factor for PSCs and PSCs were shown to express the receptor (CX3CR1) for this chemokine[76,77]. Interestingly, this chemokine and its receptor system was reported to regulate the insulin secretion by β-cells[78]. Recently, another new activation factor, namely parathyroid hormone related protein (PTHrP) was demonstrated to be expressed by acinar cells during experimental pancreatitis using an acinar cell specific PTHrP gene knock-out model. Receptor for this factor (PHT1R) has been shown to be expressed in PSCs and receptor-ligand interaction between the two resulted in fibrogenesis[79]. Of note, IL-6 has been shown to inhibit both PSCs proliferation and collagen synthesis[80]. Recently it was also demonstrated that IL-4 and IL-13 secreted by PSCs mediate macrophage activation, which in turn participate in promoting the pancreatic fibrosis[81].

To summarize the effect of the above experimental evidence, different paracrine factors released during the injury will result in activation, proliferation and migration of PSCs and the activated phenotype is further retained by an autocrine loop, even in the absence of paracrine triggers.

MOLECULAR PATHWAYS, MICRORNAS, TRANSCRIPTION FACTORS AND PROTEOMICS IN PSC MEDIATED PANCREATIC FIBROSIS

Studies conducted over the past decade have implicated the involvement of several proteins and molecular pathways (Figure 4) in perpetuating the profibrogenic role of PSCs.

Figure 4
Figure 4 Signaling pathways mediating pancreatic stellate cell activation. Expression of α-SMA, proliferation, migration and deposition of matrix proteins are the important properties attained by activated pancreatic stellate cells (PSCs) when stimulated with various growth factors and proinflammatory cytokines. Proliferation and migration is mediated through the MAP kinase and PI3K pathways when PSCs are stimulated with HNE, alcohol, PDGF and IL-33 and other cytokines. TGF-β1 induces the Smad proteins and stimulates the proliferation and collagen secretion by PSCs. Activation of Indian Hedgehog (IHH) signaling in PSCs promotes their migration, proliferation and collagen deposition. PSC mediated Sonic Hedgehog (SHH) signaling promotes cancer cell invasion and migration. Wnt signaling can cause collagen deposition and cancer progression. PDGF: Platelet derived growth factor; HNE: Hydroxy-nonenal; ERK: Extracellular signal-regulated kinases; JNK: c-Jun N-terminal kinase; TGF-β: Transforming growth factor-β; α-SMA: α-smooth muscle actin; COX-2: Cyclooxygenase-2; IL: Interleukin.
MAPK, JAK-STAT and PI3K signaling pathways

Mitogen activated protein kinases (MAPKs) are serine/threonine protein kinases with three families: extracellular signal regulated kinase (ERKs), c-Jun N-terminal kinase (JNK) and p38[82], and all the three MAPKs have been studied extensively for their role in PSCs activation. In vitro studies demonstrated that the activation of ERK1/2 is the initial pathway that precedes the transformation of PSCs into activated phenotype and PDGF was shown to mediate ERK1/2 and Activator protein-1 (AP-1) dependent proliferation and migration of PSCs[72,73]. Studies have also demonstrated involvement of the Janus-activated kinase-signal transducers and activators of transcription (JAK2-STAT3) pathway in PDGF-BB induced PSC proliferation[83]. PI3K and all the MAPKs were described in human PSCs to express IL-32α and IL-33 when treated with proinflammatory cytokines. IL-33 was shown to activate PSCs[84-86]. HNE was reported to activate all the 3 classes of MAPKs and AP-1. PSCs treated with HNE showed increased production of type I collagen with no significant effect on proliferation and transformation, implicating oxidative stress mediated pathogenesis of pancreatic inflammation and fibrosis[87]. All the three MAPKs including AP-1 were triggered in PSCs when stimulated with ethanol and acetaldehyde. Inhibition of p38, JNK and Rho associated protein kinase (ROCK) pathways demonstrated the inhibition of PSC activation, supporting the involvement of above mentioned pathways in the pathogenesis of alcohol induced pancreatic injury[54,88,89].

Smad signaling pathway

TGF-β1, which is a proven profibrogenic cytokine, is required in the regulation of PSC activation[90]. Smads are the signaling effectors of TGF-β mediated functions and have also been ascribed a regulatory role in PSC functions. Results from co-expression of Smad2/3 with dominant negative Smad2/3 mutants and inhibition of ERK showed that the activation, proliferation and TGF-β1 mRNA expression are mediated through the Smad2/3 and ERK dependent pathways in PSCs[91]. The autocrine loop between IL-1β and TGF-β1 and the one existing between the IL-6 and TGF-β1 were mediated by Smad3/ERK dependent and Smad2/3 and ERK dependent pathways. Further investigations had confirmed the existence of a TGF-β1 autocrine loop and supported the role of PSCs in preserving the activated phenotype and collagen synthesis[92,93]. TGF-β1 induced expression of cyclooxygenase-2 (COX-2) by PSCs also followed Smad2/3 dependent pathway in response to proinflammatory cytokines[94]. This pathway has been suggested to be protective against the inhibitory activity of reversion-inducing-cysteine-rich protein with kazal motifs (RECK), a membrane anchored MMP inhibitor in the activated PSCs[95]. The stimulation of activated PSCs with TGF-β unveiled the possible role of Ras-ERK and PI3/Akt pathways in the expression of MMP-1[68].

Wnt signaling and β-catenin pathway

Yet another signaling pathway aberration that could result in PSC activation, proliferation and transformation into a profibrotic phenotype is that of Wnt signaling. This observation came from an experimental CP model by Hu et al[96] where the authors have shown that there was increased expression of Wnt and its second messenger β-catenin and that this imbalance could result in persistent activation of PSCs. Yet another study by Xu et al[97] showed that cancer cell invasion and migration are promoted by Wnt2 protein secreted by the PSCs.

Hedgehog signaling pathway

Indian hedgehog (IHH) and sonic hedgehog (SHH) are the other important pathways in PSCs. Receptors, namely smoothened and patched-1, for the IHH protein are expressed on the surface of PSCs and the receptor-ligand binding results in localization of the membrane-type 1 matrix metalloproteinase on PSC plasma membrane, which in turn could mediate PSC migration[98]. SHH was shown to influence the PSC mobility and differentiation[99] and also perineural invasion, metastasis, tumour growth and pain in pancreatic cancer[100,101].

microRNAs

Implications on the involvement of microRNAs (miRs) has recently being reported frequently in the context of CP and PSCs. A recent study reported upregulation and downregulation of 42 miRs each in activated PSCs[102]. miR-15b and 16 have been shown to induce apoptosis of rat PSCs via influencing the anti-apoptotic Bcl-2 protein[103]. An even more recent study demonstrated a paracrine pathway wherein CCN2 mRNA and miR-21 containing exosomes liberated by PSCs were engulfed by surrounding PSCs. This results in further expression of the CCN2 and miR-21 by the activated PSCs[59].

Transcription factors and interactions with cytokines

Different cytokines exert their effect by inducing various transcription factors such as nuclear factor-κB (NF-κB), Activator protein-1 (AP-1), STAT proteins and Gli, to name a few. NF-κB is stimulated by various cytokines associated with different cellular functions[104]. Activated PSCs showed NF-κB mediated expression of intracellular adhesion molecule when stimulated with IL-1β and tumor necrosis factor (TNF)-α, which was not observed in the quiescent phenotype[105]. Expression of MCP-1, cytokine induced neutrophil chemoattractant-1, IL-6, IL-8 and RANTES was observed via NF-κB activation when induced with galectin-1, various ligands of TLR and cytokines, substantiating the role of PSCs in mediating the infiltration and accumulation of inflammatory cells[106-108].

Proteomics

Proteomic studies using the immortalized PSC lines from Mus musculus and Rattus norvegicus showed the expression of cytoskeletal and ribosomal proteins by activated PSCs. The studies also demonstrated proteins involved in protein degradation, MAPK 3 and Ras related proteins by pseudo-quiescent PSCs[109,110]. Proteomic profiling of mild and severe CP by label free quantitative proteomic approach displayed varied expression of proteins with a relative change in the proteins related to ECM and PSC activation which includes periostin, fibrillin 1, transgelin and a group of collagens. An accompanying study showed increased expression of transgelin in stromal and periacinar regions of CP, confirming its role in PSC activation[111,112].

A comparative proteomic profiling of human HSC and PSC lines LX-2 and RLT-PSCs identified 1223 different proteins. Among 1223 proteins 1222 were found to be commonly expressed in both cell lines and a single protein (amino transferase) was found expressed in HSCs alone. The proteins in abundance from human PSC lines in this study were implicated for their role in maintaining the cellular structure[113]. The proteomic analysis of nicotine treated human, mouse and rat PSCs by GeLC-MS/MS approach demonstrated the differential expression of proteins and signaling pathways, while the expression of integral protein 2B, procollagen type VI alpha, toll interacting protein and amyloid interacting proteins was found to be common[114]. Expression of lysosomal proteins, indicators of pancreatic disease, proteins involved in defense mechanism and alteration in the phosphorylation sites were observed in another study[115]. Few other proteomic studies of similar kind have reported the expression of proteins related to inflammation, fibrosis, apoptosis, wound healing and proliferation[116]. The change in the expression profile of the proteome in response to various (TNF-α, FGF-2, CCL4 and IL-6) proinflammatory factors on immortalized human PSCs described their unique functions[117].

PSCS-PANCREATIC CANCER CELLS-CANCER STROMAL INTERACTIONS

It has now been conclusively demonstrated that majority (50%-80%) of PDAC volume is composed of a fibrous stroma, amidst which lay the islands of cancer cells[118]. There has been increasingly accumulating evidence that supports substantial two-way interactions between the stromal components and cancer cells and the association between the cancer cells and cancer associated PSCs have received several monikers such as "dangerous liaisons"[119], "friend or foe"[120] and "unholy alliance"[121]. The stroma in pancreatic cancer consists predominantly of collagenous fibres laid down by the PSCs, along with other cellular [mast cells, macrophages, lymphocytes, myeloid derived suppressor cells (MDSC) and endothelial cells][122-131] and non-cellular (ECM proteins such as collagen, laminin, fibronectin, glycoproteins, proteoglycans and glycosaminoglycans; non-ECM proteins such as growth factors, osteopontin, periostin and serine protein-acidic and rich in cysteine) components[132,133]. These stromal components can mediate the interaction between the PSCs and cancer cells and eventually influence the biological behavior and clinical outcomes of PDAC. Apart from vascular endothelial growth factor (VEGF) and angiopoietin-1, PSCs also secrete hepatocyte growth factor (HGF) and mediators responsible for endothelial cell proliferation and tube formation. This appears to operate through the HGF/c-MET pathway via induction of the downstream PI3K and p38 signaling pathways[134]. Of note, upon neutralizing the HGF activity, proliferation and migration of cancer cells could be inhibited and apoptosis could be induced[135].

Even though fibrosis that was observed early in development of PDAC led to the belief that PSC produced stroma is protective, this eventually shifted towards the concept of the stroma having a tumour permissive effect. However, the current opinion holds that PSC-stroma-cancer cell interaction is dynamic and stage-dependent, with protective effect in the earliest stage and harmful effect in later stages[38]. The mechanism of PSC induced fibrosis in PDAC is similar to that seen in CP. Therefore, in the next section we will discuss only the cancer specific interactions and phenotypic effects of stroma-cancer cell interactions. While the PSC-pancreatic cancer cell interactions result in cancer growth and PSC activation, interaction between PSCs and stromal cells may be instrumental in metastasis, immune evasion, tumour hypoxia and resistance to chemoradiotherapy.

PSC-PDAC crosstalk

Pancreatic intraepithelial neoplasia (PanINs) are the precursor lesions of PDAC. It is now well known that PSCs get activated even at the early PanIN stages of PDAC and initiates fibrosis around these precursor lesions. Several in vitro and in vivo studies have provided insight into the bipolar interactions between the PSCs and PDAC. In vitro co-culturing of PSCs with pancreatic cancer cells accelerated the proliferation and increased survival by inhibiting apoptosis[49,136]. Furthermore, co-culturing also resulted in epithelial-mesenchymal transition (EMT) as evidenced by increased expression of vimentin and snail (mesenchymal marker) with corresponding reduction in E-cadherin and cytokeratin (both epithelial markers)[137]. This was associated with migration of the cancer cells, which indicate the capability of PSCs to trigger the metastasis of pancreatic cancer cells[138].

Recurrence of PDAC after therapy has been postulated to be an effect of persistence of a treatment resistant cancer stem cell niche. PSCs have been shown to regulate the genesis of a cancer stem cell niche as marked by increased expression of stem cell markers such as ABCG-2, Lin28 and nestin, while also attaining capability to form spheroids in vitro[139]. Interestingly, it has been shown that the same PDAC can contain a heterogeneous population of PSCs based on the expression of CD10, which is a cell-membrane associated MMP. CD10(+) cells are associated with a higher propensity to proliferate and invade, thereby indicating that the relative proportion of PSC subtypes could also determine the disease biology and prognosis[140].

While the foregoing paragraphs discussed the effect of PSCs on pancreatic cancer cells, the cancer cells also induce profound effects on the PSCs. Pancreatic cancer cells produce factors such as PDGF, trefoil factor 1[141] and COX-2, which could induce PSC proliferation. COX-2 expression is upregulated not only in the cancer cells, but also in the PanINs and PSCs exposed to conditioned medium from cancer cell lines[142-145]. Galectin-1 and Galectin-3, members of galectin family of β-galactoside binding proteins, are also important drivers of the PSC-PDAC crosstalk. Galectin-3 expression by pancreatic cancer cell lines was found to promote its own proliferation along with PSCs[146,147]. Figure 5 outlines the overall crosstalk between PSCs and pancreatic cancer cells.

Figure 5
Figure 5 Crosstalk between pancreatic stellate cells and pancreatic cancer cells. Pancreatic stellate cells (PSCs) promote cancer cell proliferation, migration, invasion, EMT and metastasis. They also promote the cancer cell survival by decreasing cancer cell apoptosis and helps in chemoresistance. The cancer cells in turn promote PSC proliferation, contractility, migration and increased collagen synthesis. Apart from this, PSCs induce T cell anergy, activate mast cells and promote endothelial cell proliferation and tube formation. Together, these events mediated by PSCs and pancreatic cancer cells further aggravate pancreatic cancer progression. EMT: Epithelial-mesenchymal transition.
Role of PSCs on invasion and metastasis

Galectin-3[147], thrombospondin-2[148], stromal cell derived factor[149] and nerve growth factor (NGF)[150] expressed by PSCs are shown to drive the invasion of PDACs. Studies on xenograft models showed that PSCs exert a modulatory function and potentiate the invasiveness of SUIT2 pancreatic cancer cells expressing serine protease inhibitor nexin2 (SERPINE2)[151,152]. Pancreatic cancer cells and PSCs express fibroblast growth factor (FGF) and fibroblast growth factor receptor (FGFR) that have been shown to mediate interaction between the tumour and stromal cells resulting in development of an invasive phenotype[153]. A recent study confirmed that pro-invasive character results from nuclear localization of FGFR1 and FGF2 in PSCs[154]. Perhaps the most convincing and concept changing data on the role of PSCs in metastasis was reported in the study by Xu et al[155] which showed that the PSCs could rapidly acquire a tumour inductive property even after a short exposure of pancreatic cancer cells, thereby facilitating tumour growth and metastasis. The authors used a gender mismatch approach in which they injected a combination of female pancreatic cancer cells and male human PSCs into the pancreas of female nude mice. Interestingly, they could demonstrate Y-chromosome positive (i.e., the injected male human PSCs) in metastatic liver nodules. This implied that the PSCs from the liver could intravasate blood vessels, transport in circulation and extravasate into metastatic nodules alongside the metastatic cancer cells. The findings also suggest that the metastatic PSCs could mount an active stromal reaction even in the metastatic nodule. The property of transendothelial migration of the PSCs was further supported by in vitro studies and was found to be mediated by PDGF.

Besides contributing to distant metastasis, PSCs have also been implicated in neural invasion. This notion has been supported by studies that reported expression of neurotrophic factors such as glial derived neurotrophic factor and brain derived neurotrophic factor and stimulation of neurite formation towards pancreatic cancer cells by activated PSCs. These effects appear to be mediated by SHH paracrine signaling pathway[100,101].

Tumour hypoxia and resistance

Similar to CP, PDAC is also characterized by hypoxic microenvironment. Tumour hypoxia arising from fibro-inflammatory environment is shown to induce the expression of hypoxia-inducible factor-1α (HIF-1α) and stimulate the secretion of SHH ligand by cancer cells, leading to stromal deposition by tumour associated fibroblasts. Organotypic culture of thick pancreatic sections under hypoxic conditions depicted the activation (α-SMA) and proliferation (Ki67) of PSCs along with higher expression of HIF-1α, mediating the activation of hypoxic pathways[156]. In vitro studies on the role of hypoxic milieu on the interactions between PSCs and PDACs led to interesting observations. The hypoxia exposed PSCs expressed type I collagen and VEGF, showed increased migration and also promoted the endothelial cell proliferation, migration and angiogenesis[157]. Another study also yielded similar results where the hypoxia induced PSCs showed increased expression of periostin, collagen type I, VEGF and fibronectin. In co-cultures, the hypoxia treated PSCs enhanced the endostatin production by cancer cells and increased the endothelial cell growth[158]. A similar kind of study using 3D matrices also reported that the hypoxia induced procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2) in PSCs promotes cancer cell migration[159]. Periostin, a matrix protein, with its persevered autocrine loop was shown to promote ECM synthesis and cancer cell growth under hypoxia and starvation during chemotherapy by maintaining the activated phenotype of PSCs[160]. PSCs were also shown to express miR-21 and miR-210 under hypoxic conditions, where miR-210 was reported to regulate the interactions between PSCs and PDACs via ERK and Akt pathways[161] and miR-21 contributing to cancer cell invasion and metastasis[162]. Apart from these 2 miRs, miR-199a and miR-214 expressed by PSCs shown to have a pro-tumoral effect and also promote their own proliferation and migration[163]. Overexpression of miR-29, the expression of which was lost during the transformation of PSCs into activated phenotype, resulted in the reduction of collagen deposition, cancer cell growth and viability[164].

The outcomes of the above studies not only confirmed the central role of PSCs in desmoplasia but also exhibited the proangiogenic functions mediated by them in tumour hypoxia.

Immune escape of PDAC

Emerging data over the recent years have strongly suggested that pancreatic cancer cells could evade host immune surveillance. One of the major factors that mediate immune evasion of pancreatic cancer cells is by sequestration of CD8(+) cells within the stroma, thereby preventing them from invading peritumoral areas where they could mediate immune mediated injury to the cancer cells. This appears to be mediated by the chemokine CXCL12[165]. The other important mediator that sustain an immunosuppressive milieu is the β-galactoside binding lectin galectin-1, which is overexpressed by PSCs in pancreatic cancer. Using siRNA induced knockout and overexpression studies, it was shown that galectin-1 could induce T-cell apoptosis and reduced Th-1 response (with concomitant increase in Th-2 responses) and thereby reducing the immune mediated injury to the cancer cells. This was further reconfirmed and was shown that the effects were significantly higher in poorly differentiated tumours compared to the well-differentiated ones[166].

Other PSC mediated mechanisms that has been proposed to result in disruption of anti-tumour immunity are cytokine secretion by macrophages[167] and expression of the fibroblast activation protein-α[168], migration of MDSC[130], mast cell degradation leading to release of tryptase and IL-13[124].

THERAPEUTIC IMPLICATIONS

Given the background of substantial understanding of the mechanisms of PSC involvement in pancreatic fibrosis and pancreatic cancer, several experimental, preclinical and early phase clinical studies on CP and pancreatic cancer have appeared in the literature over the recent years. Experimental studies (both in vivo and in vitro) that have targeted the profibrogenic function of PSCs have shown favorable results; however these results have not yet been satisfactorily reproduced in human CP. Table 2 summarizes the drugs and their effects in experimental studies of CP[169-218].

Table 2 Therapeutic agents that have been used in experimental studies for inhibition of Pancreatic stellate cells in chronic pancreatitis.
RefAgentClass/type of agentIn vivo/in vitro (study) modelOutcome of the study
Nakamura et al[169] 2000FOY-007 FOY-305Synthetic serine protease inhibitorCytokine stimulated human periacinar fibroblast like cellsBoth attenuated proliferation and procollagen type I C-terminal peptide (PIP)
FOY-007 also inhibited collagen synthesis
Xie et al[170] 2002IS-741Carboxamide derivativeWistar Bonn/Kobori ratsSuppressed the expression of IL-6 and CINC and pancreatic acute phase proteins (PAP and p8)
Kuno et al[171] 2003LisinoprilAngiotensin-converting enzyme (ACE) inhibitorWistar Bonn/Kobori ratsIncreased pancreatic weight and decreased pancreatic MPO and serum ACE activity was observed
Decrease in serum MCP-1 levels, intra-pancreatic hydroxyproline content was identified
TGF-β1 mRNA overexpression was suppressed
Yamada et al[172] 2003CandesartanAngiotensin II receptor antagonistWistar Bonn/Kobori ratsIncreased pancreatic weight and expression of angiotensinogen and angiotensin II receptor type 2 mRNA
Decreased pancreatic MPO and serum ACE activity and hydroxyl proline content
Suppressed TGF-β1 mRNA overexpression
Masamune et al[173] 2003Y-27632 and HA-1077Rho kinase inhibitorsIsolated PSCs from male Wistar ratsInhibited α-SMA expression, proliferation, type I collagen production and chemotaxis
Nagashio et al[174] 2004AdTb-ExRAdenoviral vector system expressing TGF-β receptorCaerulein induced CP in BALB/c miceReduced the activated PSCs, number of apoptotic acinar cells and fibrosis
Weight of the pancreas increased
Zhao et al[175] 2005Mutant MCP-1DBTC induced CP in Lewis ratsDecreased MCP-1, fibrosis and hydroxyproline levels
Reduced IL6, TGF-β, IL-1β, MCP-1 and PDGF expression
Gibo et al[176] 2005Camostat mesilateOral protease inhibitorIn vivo: DBTC induced CP in Lewis rat In vitro: Culture activated PSCsIn vivo: Inflammation, fibrosis and cytokines expression was inhibited
In vitro: PSC proliferation and MCP-1 production was reduced
Yamada et al[177] 2005Lisinopril and candesartanAngiotensin-converting enzyme inhibitor and angiotensin II type 1 receptor blockerWistar Bonn/Kobori ratsReduced MPO activity, hydroxyproline content, inflammation and fibrosis in combination therapy
Suppressed mRNA expression of TGF-β1, PDGF- β and TNF-α
van Westerloo et al[178] 2005TroglitazonePPAR-γ ligandCaerulein induced experimental CP in female C57BL6 miceIntrapancreatic fibrosis and hydroxy proline contents were reduced
Attenuated increase in MPO content and TGF-β1 levels
Reding et al[179] 2006RofecoxibCOX-2 inhibitorWistar Bonn/Kobori ratsReduced TGF-β, collagen synthesis, inflammation and macrophage infiltration
Asaumi et al[180] 2006Epigallocatechin-3-gallate EGCGAntioxidant of polyphenolsEthanol stimulated PSCs isolated from Wistar ratsInhibited lipid peroxidation, SOD activity and p38 phosphorylation
Decreased TGF-β1 and collagen secretion
Baumert et al[181] 2005 Fitzner et al[182] 2006IFN-γAntifibrotic cytokineCulture activated PSCs, isolated from LEW.1W rats and pancreatic stellate cell linesDiminished PSC proliferation and collagen synthesis
Inhibited α-SMA expression
Induction of quiescent phenotype mediated through activated STAT1
Ohashi et al[183] 2006Thioredoxin-1 (TRX-1)Redox-regulating protein with antioxidative activityCaerulein induced CP in wild type C57BL/6 mice and transgenic mice overexpressing TRX-1Attenuated PSC activation and fibrosis
TGF-β1 and PDGF expression was reduced
Lower levels of MCP-1 in serum and acinar cells
McCarroll et al[184] 2006Retinol and its metabolitesVitamers of vitamin AEthanol stimulated culture activated rat PSCsInhibited PSC activation, proliferation, expression of collagen I. All MAP kinases were activated
Tasci et al[185] 2007AllopurinolXanthine oxidase inhibitorTrinitrobenzene sulfonic acid (TNBS) induced CP in Sprague-Dawley ratsPSC activation was inhibited in vivo
Lower collagen deposition and lobular and sub-lobular atrophy was observed
Lu et al[186] 2007Ascorbic acidAntioxidantDBTC induced CP in Sprague-Dawley ratsDecreased malondialdehyde (MDA), hyaluronic acid, laminin concentrations and pancreatic injury
Increased superoxide dismutase activity
Shirahige et al[187] 2007TaurineAmino sulfonic acidDBTC induced pancreatic fibrosis in Wistar rats Culture activated PSCs from Wistar ratsImproved pancreatic fibrosis in rats. Increased IL-6 and decreased IL-2 were observed in pancreatic tissue homogenates
PSC culture supernatants showed decreased type I collagen, MMP-2 and TGF-β1
Rickmann et al[188]2007TocotrienolsVitamin E family membersCulture activated PSCs isolated from Wistar ratsReduced viability of activated PSCs by apoptosis and autophagy
Michalski CW et al[189] 2008Canabinoid WIN 55,212-2Aminoalkylindole derivativeHuman PSCs from CP tissuesReduced fibronectin, collagen1 and α-SMA levels
Decreased IL-6, MCP-1 and MMP-2 secretion and invasiveness by PSCs
Weylandt et al[190] 2008Omega-3 polyunsaturated fatty acids (n-3 PUFA )Polyunsaturated fatsCaerulein-induced CP in fat-1 transgenic miceIncreased n-3 PUFA tissue levels
Decreased PSC activation
Less pancreatic fibrosis and collagen content
Karatas et al[191] 2008HalofuginoneSynthetic halogenated derivative of febrifugineComplete pancreatic duct obstruction and caerulein hyperstimulation in female Wistar ratsLower serum amylase, lipase, hyaluronic acid, nitric oxide levels and tissue hydroxyproline levels
Pancreatic inflammation and acinar cell atrophy was reduced
Fitzner et al[192] 2008BosentanET-1-receptor antagonistCulture activated rat PSCsInhibited PSC proliferation and collagen synthesis
Reduced the expression of ET-1, α-SMA and CTGF
Schwer et al[193] 2010Carbon monoxide-releasing molecules-2 (CORMs)Metal carbonyl compounds delivering carbon monoxideCulture activated PSCs isolated from Wistar ratsPSC proliferation was inhibited through p38/HO-1 pathway activation
Nathan et al[194] 2010Pancreatic secretory trypsin inhibitor (PSTI)Caerulein induced CP in C57Bl/6 PSTI transgenic miceDecreased MPO activity and inflammatory cell infiltration
Reduction in collagen I and fibronectin mRNA levels
González et al[195] 2011Palm oil tocotrienol-rich fractionVitamin E family membersArginine induced chronic like pancreatitis in Wistar ratsReduced amylase, hydroxyproline and TGF-β1 levels were observed
Diminished α-SMA, fibronectin and collagen expression was identified
Long et al[196] 2011OctreotideAnalog of somatostatinIn vivo: PSC induced Pancreas Graft Fibrosis in Rats (Sprague Dawley rats as donors and Wistar rats as recipients) In vitro: PSCs isolated from Sprague Dawley ratsIn vivo: Reduced inflammatory cell infiltration and expression of α-SMA, collagen I and TGF-β1
In vitro: PSC proliferation and activation was inhibited
Li et al[197] 2011Pancreatic stone protein ⁄ regenerating proteinSecretory stress proteins familyCulture activated PSCs from human CP tissue obtained by outgrowth methodInhibited PSC proliferation, migration and reduced. collagen I and fibronectin
Increased MMP/TIMP ratio and promoted fibrolysis
Tang et al[198] 2011SinisanChinese herbTNBS induced CP in Sprague-Dawley ratsDecreased serum amylase
mRNA expression of TNF-α, IL-1β and COX-2 were reduced and IL-10 was increased
α-SMA expression was reduced
Wei et al[199] 2011PravastatinCompetitive inhibitor of HMG-CoAPancreatic ductal hypertension induced CP in Wistar ratsAttenuated fibrosis and mRNA levels of TNF-α and TGF-β1 and increased IL-10 expression
Exocrine secretion was improved
SOD activity was increased
Li et al[200] 2011α-TocopherolVitamin E family memberTNBS induced CP in Sprague-Dawley ratsReduced fibrosis and enhanced survival rate
Pancreatic weight was increased in CP model
Monteiro et al[201] 2012Vitamin E supplementationEthanol induced (alcoholic) CP in Wistar ratsmRNA levels of α-SMA, COX-2, IL-6, MIP-3α and TNF-α were decreased and PAP was increased
Matsushita et al[202] 2012TaurineAmino sulfonic acidIn vivo: DBTC induced CP in Wistar rats In vitro: AR42J acinar cellsInhibited acinar cell apoptosis
Yang et al[203] 2012L-CysteineAmino acidIn vivo: TNBS induced CP in Sprague-Dawley rats In vitro: Culture activated PSCsDecreased α-SMA, TIMP-1, IL-1β TGF-β1 expression and hydroxylproline levels and increased MMP-2 levels
Suppressed PSC proliferation and ECM synthesis
Bai et al[204] 2012SulindacNon-steroidal anti-inflammatory drugCaerulein induced CP in C57BL/6 miceReduced fibrosis, acinar cell loss and inflammatory cell infiltration
TNF-α and MCP-1 levels were decreased
Expression of TGF-β, PDGF-β, SHH and Gli was reduced
Lee et al[205] 2012Simvastatin and TroglitazoneHMG-CoA reductase inhibitor and PPAR agonistsCulture activated PSCs isolated from Sprague-Dawley ratsPSC proliferation was inhibited synergistically
Shen et al[206] 2013rCXCL9ChemokineIn vivo: TNBS induced CP in Sprague-Dawley rats In vitro: Culture activated PSCs from Sprague-Dawley ratsIn vivo: reduced fibrosis
In vitro: protein expression of collagen 1α1 and TGF-β1 decreased
Gao et al[207] 2013Bone morphogeneic proteinsTGF-β superfamily members,In vivo: Caerulein induced CP in female Swiss Webster mice In vitro: PSCs isolated from female Swiss Webster mice and human pancreatic tissueIn vivo: BMP2 protein levels were increased
In vitro: PSC pre-treatment with BMP2 attenuated TGF-β1, α-SMA, fibronectin and collagen type Iα expression
Zhou et al[208] 2013EdaravoneFree radical scavengerDBTC induced CP in Sprague-Dawley ratsRats body weight was improved and reduced the fibrosis
SOD activity was increased and MDA levels were decreased
TGF-β, TNF-α and IL-6 levels were downregulated
NF-κB and PSC activation was inhibited
Niina et al[209] 2014ONO-1301Prostacyclin agonist,DBTC induced CP in Lewis ratsReduced interstitial fibrosis and inflammatory cell infiltration
Increased HGF and decreased IL-1β, TNF-α, TGF-β, MCP-1 and collagen mRNA expression was observed
Mrazek et al[210] 2015ApigeninHydroxyflavoneIn vivo: Caerulein induced pancreatitis in C57/BL6 mice In vitro: PSCs isolated from human pancreatic tissuesIn vivo: Reduced pancreatic fibrosis and retained acinar cell morphology
In vitro: Induced PSC apoptosis and death
Downregulated the parathyroid hormone related protein induced fibronectin, collagen 1α1, PCNA, TGF-β and IL-6 expression
Tsang et al[211] 2015Trans-resveratrolNatural stilbenoidRat pancreatic stellate cell line LTC-14Collagen type I, α-SMA and fibronectin was downregulated both at mRNA and protein level
NF-κB activation was decreased
Lin et al[212] 2015Rhein, Emodin, and CurcuminPhenolic compoundsRat pancreatic stellate cell line LTC-14Collagen type I, α-SMA and fibronectin expression was decreased
Gundewar et al[213] 2015L49H37Curcumin analogImmortalized human pancreatic stellate cell lineInhibited PSC proliferation and promoted apoptosis
Decreased the phosphorylation of ERK1/2
Tsang et al[214] 2015Eruberin AFlavanol glycosideRat PSC line LTC-14Inhibited expression of α-SMA Collagen type I and fibronectin. Reduced the activation of NF-κB and phosphorylation PI3K/AKT
Bläuer et al[215] 20151,25-dihydroxyvitamin D3Vitamin D metabolitePSCs isolated from C57BL6JOlaHsd mouseAntiproliferative and antifibrotic effects were observed
Xiao et al[216] 2015Retinoic acidVitamin A MetaboliteIn vivo: Caerulein induced CP in Balb/c mice In vitro: Culture activated PSCsDecreased expression of TGF-βRII, collagen 1α1 PDGF-Rβ and β-catenin
Nuclear translation of β-catenin was decreased
Wnt 2 and β-catenin protein expression was downregulated
Inhibited PSC proliferation and induced apoptosis
Witteck et al[217] 2015Trametinib and dactolisibMEK inhibitor and PI3kinase/mTOR inhibitorCulture activated PSCs isolated from Lewis ratsBoth drugs inhibited PSC proliferation
Trametinib suppressed the expression of IL-6 and TGF-β1
Dactolisib decreased the levels of α-SMA and Collagen type Iα1
Ulmasov et al[218] 2016CWHM-12 (RGD peptidomimetic compound)Integrin inhibitorIn vivo: Caerulein induced pancreatitis in C57/BL6 mice In vitro: Rat PSC line LTC-14Pancreatic fibrosis, acinar cell atrophy and loss was reduced
Decreased the expression of TGF-β regulated genes and PSC activation

In the context of pancreatic cancer, where conventional chemotherapy has shown dismal results, the current concept is to target the stroma along with conventional chemotherapy. Since the pancreatic cancer stroma has been shown to be associated with tumour hypoxia, metastasis, drug resistance, it is expected that prior stromal degradation could result in chemosensitivity of the tumour even with the conventional chemotherapeutic drugs. Table 3 shows the recently tested drugs/biologics that targeted pancreatic cancer stroma in preclinical studies[219-242]. Besides the preclinical studies, several SHH pathway inhibitors have also been tested in advanced or metastatic PDACs in phase I and II studies (both open labeled and randomized double-blind controlled trials). Few of these include Vismodegib (GDC-0449), Saridegib (IPI-926) and Erismodegib (LDE225), PDGFR inhibitor (TKI258), hyaluran (PEGPH20) and dasatinib, to name a few. These have been used along with gemcitabine and/or nab-Paclitaxel and FOLFIRINOX. Discussion of details of the study designs and results of these trials are out of the scope of this review and can be obtained from recent high quality reviews[243,244].

Table 3 Therapeutic agents that have been evaluated in experimental/pre-clinical studies targeting pancreatic stellate cells and cancer stroma in pancreatic ductal adenocarcinoma.
RefAgentClass/type of agentIn vivo/in vitro (study) modelOutcome of the study
Feldmann et al[219] 2007CyclopamineSteroidal alkaloidOrthotopic xenograft modelInhibited cancer cell invasion and metastasis by suppressing hedgehog
Diep et al[220] 2011Erlotinib RDEA119 and AZD6244EGFR tyrosine Kinase and MAP kinase inhibitorsIn vitro: Pancreatic cancer cell lines In vivo: BxPC-3 and MIA PaCa-2 mice xenograft modelInhibited cancer cell proliferation, EGF receptor signaling and induced apoptosis
Suppressed tumour growth
Froeling et al[221] 2011ATRA, 9-cis-RA and 13-cis-RAMetabolites of vitamin AIn vivo: LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre mice In vitro: AsPc1 and Capan1 pancreatic cancer cell lines, PS1 and other PSC cell linesRetinoic acid induced PSC quiescence and decreased migration
Decreased and induced proliferation and apoptosis of cancer cells
Chauhan et al[222] 2013LosartanAngiotensin inhibitorOrthotopic mice modelReduced stromal collagen production, expression of TGF-β1, CCN2 and ET-1
Improved drug and oxygen delivery to tumour
Sun et al[223] 2013CurcuminPhenolic compoundTGF-β1 stimulated PANC-1 cell lineInhibited proliferation and promoted apoptosis
Cancer cell invasion and migration was decreased
Edderkaoui et al[224] 2013Ellagic acid EmbelinPolyphenolic and benzoquinone phytochemicalPancreatic cancer cells and PSCsInduced apoptosis and inhibited proliferation
NF-κB activity was decreased
Macha et al[225] 2013GuggulsteronePlant polyphenolCD18/HPAF and Capan1 cell clonesInhibited growth and colony formation
Induced apoptosis and arrested cell cycle
Decreased motility and invasion
Kozono et al[226] 2013PirfenidonePyridone compoundIn vivo: Orthotopic tumour mice Model In vitro: PSCs isolated from pancreatic tissueIn vivo: Reduced tumour growth, PSC proliferation and the deposition of collagen type I and periostin in tumours was decreased
In vitro: Proliferation, invasiveness and migration of PSCs was inhibited
Guan et al[227] 2014Retinoic acidVitamin A derivativePanc-1 and Aspc-1 cell lines Cancer associated fibroblastsReduced α-SMA, FAP and IL-6 expression
Inhibited cancer cell migration and EMT
Gonzalez-Villasana et al[228] 2014Bisphosphonates and nab-paclitaxelMonocyte-macrophage lineage inhibitorsIn vitro: Human PSCs and cancer cell line In vivo: Orthotopic mice modelIn vitro: Inhibited PSC activation, proliferation MCP-1 release and collagen 1 expression and induced apoptosis
In vivo: Reduced tumour size, fibrosis, proliferation and increased apoptosis
Pomianowska et al[229] 2014Prostaglandin E2 (PGE2)Lipid compoundHuman PSCs isolated from resected pancreatic tumour tissueIL-1β and EGF induced COX-2 expression, TGF-β induced collagen synthesis and PDGF induced PSC proliferation was inhibited
Gong et al[230] 2014NexrutinePhytoceutical with COX-2 Inhibitor activityIn vitro: pancreatic cancer cell lines In vivo: BK5-COX-2 transgenic miceIn vitro: promoted cancer cell apoptosis and reduced their growth
Suppressed COX-2 expression
In vivo: NF-κB and Stat3 activity and fibrosis was decreased
Yan et al[231] 2014Crizotinibc-MET/HGF receptor and ALK tyrosine kinases inhibitorIn vitro: Human pancreatic cancer cell lines AsPC-1, PANC-1, MIA PaCa-2 and Capan-1 In vivo: Mouse xenograft modelIn vitro: Growth and proliferation was inhibited
Induced apoptosis
Inhibited ALK activity
In vivo: Inhibited angiogenesis, tumour growth and ALK activity
Zhang et al[232] 20145-AzacytidineCytidine analogueBxpc-3 cancer cell lineInhibited cancer cell proliferation by suppressing Wnt/β-catenin signaling
Wang et al[233] 2014miR-216amicroRNAIn vitro: Capan-2 and PANC-1 pancreatic cancer cell lines In vivo: BALB/c nude miceIn vitro: Inhibited cell growth and induced apoptosis
Down regulated survivin and XIAP expression
In vivo: Inhibited xenograft tumour growth by suppressing JAK2/STAT3 signaling pathway
Kumar et al[234] 2015miR-let7b and GDC-0449microRNA and Hedgehog inhibitorIn vitro: Capan-1, HPAF-II, T3M4 and MIA PaCa-2 cell lines In vivo: Athymic nude mice bearing ectopic tumourIn vitro: Decreased cell proliferation and induced apoptosis via Gli dependent mechanism
In vivo: Reduced tumour cell proliferation with increased apoptosis and tumour growth was inhibited
Petrova et al[235] 2015RU-SKI 43Hedgehog acyltransferase inhibitorIn vitro: Pancreatic cancer cell lines In vivo: Panc-1 xenograft mouse modelIn vitro: Reduced cancer cell proliferation and Gli-1 activation through Smo independent signaling
Decreased Akt and mTOR activity
In vivo: Tumour growth decreased
Massó-Vallés et al[236] 2015IbrutinibTyrosine kinase inhibitorTransgenic mouse and xenograft mice modelsReduced fibrosis and extended survival
Zhou et al[237] 2015Zileuton5-LOX inhibitorPancreatic cancer SW1990 cell lineInduced apoptosis, decreased proliferation and expression of 5-lipoxygenase
Lui et al[238] 2015Desferrioxamine, Di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone and Di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazoneThiosemicarbazonesIn vitro: PANC-1 and MIAPaCa-2 In vivo: PANC-1 tumour xenograft miceActivation of the non-receptor tyrosine kinase Src and cAbl was decreased in vitro and STAT3 activation was reduced in both in vivo and in vitro condition
Khan et al[239] 2015OrmeloxifeneNonsteroidal drugPancreatic cancer cell lines and PDAC xenograft miceInhibited cell proliferation, tumour stroma through SHH pathway and stromal cell infiltration
Decreased collagen I expression
Restored the tumour-suppressor miR-132 expression
Liu et al[240] 2016OridoninTetracycline diterpenoid compoundAspc1, Bxpc3, Panc1 and SW1990 cell linesMigration and EMT was inhibited by affecting Wnt/β-catenin signal events
Haqq et al[241] 2016Gemcitabine with omega-3 polyunsaturated fatty acid emulsion, (LipidemTM)Nucleoside analogIn vitro studies using pancreatic cancer cell lines Capan-1 and Panc-1 and PSC cell line; RLT-PSCDrugs showed antiproliferative and anti-invasive effects
Ji et al[242] 2016MMP2 responsive liposome loaded with Pirfenidone and gemcitabine---In vivo: BALB/c nude Orthotopic tumour mice model In vitro: Human PSCs isolated from surgical specimens of Pancreatic cancerPirfenidone inhibited collagen I and TGF-β expression in PSCs
Gemcitabine killed pancreatic tumour cells
Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: India

Peer-review report classification

Grade A (Excellent): A, A

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Larentzakis A, Peng SY, Shimizu Y S- Editor: Qi Y L- Editor: A E- Editor: Liu WX

References
1.  Ito T. Cytological studies on stellate cells of Kupffer and fat storing cells in the capillary wall of the human liver. Acta Anat Nippon. 1951;26:42.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Wake K. Development of vitamin A-rich lipid droplets in multivesicular bodies of rat liver stellate cells. J Cell Biol. 1974;63:683-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 94]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
3.  Wake K. "Sternzellen" in the liver: perisinusoidal cells with special reference to storage of vitamin A. Am J Anat. 1971;132:429-462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 244]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
4.  Wake K, Motomatsu K, Senoo H. Stellate cells storing retinol in the liver of adult lamprey, Lampetra japonica. Cell Tissue Res. 1987;249:289-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
5.  Watari N, Hotta Y, Mabuchi Y. Morphological studies on a vitamin A-storing cell and its complex with macrophage observed in mouse pancreatic tissues following excess vitamin A administration. Okajimas Folia Anat Jpn. 1982;58:837-858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 90]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
6.  Ikejiri N. The vitamin A-storing cells in the human and rat pancreas. Kurume Med J. 1990;37:67-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 56]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
7.  Apte MV, Haber PS, Applegate TL, Norton ID, McCaughan GW, Korsten MA, Pirola RC, Wilson JS. Periacinar stellate shaped cells in rat pancreas: identification, isolation, and culture. Gut. 1998;43:128-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 658]  [Cited by in F6Publishing: 687]  [Article Influence: 26.4]  [Reference Citation Analysis (0)]
8.  Bachem MG, Schneider E, Gross H, Weidenbach H, Schmid RM, Menke A, Siech M, Beger H, Grünert A, Adler G. Identification, culture, and characterization of pancreatic stellate cells in rats and humans. Gastroenterology. 1998;115:421-432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 744]  [Cited by in F6Publishing: 766]  [Article Influence: 29.5]  [Reference Citation Analysis (0)]
9.  Masamune A, Satoh M, Kikuta K, Suzuki N, Shimosegawa T. Establishment and characterization of a rat pancreatic stellate cell line by spontaneous immortalization. World J Gastroenterol. 2003;9:2751-2758.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 24]  [Cited by in F6Publishing: 29]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
10.  Sparmann G, Hohenadl C, Tornøe J, Jaster R, Fitzner B, Koczan D, Thiesen HJ, Glass A, Winder D, Liebe S. Generation and characterization of immortalized rat pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2004;287:G211-G219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 51]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
11.  Jesnowski R, Fürst D, Ringel J, Chen Y, Schrödel A, Kleeff J, Kolb A, Schareck WD, Löhr M. Immortalization of pancreatic stellate cells as an in vitro model of pancreatic fibrosis: deactivation is induced by matrigel and N-acetylcysteine. Lab Invest. 2005;85:1276-1291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 120]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
12.  Rosendahl AH, Gundewar C, Said Hilmersson K, Ni L, Saleem MA, Andersson R. Conditionally immortalized human pancreatic stellate cell lines demonstrate enhanced proliferation and migration in response to IGF-I. Exp Cell Res. 2015;330:300-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 22]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
13.  Zha M, Li F, Xu W, Chen B, Sun Z. Isolation and characterization of islet stellate cells in rat. Islets. 2014;6:e28701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 39]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
14.  Zha M, Xu W, Jones PM, Sun Z. Isolation and characterization of human islet stellate cells. Exp Cell Res. 2016;341:61-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 24]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
15.  Cassiman D, Barlow A, Vander Borght S, Libbrecht L, Pachnis V. Hepatic stellate cells do not derive from the neural crest. J Hepatol. 2006;44:1098-1104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 64]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
16.  Asahina K, Tsai SY, Li P, Ishii M, Maxson RE, Sucov HM, Tsukamoto H. Mesenchymal origin of hepatic stellate cells, submesothelial cells, and perivascular mesenchymal cells during mouse liver development. Hepatology. 2009;49:998-1011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 163]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
17.  Asahina K, Zhou B, Pu WT, Tsukamoto H. Septum transversum-derived mesothelium gives rise to hepatic stellate cells and perivascular mesenchymal cells in developing mouse liver. Hepatology. 2011;53:983-995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 229]  [Cited by in F6Publishing: 231]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
18.  Marrache F, Pendyala S, Bhagat G, Betz KS, Song Z, Wang TC. Role of bone marrow-derived cells in experimental chronic pancreatitis. Gut. 2008;57:1113-1120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 51]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
19.  Scarlett CJ, Colvin EK, Pinese M, Chang DK, Morey AL, Musgrove EA, Pajic M, Apte M, Henshall SM, Sutherland RL. Recruitment and activation of pancreatic stellate cells from the bone marrow in pancreatic cancer: a model of tumor-host interaction. PLoS One. 2011;6:e26088.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 52]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
20.  Sparmann G, Kruse ML, Hofmeister-Mielke N, Koczan D, Jaster R, Liebe S, Wolff D, Emmrich J. Bone marrow-derived pancreatic stellate cells in rats. Cell Res. 2010;20:288-298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
21.  Ino K, Masuya M, Tawara I, Miyata E, Oda K, Nakamori Y, Suzuki K, Ohishi K, Katayama N. Monocytes infiltrate the pancreas via the MCP-1/CCR2 pathway and differentiate into stellate cells. PLoS One. 2014;9:e84889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
22.  Kim N, Yoo W, Lee J, Kim H, Lee H, Kim YS, Kim DU, Oh J. Formation of vitamin A lipid droplets in pancreatic stellate cells requires albumin. Gut. 2009;58:1382-1390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
23.  Kim N, Choi S, Lim C, Lee H, Oh J. Albumin mediates PPAR-gamma or C/EBP-alpha-induced phenotypic changes in pancreatic stellate cells. Biochem Biophys Res Commun. 2010;391:640-644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
24.  Omary MB, Lugea A, Lowe AW, Pandol SJ. The pancreatic stellate cell: a star on the rise in pancreatic diseases. J Clin Invest. 2007;117:50-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 473]  [Cited by in F6Publishing: 526]  [Article Influence: 30.9]  [Reference Citation Analysis (0)]
25.  Ding Z, Maubach G, Masamune A, Zhuo L. Glial fibrillary acidic protein promoter targets pancreatic stellate cells. Dig Liver Dis. 2009;41:229-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
26.  Riopel MM, Li J, Liu S, Leask A, Wang R. β1 integrin-extracellular matrix interactions are essential for maintaining exocrine pancreas architecture and function. Lab Invest. 2013;93:31-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
27.  Means AL. Pancreatic stellate cells: small cells with a big role in tissue homeostasis. Lab Invest. 2013;93:4-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
28.  Masamune A, Watanabe T, Kikuta K, Shimosegawa T. Roles of pancreatic stellate cells in pancreatic inflammation and fibrosis. Clin Gastroenterol Hepatol. 2009;7:S48-S54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 193]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
29.  Phillips PA, McCarroll JA, Park S, Wu MJ, Pirola R, Korsten M, Wilson JS, Apte MV. Rat pancreatic stellate cells secrete matrix metalloproteinases: implications for extracellular matrix turnover. Gut. 2003;52:275-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 212]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
30.  Phillips PA, Yang L, Shulkes A, Vonlaufen A, Poljak A, Bustamante S, Warren A, Xu Z, Guilhaus M, Pirola R. Pancreatic stellate cells produce acetylcholine and may play a role in pancreatic exocrine secretion. Proc Natl Acad Sci USA. 2010;107:17397-17402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 71]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
31.  Shimizu K, Kobayashi M, Tahara J, Shiratori K. Cytokines and peroxisome proliferator- activated receptor gamma ligand regulate phagocytosis by pancreatic stellate cells. Gastroenterology. 2005;128:2105-2118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 68]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
32.  Berna MJ, Seiz O, Nast JF, Benten D, Bläker M, Koch J, Lohse AW, Pace A. CCK1 and CCK2 receptors are expressed on pancreatic stellate cells and induce collagen production. J Biol Chem. 2010;285:38905-38914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
33.  Mato E, Lucas M, Petriz J, Gomis R, Novials A. Identification of a pancreatic stellate cell population with properties of progenitor cells: new role for stellate cells in the pancreas. Biochem J. 2009;421:181-191.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Docherty K. Pancreatic stellate cells can form new beta-like cells. Biochem J. 2009;421:e1-e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
35.  Kordes C, Sawitza I, Götze S, Häussinger D. Stellate cells from rat pancreas are stem cells and can contribute to liver regeneration. PLoS One. 2012;7:e51878.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
36.  Tahara J, Shimizu K, Shiratori K. Engulfment of necrotic acinar cells by pancreatic stellate cells inhibits pancreatic fibrogenesis. Pancreas. 2008;37:69-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 14]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
37.  Nakamura T, Ito T, Oono T, Igarashi H, Fujimori N, Uchida M, Niina Y, Yasuda M, Suzuki K, Takayanagi R. Bacterial DNA promotes proliferation of rat pancreatic stellate cells thorough toll-like receptor 9: potential mechanisms for bacterially induced fibrosis. Pancreas. 2011;40:823-831.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
38.  Apte M, Pirola RC, Wilson JS. Pancreatic stellate cell: physiologic role, role in fibrosis and cancer. Curr Opin Gastroenterol. 2015;31:416-423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 86]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
39.  Hong OK, Lee SH, Rhee M, Ko SH, Cho JH, Choi YH, Song KH, Son HY, Yoon KH. Hyperglycemia and hyperinsulinemia have additive effects on activation and proliferation of pancreatic stellate cells: possible explanation of islet-specific fibrosis in type 2 diabetes mellitus. J Cell Biochem. 2007;101:665-675.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
40.  Kikuta K, Masamune A, Hamada S, Takikawa T, Nakano E, Shimosegawa T. Pancreatic stellate cells reduce insulin expression and induce apoptosis in pancreatic β-cells. Biochem Biophys Res Commun. 2013;433:292-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
41.  Zha M, Xu W, Zhai Q, Li F, Chen B, Sun Z. High glucose aggravates the detrimental effects of pancreatic stellate cells on Beta-cell function. Int J Endocrinol. 2014;2014:165612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
42.  Li FF, Chen BJ, Li W, Li L, Zha M, Zhou S, Bachem MG, Sun ZL. Islet Stellate Cells Isolated from Fibrotic Islet of Goto-Kakizaki Rats Affect Biological Behavior of Beta-Cell. J Diabetes Res. 2016;2016:6924593.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 19]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
43.  Zang G, Sandberg M, Carlsson PO, Welsh N, Jansson L, Barbu A. Activated pancreatic stellate cells can impair pancreatic islet function in mice. Ups J Med Sci. 2015;120:169-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 39]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
44.  Li F, Chen B, Li L, Zha M, Zhou S, Wu T, Bachem MG, Sun Z. INS-1 cells inhibit the production of extracellular matrix from pancreatic stellate cells. J Mol Histol. 2014;45:321-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
45.  Xu W, Li W, Wang Y, Zha M, Yao H, Jones PM, Sun Z. Regenerating islet-derived protein 1 inhibits the activation of islet stellate cells isolated from diabetic mice. Oncotarget. 2015;6:37054-37065.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
46.  Fitzner B, Müller S, Walther M, Fischer M, Engelmann R, Müller-Hilke B, Pützer BM, Kreutzer M, Nizze H, Jaster R. Senescence determines the fate of activated rat pancreatic stellate cells. J Cell Mol Med. 2012;16:2620-2630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 45]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
47.  Patel M, Fine DR. Fibrogenesis in the pancreas after acinar cell injury. Scand J Surg. 2005;94:108-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
48.  Li N, Li Y, Li Z, Huang C, Yang Y, Lang M, Cao J, Jiang W, Xu Y, Dong J. Hypoxia Inducible Factor 1 (HIF-1) Recruits Macrophage to Activate Pancreatic Stellate Cells in Pancreatic Ductal Adenocarcinoma. Int J Mol Sci. 2016;17:E799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 67]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
49.  Vonlaufen A, Joshi S, Qu C, Phillips PA, Xu Z, Parker NR, Toi CS, Pirola RC, Wilson JS, Goldstein D. Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 2008;68:2085-2093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 355]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
50.  Bachem MG, Schünemann M, Ramadani M, Siech M, Beger H, Buck A, Zhou S, Schmid-Kotsas A, Adler G. Pancreatic carcinoma cells induce fibrosis by stimulating proliferation and matrix synthesis of stellate cells. Gastroenterology. 2005;128:907-921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 460]  [Cited by in F6Publishing: 485]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
51.  Apte MV, Norton ID, Wilson JS. Ethanol induced acinar cell injury. Alcohol Alcohol Suppl. 1994;2:365-368.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Haber PS, Apte MV, Applegate TL, Norton ID, Korsten MA, Pirola RC, Wilson JS. Metabolism of ethanol by rat pancreatic acinar cells. J Lab Clin Med. 1998;132:294-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
53.  Wilson JS, Apte MV. Role of alcohol metabolism in alcoholic pancreatitis. Pancreas. 2003;27:311-315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 50]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
54.  Masamune A, Kikuta K, Satoh M, Satoh A, Shimosegawa T. Alcohol activates activator protein-1 and mitogen-activated protein kinases in rat pancreatic stellate cells. J Pharmacol Exp Ther. 2002;302:36-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 85]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
55.  Apte MV, Pirola RC, Wilson JS. Battle-scarred pancreas: role of alcohol and pancreatic stellate cells in pancreatic fibrosis. J Gastroenterol Hepatol. 2006;21 Suppl 3:S97-S101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 43]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
56.  Casini A, Galli A, Pignalosa P, Frulloni L, Grappone C, Milani S, Pederzoli P, Cavallini G, Surrenti C. Collagen type I synthesized by pancreatic periacinar stellate cells (PSC) co-localizes with lipid peroxidation-derived aldehydes in chronic alcoholic pancreatitis. J Pathol. 2000;192:81-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
57.  Hu R, Wang YL, Edderkaoui M, Lugea A, Apte MV, Pandol SJ. Ethanol augments PDGF-induced NADPH oxidase activity and proliferation in rat pancreatic stellate cells. Pancreatology. 2007;7:332-340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
58.  Gao R, Brigstock DR. Connective tissue growth factor (CCN2) in rat pancreatic stellate cell function: integrin alpha5beta1 as a novel CCN2 receptor. Gastroenterology. 2005;129:1019-1030.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 96]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
59.  Charrier A, Chen R, Chen L, Kemper S, Hattori T, Takigawa M, Brigstock DR. Connective tissue growth factor (CCN2) and microRNA-21 are components of a positive feedback loop in pancreatic stellate cells (PSC) during chronic pancreatitis and are exported in PSC-derived exosomes. J Cell Commun Signal. 2014;8:147-156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 66]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
60.  Lee AT, Xu Z, Pothula SP, Patel MB, Pirola RC, Wilson JS, Apte MV. Alcohol and cigarette smoke components activate human pancreatic stellate cells: implications for the progression of chronic pancreatitis. Alcohol Clin Exp Res. 2015;39:2123-2133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 40]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
61.  Tolstrup JS, Kristiansen L, Becker U, Grønbaek M. Smoking and risk of acute and chronic pancreatitis among women and men: a population-based cohort study. Arch Intern Med. 2009;169:603-609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 124]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
62.  Asaumi H, Watanabe S, Taguchi M, Tashiro M, Otsuki M. Externally applied pressure activates pancreatic stellate cells through the generation of intracellular reactive oxygen species. Am J Physiol Gastrointest Liver Physiol. 2007;293:G972-G978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 41]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
63.  Watanabe S, Nagashio Y, Asaumi H, Nomiyama Y, Taguchi M, Tashiro M, Kihara Y, Nakamura H, Otsuki M. Pressure activates rat pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2004;287:G1175-G1181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 38]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
64.  Ko SH, Hong OK, Kim JW, Ahn YB, Song KH, Cha BY, Son HY, Kim MJ, Jeong IK, Yoon KH. High glucose increases extracellular matrix production in pancreatic stellate cells by activating the renin-angiotensin system. J Cell Biochem. 2006;98:343-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 68]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
65.  Nomiyama Y, Tashiro M, Yamaguchi T, Watanabe S, Taguchi M, Asaumi H, Nakamura H, Otsuki M. High glucose activates rat pancreatic stellate cells through protein kinase C and p38 mitogen-activated protein kinase pathway. Pancreas. 2007;34:364-372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 42]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
66.  Kiss K, Baghy K, Spisák S, Szanyi S, Tulassay Z, Zalatnai A, Löhr JM, Jesenofsky R, Kovalszky I, Firneisz G. Chronic hyperglycemia induces trans-differentiation of human pancreatic stellate cells and enhances the malignant molecular communication with human pancreatic cancer cells. PLoS One. 2015;10:e0128059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
67.  Apte MV, Haber PS, Darby SJ, Rodgers SC, McCaughan GW, Korsten MA, Pirola RC, Wilson JS. Pancreatic stellate cells are activated by proinflammatory cytokines: implications for pancreatic fibrogenesis. Gut. 1999;44:534-541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 433]  [Cited by in F6Publishing: 464]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
68.  Tahara H, Sato K, Yamazaki Y, Ohyama T, Horiguchi N, Hashizume H, Kakizaki S, Takagi H, Ozaki I, Arai H. Transforming growth factor-α activates pancreatic stellate cells and may be involved in matrix metalloproteinase-1 upregulation. Lab Invest. 2013;93:720-732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
69.  Andoh A, Takaya H, Saotome T, Shimada M, Hata K, Araki Y, Nakamura F, Shintani Y, Fujiyama Y, Bamba T. Cytokine regulation of chemokine (IL-8, MCP-1, and RANTES) gene expression in human pancreatic periacinar myofibroblasts. Gastroenterology. 2000;119:211-219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 126]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
70.  Ohnishi N, Miyata T, Ohnishi H, Yasuda H, Tamada K, Ueda N, Mashima H, Sugano K. Activin A is an autocrine activator of rat pancreatic stellate cells: potential therapeutic role of follistatin for pancreatic fibrosis. Gut. 2003;52:1487-1493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 73]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
71.  Hama K, Ohnishi H, Aoki H, Kita H, Yamamoto H, Osawa H, Sato K, Tamada K, Mashima H, Yasuda H. Angiotensin II promotes the proliferation of activated pancreatic stellate cells by Smad7 induction through a protein kinase C pathway. Biochem Biophys Res Commun. 2006;340:742-750.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 35]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
72.  Jaster R, Sparmann G, Emmrich J, Liebe S. Extracellular signal regulated kinases are key mediators of mitogenic signals in rat pancreatic stellate cells. Gut. 2002;51:579-584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 113]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
73.  Masamune A, Kikuta K, Satoh M, Kume K, Shimosegawa T. Differential roles of signaling pathways for proliferation and migration of rat pancreatic stellate cells. Tohoku J Exp Med. 2003;199:69-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 68]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
74.  Klonowski-Stumpe H, Reinehr R, Fischer R, Warskulat U, Lüthen R, Häussinger D. Production and effects of endothelin-1 in rat pancreatic stellate cells. Pancreas. 2003;27:67-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
75.  Masamune A, Satoh M, Kikuta K, Suzuki N, Shimosegawa T. Endothelin-1 stimulates contraction and migration of rat pancreatic stellate cells. World J Gastroenterol. 2005;11:6144-6151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 35]  [Cited by in F6Publishing: 35]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
76.  Uchida M, Ito T, Nakamura T, Igarashi H, Oono T, Fujimori N, Kawabe K, Suzuki K, Jensen RT, Takayanagi R. ERK pathway and sheddases play an essential role in ethanol-induced CX3CL1 release in pancreatic stellate cells. Lab Invest. 2013;93:41-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
77.  Uchida M, Ito T, Nakamura T, Hijioka M, Igarashi H, Oono T, Kato M, Nakamura K, Suzuki K, Takayanagi R. Pancreatic stellate cells and CX3CR1: occurrence in normal pancreas and acute and chronic pancreatitis and effect of their activation by a CX3CR1 agonist. Pancreas. 2014;43:708-719.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
78.  Lee YS, Morinaga H, Kim JJ, Lagakos W, Taylor S, Keshwani M, Perkins G, Dong H, Kayali AG, Sweet IR. The fractalkine/CX3CR1 system regulates β cell function and insulin secretion. Cell. 2013;153:413-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 106]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
79.  Bhatia V, Rastellini C, Han S, Aronson JF, Greeley GH, Falzon M. Acinar cell-specific knockout of the PTHrP gene decreases the proinflammatory and profibrotic responses in pancreatitis. Am J Physiol Gastrointest Liver Physiol. 2014;307:G533-G549.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Mews P, Phillips P, Fahmy R, Korsten M, Pirola R, Wilson J, Apte M. Pancreatic stellate cells respond to inflammatory cytokines: potential role in chronic pancreatitis. Gut. 2002;50:535-541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 284]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
81.  Xue J, Sharma V, Hsieh MH, Chawla A, Murali R, Pandol SJ, Habtezion A. Alternatively activated macrophages promote pancreatic fibrosis in chronic pancreatitis. Nat Commun. 2015;6:7158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 250]  [Article Influence: 27.8]  [Reference Citation Analysis (0)]
82.  Pearson G, Robinson F, Beers Gibson T, Xu BE, Karandikar M, Berman K, Cobb MH. Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev. 2001;22:153-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 297]  [Cited by in F6Publishing: 1318]  [Article Influence: 57.3]  [Reference Citation Analysis (0)]
83.  Masamune A, Satoh M, Kikuta K, Suzuki N, Shimosegawa T. Activation of JAK-STAT pathway is required for platelet-derived growth factor-induced proliferation of pancreatic stellate cells. World J Gastroenterol. 2005;11:3385-3391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
84.  Nishida A, Andoh A, Shioya M, Kim-Mitsuyama S, Takayanagi A, Fujiyama Y. Phosphatidylinositol 3-kinase/Akt signaling mediates interleukin-32alpha induction in human pancreatic periacinar myofibroblasts. Am J Physiol Gastrointest Liver Physiol. 2008;294:G831-G838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
85.  Masamune A, Watanabe T, Kikuta K, Satoh K, Kanno A, Shimosegawa T. Nuclear expression of interleukin-33 in pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2010;299:G821-G832.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 68]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
86.  Nishida A, Andoh A, Imaeda H, Inatomi O, Shiomi H, Fujiyama Y. Expression of interleukin 1-like cytokine interleukin 33 and its receptor complex (ST2L and IL1RAcP) in human pancreatic myofibroblasts. Gut. 2010;59:531-541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 48]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
87.  Kikuta K, Masamune A, Satoh M, Suzuki N, Shimosegawa T. 4-hydroxy-2, 3-nonenal activates activator protein-1 and mitogen-activated protein kinases in rat pancreatic stellate cells. World J Gastroenterol. 2004;10:2344-2351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 28]  [Cited by in F6Publishing: 216]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
88.  McCarroll JA, Phillips PA, Park S, Doherty E, Pirola RC, Wilson JS, Apte MV. Pancreatic stellate cell activation by ethanol and acetaldehyde: is it mediated by the mitogen-activated protein kinase signaling pathway? Pancreas. 2003;27:150-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 61]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
89.  Masamune A, Kikuta K, Suzuki N, Satoh M, Satoh K, Shimosegawa T. A c-Jun NH2-terminal kinase inhibitor SP600125 (anthra[1,9-cd]pyrazole-6 (2H)-one) blocks activation of pancreatic stellate cells. J Pharmacol Exp Ther. 2004;310:520-527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 35]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
90.  Shek FW, Benyon RC, Walker FM, McCrudden PR, Pender SL, Williams EJ, Johnson PA, Johnson CD, Bateman AC, Fine DR. Expression of transforming growth factor-beta 1 by pancreatic stellate cells and its implications for matrix secretion and turnover in chronic pancreatitis. Am J Pathol. 2002;160:1787-1798.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 233]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
91.  Ohnishi H, Miyata T, Yasuda H, Satoh Y, Hanatsuka K, Kita H, Ohashi A, Tamada K, Makita N, Iiri T. Distinct roles of Smad2-, Smad3-, and ERK-dependent pathways in transforming growth factor-beta1 regulation of pancreatic stellate cellular functions. J Biol Chem. 2004;279:8873-8878.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 93]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
92.  Aoki H, Ohnishi H, Hama K, Ishijima T, Satoh Y, Hanatsuka K, Ohashi A, Wada S, Miyata T, Kita H. Autocrine loop between TGF-beta1 and IL-1beta through Smad3- and ERK-dependent pathways in rat pancreatic stellate cells. Am J Physiol Cell Physiol. 2006;290:C1100-C1108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 62]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
93.  Aoki H, Ohnishi H, Hama K, Shinozaki S, Kita H, Yamamoto H, Osawa H, Sato K, Tamada K, Sugano K. Existence of autocrine loop between interleukin-6 and transforming growth factor-beta1 in activated rat pancreatic stellate cells. J Cell Biochem. 2006;99:221-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 59]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
94.  Aoki H, Ohnishi H, Hama K, Shinozaki S, Kita H, Osawa H, Yamamoto H, Sato K, Tamada K, Sugano K. Cyclooxygenase-2 is required for activated pancreatic stellate cells to respond to proinflammatory cytokines. Am J Physiol Cell Physiol. 2007;292:C259-C268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
95.  Lee H, Lim C, Lee J, Kim N, Bang S, Lee H, Min B, Park G, Noda M, Stetler-Stevenson WG. TGF-beta signaling preserves RECK expression in activated pancreatic stellate cells. J Cell Biochem. 2008;104:1065-1074.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 29]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
96.  Hu Y, Wan R, Yu G, Shen J, Ni J, Yin G, Xing M, Chen C, Fan Y, Xiao W. Imbalance of Wnt/Dkk negative feedback promotes persistent activation of pancreatic stellate cells in chronic pancreatitis. PLoS One. 2014;9:e95145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
97.  Xu Y, Li H, Huang C, Zhao T, Zhang H, Zheng C, Ren H, Hao J. Wnt2 protein plays a role in the progression of pancreatic cancer promoted by pancreatic stellate cells. Med Oncol. 2015;32:97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
98.  Shinozaki S, Ohnishi H, Hama K, Kita H, Yamamoto H, Osawa H, Sato K, Tamada K, Mashima H, Sugano K. Indian hedgehog promotes the migration of rat activated pancreatic stellate cells by increasing membrane type-1 matrix metalloproteinase on the plasma membrane. J Cell Physiol. 2008;216:38-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
99.  Bailey JM, Swanson BJ, Hamada T, Eggers JP, Singh PK, Caffery T, Ouellette MM, Hollingsworth MA. Sonic hedgehog promotes desmoplasia in pancreatic cancer. Clin Cancer Res. 2008;14:5995-6004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 370]  [Cited by in F6Publishing: 397]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]
100.  Li X, Wang Z, Ma Q, Xu Q, Liu H, Duan W, Lei J, Ma J, Wang X, Lv S. Sonic hedgehog paracrine signaling activates stromal cells to promote perineural invasion in pancreatic cancer. Clin Cancer Res. 2014;20:4326-4338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 112]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
101.  Han L, Ma J, Duan W, Zhang L, Yu S, Xu Q, Lei J, Li X, Wang Z, Wu Z. Pancreatic stellate cells contribute pancreatic cancer pain via activation of sHH signaling pathway. Oncotarget. 2016;7:18146-18158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 33]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
102.  Masamune A, Nakano E, Hamada S, Takikawa T, Yoshida N, Shimosegawa T. Alteration of the microRNA expression profile during the activation of pancreatic stellate cells. Scand J Gastroenterol. 2014;49:323-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 68]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
103.  Shen J, Wan R, Hu G, Yang L, Xiong J, Wang F, Shen J, He S, Guo X, Ni J. miR-15b and miR-16 induce the apoptosis of rat activated pancreatic stellate cells by targeting Bcl-2 in vitro. Pancreatology. 2012;12:91-99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
104.  Aggarwal BB. Nuclear factor-kappaB: the enemy within. Cancer Cell. 2004;6:203-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1147]  [Cited by in F6Publishing: 1164]  [Article Influence: 58.2]  [Reference Citation Analysis (0)]
105.  Masamune A, Sakai Y, Kikuta K, Satoh M, Satoh A, Shimosegawa T. Activated rat pancreatic stellate cells express intercellular adhesion molecule-1 (ICAM-1) in vitro. Pancreas. 2002;25:78-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 48]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
106.  Shimada M, Andoh A, Hata K, Tasaki K, Araki Y, Fujiyama Y, Bamba T. IL-6 secretion by human pancreatic periacinar myofibroblasts in response to inflammatory mediators. J Immunol. 2002;168:861-868.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 109]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
107.  Masamune A, Kikuta K, Watanabe T, Satoh K, Satoh A, Shimosegawa T. Pancreatic stellate cells express Toll-like receptors. J Gastroenterol. 2008;43:352-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 70]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
108.  Masamune A, Satoh M, Hirabayashi J, Kasai K, Satoh K, Shimosegawa T. Galectin-1 induces chemokine production and proliferation in pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2006;290:G729-G736.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 60]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
109.  Paulo JA, Urrutia R, Banks PA, Conwell DL, Steen H. Proteomic analysis of an immortalized mouse pancreatic stellate cell line identifies differentially-expressed proteins in activated vs nonproliferating cell states. J Proteome Res. 2011;10:4835-4844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 31]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
110.  Paulo JA, Urrutia R, Banks PA, Conwell DL, Steen H. Proteomic analysis of a rat pancreatic stellate cell line using liquid chromatography tandem mass spectrometry (LC-MS/MS). J Proteomics. 2011;75:708-717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
111.  Pan S, Chen R, Stevens T, Bronner MP, May D, Tamura Y, McIntosh MW, Brentnall TA. Proteomics portrait of archival lesions of chronic pancreatitis. PLoS One. 2011;6:e27574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 42]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
112.  Apte MV, Yang L, Phillips PA, Xu Z, Kaplan W, Cowley M, Pirola RC, Wilson JS. Extracellular matrix composition significantly influences pancreatic stellate cell gene expression pattern: role of transgelin in PSC function. Am J Physiol Gastrointest Liver Physiol. 2013;305:G408-G417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
113.  Paulo JA, Kadiyala V, Banks PA, Conwell DL, Steen H. Mass spectrometry-based quantitative proteomic profiling of human pancreatic and hepatic stellate cell lines. Genomics Proteomics Bioinformatics. 2013;11:105-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
114.  Paulo JA, Urrutia R, Kadiyala V, Banks P, Conwell DL, Steen H. Cross-species analysis of nicotine-induced proteomic alterations in pancreatic cells. Proteomics. 2013;13:1499-1512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
115.  Paulo JA, Gaun A, Gygi SP. Global Analysis of Protein Expression and Phosphorylation Levels in Nicotine-Treated Pancreatic Stellate Cells. J Proteome Res. 2015;14:4246-4256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 47]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
116.  Wehr AY, Furth EE, Sangar V, Blair IA, Yu KH. Analysis of the human pancreatic stellate cell secreted proteome. Pancreas. 2011;40:557-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 76]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
117.  Marzoq AJ, Giese N, Hoheisel JD, Alhamdani MS. Proteome variations in pancreatic stellate cells upon stimulation with proinflammatory factors. J Biol Chem. 2013;288:32517-32527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
118.  Apte MV, Xu Z, Pothula S, Goldstein D, Pirola RC, Wilson JS. Pancreatic cancer: The microenvironment needs attention too! Pancreatology. 2015;15:S32-S38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 61]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
119.  Apte MV, Wilson JS. Dangerous liaisons: pancreatic stellate cells and pancreatic cancer cells. J Gastroenterol Hepatol. 2012;27 Suppl 2:69-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 111]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
120.  Gore J, Korc M. Pancreatic cancer stroma: friend or foe? Cancer Cell. 2014;25:711-712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 149]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
121.  Vonlaufen A, Phillips PA, Xu Z, Goldstein D, Pirola RC, Wilson JS, Apte MV. Pancreatic stellate cells and pancreatic cancer cells: an unholy alliance. Cancer Res. 2008;68:7707-7710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 170]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
122.  Chang DZ, Ma Y, Ji B, Wang H, Deng D, Liu Y, Abbruzzese JL, Liu YJ, Logsdon CD, Hwu P. Mast cells in tumor microenvironment promotes the in vivo growth of pancreatic ductal adenocarcinoma. Clin Cancer Res. 2011;17:7015-7023.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 117]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
123.  Chang DZ. Mast cells in pancreatic ductal adenocarcinoma. Oncoimmunology. 2012;1:754-755.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
124.  Ma Y, Hwang RF, Logsdon CD, Ullrich SE. Dynamic mast cell-stromal cell interactions promote growth of pancreatic cancer. Cancer Res. 2013;73:3927-3937.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 145]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
125.  Pandol SJ, Edderkaoui M. What are the macrophages and stellate cells doing in pancreatic adenocarcinoma? Front Physiol. 2015;6:125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
126.  Hu H, Jiao F, Han T, Wang LW. Functional significance of macrophages in pancreatic cancer biology. Tumour Biol. 2015;36:9119-9126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
127.  Tewari N, Zaitoun AM, Arora A, Madhusudan S, Ilyas M, Lobo DN. The presence of tumour-associated lymphocytes confers a good prognosis in pancreatic ductal adenocarcinoma: an immunohistochemical study of tissue microarrays. BMC Cancer. 2013;13:436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 51]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
128.  Stromnes IM, Brockenbrough JS, Izeradjene K, Carlson MA, Cuevas C, Simmons RM, Greenberg PD, Hingorani SR. Targeted depletion of an MDSC subset unmasks pancreatic ductal adenocarcinoma to adaptive immunity. Gut. 2014;63:1769-1781.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 246]  [Article Influence: 24.6]  [Reference Citation Analysis (0)]
129.  Xu XD, Hu J, Wang M, Peng F, Tian R, Guo XJ, Xie Y, Qin RY. Circulating myeloid-derived suppressor cells in patients with pancreatic cancer. Hepatobiliary Pancreat Dis Int. 2016;15:99-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
130.  Korc M. Pancreatic cancer-associated stroma production. Am J Surg. 2007;194:S84-S86.  [PubMed]  [DOI]  [Cited in This Article: ]
131.  Maity G, Mehta S, Haque I, Dhar K, Sarkar S, Banerjee SK, Banerjee S. Pancreatic tumor cell secreted CCN1/Cyr61 promotes endothelial cell migration and aberrant neovascularization. Sci Rep. 2014;4:4995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
132.  Lunardi S, Muschel RJ, Brunner TB. The stromal compartments in pancreatic cancer: are there any therapeutic targets? Cancer Lett. 2014;343:147-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 126]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
133.  Pothula SP, Xu Z, Goldstein D, Pirola RC, Wilson JS, Apte MV. Key role of pancreatic stellate cells in pancreatic cancer. Cancer Lett. 2016;381:194-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 95]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
134.  Patel MB, Pothula SP, Xu Z, Lee AK, Goldstein D, Pirola RC, Apte MV, Wilson JS. The role of the hepatocyte growth factor/c-MET pathway in pancreatic stellate cell-endothelial cell interactions: antiangiogenic implications in pancreatic cancer. Carcinogenesis. 2014;35:1891-1900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 62]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
135.  Pothula SP, Xu Z, Goldstein D, Biankin AV, Pirola RC, Wilson JS, Apte MV. Hepatocyte growth factor inhibition: a novel therapeutic approach in pancreatic cancer. Br J Cancer. 2016;114:269-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 69]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
136.  Hwang RF, Moore T, Arumugam T, Ramachandran V, Amos KD, Rivera A, Ji B, Evans DB, Logsdon CD. Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res. 2008;68:918-926.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 794]  [Cited by in F6Publishing: 897]  [Article Influence: 56.1]  [Reference Citation Analysis (0)]
137.  Kikuta K, Masamune A, Watanabe T, Ariga H, Itoh H, Hamada S, Satoh K, Egawa S, Unno M, Shimosegawa T. Pancreatic stellate cells promote epithelial-mesenchymal transition in pancreatic cancer cells. Biochem Biophys Res Commun. 2010;403:380-384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 199]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
138.  Fujiwara K, Ohuchida K, Ohtsuka T, Mizumoto K, Shindo K, Ikenaga N, Cui L, Takahata S, Aishima S, Tanaka M. Migratory activity of CD105+ pancreatic cancer cells is strongly enhanced by pancreatic stellate cells. Pancreas. 2013;42:1283-1290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
139.  Hamada S, Masamune A, Takikawa T, Suzuki N, Kikuta K, Hirota M, Hamada H, Kobune M, Satoh K, Shimosegawa T. Pancreatic stellate cells enhance stem cell-like phenotypes in pancreatic cancer cells. Biochem Biophys Res Commun. 2012;421:349-354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 155]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
140.  Ikenaga N, Ohuchida K, Mizumoto K, Cui L, Kayashima T, Morimatsu K, Moriyama T, Nakata K, Fujita H, Tanaka M. CD10+ pancreatic stellate cells enhance the progression of pancreatic cancer. Gastroenterology. 2010;139:1041-1051, 1051.e1-8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 144]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
141.  Arumugam T, Brandt W, Ramachandran V, Moore TT, Wang H, May FE, Westley BR, Hwang RF, Logsdon CD. Trefoil factor 1 stimulates both pancreatic cancer and stellate cells and increases metastasis. Pancreas. 2011;40:815-822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 50]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
142.  Koshiba T, Hosotani R, Miyamoto Y, Wada M, Lee JU, Fujimoto K, Tsuji S, Nakajima S, Doi R, Imamura M. Immunohistochemical analysis of cyclooxygenase-2 expression in pancreatic tumors. Int J Pancreatol. 1999;26:69-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 55]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
143.  Tucker ON, Dannenberg AJ, Yang EK, Zhang F, Teng L, Daly JM, Soslow RA, Masferrer JL, Woerner BM, Koki AT. Cyclooxygenase-2 expression is up-regulated in human pancreatic cancer. Cancer Res. 1999;59:987-990.  [PubMed]  [DOI]  [Cited in This Article: ]
144.  Kokawa A, Kondo H, Gotoda T, Ono H, Saito D, Nakadaira S, Kosuge T, Yoshida S. Increased expression of cyclooxygenase-2 in human pancreatic neoplasms and potential for chemoprevention by cyclooxygenase inhibitors. Cancer. 2001;91:333-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
145.  Merati K, said Siadaty M, Andea A, Sarkar F, Ben-Josef E, Mohammad R, Philip P, Shields AF, Vaitkevicius V, Grignon DJ. Expression of inflammatory modulator COX-2 in pancreatic ductal adenocarcinoma and its relationship to pathologic and clinical parameters. Am J Clin Oncol. 2001;24:447-452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 65]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
146.  Jiang HB, Xu M, Wang XP. Pancreatic stellate cells promote proliferation and invasiveness of human pancreatic cancer cells via galectin-3. World J Gastroenterol. 2008;14:2023-2028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
147.  Tang D, Yuan Z, Xue X, Lu Z, Zhang Y, Wang H, Chen M, An Y, Wei J, Zhu Y. High expression of Galectin-1 in pancreatic stellate cells plays a role in the development and maintenance of an immunosuppressive microenvironment in pancreatic cancer. Int J Cancer. 2012;130:2337-2348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 119]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
148.  Farrow B, Berger DH, Rowley D. Tumor-derived pancreatic stellate cells promote pancreatic cancer cell invasion through release of thrombospondin-2. J Surg Res. 2009;156:155-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
149.  Gao Z, Wang X, Wu K, Zhao Y, Hu G. Pancreatic stellate cells increase the invasion of human pancreatic cancer cells through the stromal cell-derived factor-1/CXCR4 axis. Pancreatology. 2010;10:186-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 87]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
150.  Zhou Y, Zhou Q, Chen R. Pancreatic stellate cells promotes the perineural invasion in pancreatic cancer. Med Hypotheses. 2012;78:811-813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
151.  Buchholz M, Biebl A, Neesse A, Wagner M, Iwamura T, Leder G, Adler G, Gress TM. SERPINE2 (protease nexin I) promotes extracellular matrix production and local invasion of pancreatic tumors in vivo. Cancer Res. 2003;63:4945-4951.  [PubMed]  [DOI]  [Cited in This Article: ]
152.  Neesse A, Wagner M, Ellenrieder V, Bachem M, Gress TM, Buchholz M. Pancreatic stellate cells potentiate proinvasive effects of SERPINE2 expression in pancreatic cancer xenograft tumors. Pancreatology. 2007;7:380-385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
153.  Tian X, Chen G, Zhou S, Henne-Bruns D, Bachem M, Kornmann M. Interactions of pancreatic cancer and stellate cells are mediated by FGFR1-III isoform expression. Hepatogastroenterology. 2012;59:1604-1608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 9]  [Reference Citation Analysis (0)]
154.  Coleman SJ, Chioni AM, Ghallab M, Anderson RK, Lemoine NR, Kocher HM, Grose RP. Nuclear translocation of FGFR1 and FGF2 in pancreatic stellate cells facilitates pancreatic cancer cell invasion. EMBO Mol Med. 2014;6:467-481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 128]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
155.  Xu Z, Vonlaufen A, Phillips PA, Fiala-Beer E, Zhang X, Yang L, Biankin AV, Goldstein D, Pirola RC, Wilson JS. Role of pancreatic stellate cells in pancreatic cancer metastasis. Am J Pathol. 2010;177:2585-2596.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 291]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
156.  Rebours V, Albuquerque M, Sauvanet A, Ruszniewski P, Lévy P, Paradis V, Bedossa P, Couvelard A. Hypoxia pathways and cellular stress activate pancreatic stellate cells: development of an organotypic culture model of thick slices of normal human pancreas. PLoS One. 2013;8:e76229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
157.  Masamune A, Kikuta K, Watanabe T, Satoh K, Hirota M, Shimosegawa T. Hypoxia stimulates pancreatic stellate cells to induce fibrosis and angiogenesis in pancreatic cancer. Am J Physiol Gastrointest Liver Physiol. 2008;295:G709-G717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 190]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
158.  Erkan M, Reiser-Erkan C, Michalski CW, Deucker S, Sauliunaite D, Streit S, Esposito I, Friess H, Kleeff J. Cancer-stellate cell interactions perpetuate the hypoxia-fibrosis cycle in pancreatic ductal adenocarcinoma. Neoplasia. 2009;11:497-508.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 223]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
159.  Sada M, Ohuchida K, Horioka K, Okumura T, Moriyama T, Miyasaka Y, Ohtsuka T, Mizumoto K, Oda Y, Nakamura M. Hypoxic stellate cells of pancreatic cancer stroma regulate extracellular matrix fiber organization and cancer cell motility. Cancer Lett. 2016;372:210-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 61]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
160.  Erkan M, Kleeff J, Gorbachevski A, Reiser C, Mitkus T, Esposito I, Giese T, Büchler MW, Giese NA, Friess H. Periostin creates a tumor-supportive microenvironment in the pancreas by sustaining fibrogenic stellate cell activity. Gastroenterology. 2007;132:1447-1464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 240]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
161.  Takikawa T, Masamune A, Hamada S, Nakano E, Yoshida N, Shimosegawa T. miR-210 regulates the interaction between pancreatic cancer cells and stellate cells. Biochem Biophys Res Commun. 2013;437:433-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 65]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
162.  Kadera BE, Li L, Toste PA, Wu N, Adams C, Dawson DW, Donahue TR. MicroRNA-21 in pancreatic ductal adenocarcinoma tumor-associated fibroblasts promotes metastasis. PLoS One. 2013;8:e71978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 124]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
163.  Kuninty PR, Bojmar L, Tjomsland V, Larsson M, Storm G, Östman A, Sandström P, Prakash J. MicroRNA-199a and -214 as potential therapeutic targets in pancreatic stellate cells in pancreatic tumor. Oncotarget. 2016;7:16396-16408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 59]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
164.  Kwon JJ, Nabinger SC, Vega Z, Sahu SS, Alluri RK, Abdul-Sater Z, Yu Z, Gore J, Nalepa G, Saxena R. Pathophysiological role of microRNA-29 in pancreatic cancer stroma. Sci Rep. 2015;5:11450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 50]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
165.  Ene-Obong A, Clear AJ, Watt J, Wang J, Fatah R, Riches JC, Marshall JF, Chin-Aleong J, Chelala C, Gribben JG. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology. 2013;145:1121-1132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 340]  [Cited by in F6Publishing: 399]  [Article Influence: 36.3]  [Reference Citation Analysis (0)]
166.  Tang D, Gao J, Wang S, Yuan Z, Ye N, Chong Y, Xu C, Jiang X, Li B, Yin W. Apoptosis and anergy of T cell induced by pancreatic stellate cells-derived galectin-1 in pancreatic cancer. Tumour Biol. 2015;36:5617-5626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 45]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
167.  Shi C, Washington MK, Chaturvedi R, Drosos Y, Revetta FL, Weaver CJ, Buzhardt E, Yull FE, Blackwell TS, Sosa-Pineda B. Fibrogenesis in pancreatic cancer is a dynamic process regulated by macrophage-stellate cell interaction. Lab Invest. 2014;94:409-421.  [PubMed]  [DOI]  [Cited in This Article: ]
168.  Kraman M, Bambrough PJ, Arnold JN, Roberts EW, Magiera L, Jones JO, Gopinathan A, Tuveson DA, Fearon DT. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science. 2010;330:827-830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 752]  [Cited by in F6Publishing: 884]  [Article Influence: 63.1]  [Reference Citation Analysis (0)]
169.  Nakamura F, Shintani Y, Saotome T, Fujiyama Y, Bamba T. Effects of synthetic serine protease inhibitors on proliferation and collagen synthesis of human pancreatic periacinar fibroblast-like cells. Pancreas. 2001;22:317-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
170.  Xie MJ, Motoo Y, Su SB, Iovanna JL, Sawabu N. Effect of carboxamide derivative IS-741 on rat spontaneous chronic pancreatitis. Dig Dis Sci. 2002;47:139-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
171.  Kuno A, Yamada T, Masuda K, Ogawa K, Sogawa M, Nakamura S, Nakazawa T, Ohara H, Nomura T, Joh T. Angiotensin-converting enzyme inhibitor attenuates pancreatic inflammation and fibrosis in male Wistar Bonn/Kobori rats. Gastroenterology. 2003;124:1010-1019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 95]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
172.  Yamada T, Kuno A, Masuda K, Ogawa K, Sogawa M, Nakamura S, Ando T, Sano H, Nakazawa T, Ohara H. Candesartan, an angiotensin II receptor antagonist, suppresses pancreatic inflammation and fibrosis in rats. J Pharmacol Exp Ther. 2003;307:17-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 53]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
173.  Masamune A, Kikuta K, Satoh M, Satoh K, Shimosegawa T. Rho kinase inhibitors block activation of pancreatic stellate cells. Br J Pharmacol. 2003;140:1292-1302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 68]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
174.  Nagashio Y, Ueno H, Imamura M, Asaumi H, Watanabe S, Yamaguchi T, Taguchi M, Tashiro M, Otsuki M. Inhibition of transforming growth factor beta decreases pancreatic fibrosis and protects the pancreas against chronic injury in mice. Lab Invest. 2004;84:1610-1618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 60]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
175.  Zhao HF, Ito T, Gibo J, Kawabe K, Oono T, Kaku T, Arita Y, Zhao QW, Usui M, Egashira K. Anti-monocyte chemoattractant protein 1 gene therapy attenuates experimental chronic pancreatitis induced by dibutyltin dichloride in rats. Gut. 2005;54:1759-1767.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 49]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
176.  Gibo J, Ito T, Kawabe K, Hisano T, Inoue M, Fujimori N, Oono T, Arita Y, Nawata H. Camostat mesilate attenuates pancreatic fibrosis via inhibition of monocytes and pancreatic stellate cells activity. Lab Invest. 2005;85:75-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 68]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
177.  Yamada T, Kuno A, Ogawa K, Tang M, Masuda K, Nakamura S, Ando T, Okamoto T, Ohara H, Nomura T. Combination therapy with an angiotensin-converting enzyme inhibitor and an angiotensin II receptor blocker synergistically suppresses chronic pancreatitis in rats. J Pharmacol Exp Ther. 2005;313:36-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
178.  van Westerloo DJ, Florquin S, de Boer AM, Daalhuisen J, de Vos AF, Bruno MJ, van der Poll T. Therapeutic effects of troglitazone in experimental chronic pancreatitis in mice. Am J Pathol. 2005;166:721-728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 59]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
179.  Reding T, Bimmler D, Perren A, Sun LK, Fortunato F, Storni F, Graf R. A selective COX-2 inhibitor suppresses chronic pancreatitis in an animal model (WBN/Kob rats): significant reduction of macrophage infiltration and fibrosis. Gut. 2006;55:1165-1173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 44]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
180.  Asaumi H, Watanabe S, Taguchi M, Tashiro M, Nagashio Y, Nomiyama Y, Nakamura H, Otsuki M. Green tea polyphenol (-)-epigallocatechin-3-gallate inhibits ethanol-induced activation of pancreatic stellate cells. Eur J Clin Invest. 2006;36:113-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
181.  Baumert JT, Sparmann G, Emmrich J, Liebe S, Jaster R. Inhibitory effects of interferons on pancreatic stellate cell activation. World J Gastroenterol. 2006;12:896-901.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 23]  [Cited by in F6Publishing: 22]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
182.  Fitzner B, Brock P, Nechutova H, Glass A, Karopka T, Koczan D, Thiesen HJ, Sparmann G, Emmrich J, Liebe S. Inhibitory effects of interferon-gamma on activation of rat pancreatic stellate cells are mediated by STAT1 and involve down-regulation of CTGF expression. Cell Signal. 2007;19:782-790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 23]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
183.  Ohashi S, Nishio A, Nakamura H, Asada M, Tamaki H, Kawasaki K, Fukui T, Yodoi J, Chiba T. Overexpression of redox-active protein thioredoxin-1 prevents development of chronic pancreatitis in mice. Antioxid Redox Signal. 2006;8:1835-1845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
184.  McCarroll JA, Phillips PA, Santucci N, Pirola RC, Wilson JS, Apte MV. Vitamin A inhibits pancreatic stellate cell activation: implications for treatment of pancreatic fibrosis. Gut. 2006;55:79-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 117]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
185.  Tasci I, Deveci S, Isik AT, Comert B, Akay C, Mas N, Inal V, Yamanel L, Mas MR. Allopurinol in rat chronic pancreatitis: effects on pancreatic stellate cell activation. Pancreas. 2007;35:366-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
186.  Lu XL, Song YH, Fu YB, Si JM, Qian KD. Ascorbic acid alleviates pancreatic damage induced by dibutyltin dichloride (DBTC) in rats. Yonsei Med J. 2007;48:1028-1034.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
187.  Shirahige A, Mizushima T, Matsushita K, Sawa K, Ochi K, Ichimura M, Tanioka H, Shinji T, Koide N, Tanimoto M. Oral administration of taurine improves experimental pancreatic fibrosis. J Gastroenterol Hepatol. 2008;23:321-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
188.  Rickmann M, Vaquero EC, Malagelada JR, Molero X. Tocotrienols induce apoptosis and autophagy in rat pancreatic stellate cells through the mitochondrial death pathway. Gastroenterology. 2007;132:2518-2532.  [PubMed]  [DOI]  [Cited in This Article: ]
189.  Michalski CW, Maier M, Erkan M, Sauliunaite D, Bergmann F, Pacher P, Batkai S, Giese NA, Giese T, Friess H. Cannabinoids reduce markers of inflammation and fibrosis in pancreatic stellate cells. PLoS One. 2008;3:e1701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 43]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
190.  Weylandt KH, Nadolny A, Kahlke L, Köhnke T, Schmöcker C, Wang J, Lauwers GY, Glickman JN, Kang JX. Reduction of inflammation and chronic tissue damage by omega-3 fatty acids in fat-1 transgenic mice with pancreatitis. Biochim Biophys Acta. 2008;1782:634-641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 48]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
191.  Karatas A, Paksoy M, Erzin Y, Carkman S, Gonenc M, Ayan F, Aydogan F, Uzun H, Durak H. The effect of halofuginone, a specific inhibitor of collagen type 1 synthesis, in the prevention of pancreatic fibrosis in an experimental model of severe hyperstimulation and obstruction pancreatitis. J Surg Res. 2008;148:7-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
192.  Fitzner B, Brock P, Holzhüter SA, Nizze H, Sparmann G, Emmrich J, Liebe S, Jaster R. Synergistic growth inhibitory effects of the dual endothelin-1 receptor antagonist bosentan on pancreatic stellate and cancer cells. Dig Dis Sci. 2009;54:309-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
193.  Schwer CI, Mutschler M, Stoll P, Goebel U, Humar M, Hoetzel A, Schmidt R. Carbon monoxide releasing molecule-2 inhibits pancreatic stellate cell proliferation by activating p38 mitogen-activated protein kinase/heme oxygenase-1 signaling. Mol Pharmacol. 2010;77:660-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
194.  Nathan JD, Romac J, Peng RY, Peyton M, Rockey DC, Liddle RA. Protection against chronic pancreatitis and pancreatic fibrosis in mice overexpressing pancreatic secretory trypsin inhibitor. Pancreas. 2010;39:e24-e30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 16]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
195.  González AM, Garcia T, Samper E, Rickmann M, Vaquero EC, Molero X. Assessment of the protective effects of oral tocotrienols in arginine chronic-like pancreatitis. Am J Physiol Gastrointest Liver Physiol. 2011;301:G846-G855.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
196.  Long D, Lu J, Luo L, Guo Y, Li C, Wu W, Shan J, Li L, Li S, Li Y. Effects of octreotide on activated pancreatic stellate cell-induced pancreas graft fibrosis in rats. J Surg Res. 2012;176:248-259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
197.  Li L, Bimmler D, Graf R, Zhou S, Sun Z, Chen J, Siech M, Bachem MG. PSP/reg inhibits cultured pancreatic stellate cell and regulates MMP/ TIMP ratio. Eur J Clin Invest. 2011;41:151-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
198.  Tang Y, Liao Y, Kawaguchi-Sakita N, Raut V, Fakhrejahani E, Qian N, Toi M. Sinisan, a traditional Chinese medicine, attenuates experimental chronic pancreatitis induced by trinitrobenzene sulfonic acid in rats. J Hepatobiliary Pancreat Sci. 2011;18:551-558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
199.  Wei L, Yamamoto M, Harada M, Otsuki M. Treatment with pravastatin attenuates progression of chronic pancreatitis in rat. Lab Invest. 2011;91:872-884.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
200.  Li XC, Lu XL, Chen HH. α-Tocopherol treatment ameliorates chronic pancreatitis in an experimental rat model induced by trinitrobenzene sulfonic acid. Pancreatology. 2011;11:5-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
201.  Monteiro TH, Silva CS, Cordeiro Simões Ambrosio LM, Zucoloto S, Vannucchi H. Vitamin E alters inflammatory gene expression in alcoholic chronic pancreatitis. J Nutrigenet Nutrigenomics. 2012;5:94-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
202.  Matsushita K, Mizushima T, Shirahige A, Tanioka H, Sawa K, Ochi K, Tanimoto M, Koide N. Effect of taurine on acinar cell apoptosis and pancreatic fibrosis in dibutyltin dichloride-induced chronic pancreatitis. Acta Med Okayama. 2012;66:329-334.  [PubMed]  [DOI]  [Cited in This Article: ]
203.  Yang L, Shen J, He S, Hu G, Shen J, Wang F, Xu L, Dai W, Xiong J, Ni J. L-cysteine administration attenuates pancreatic fibrosis induced by TNBS in rats by inhibiting the activation of pancreatic stellate cell. PLoS One. 2012;7:e31807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 17]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
204.  Bai H, Chen X, Zhang L, Dou X. The effect of sulindac, a non-steroidal anti-inflammatory drug, attenuates inflammation and fibrosis in a mouse model of chronic pancreatitis. BMC Gastroenterol. 2012;12:115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 22]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
205.  Lee BJ, Lee HS, Kim CD, Jung SW, Seo YS, Kim YS, Jeen YT, Chun HJ, Um SH, Lee SW. The Effects of Combined Treatment with an HMG-CoA Reductase Inhibitor and PPARγ Agonist on the Activation of Rat Pancreatic Stellate Cells. Gut Liver. 2012;6:262-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
206.  Shen J, Gao J, Chen C, Lu H, Hu G, Shen J, Zhu S, Wu M, Wang X, Qian L. Antifibrotic role of chemokine CXCL9 in experimental chronic pancreatitis induced by trinitrobenzene sulfonic acid in rats. Cytokine. 2013;64:382-394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
207.  Gao X, Cao Y, Yang W, Duan C, Aronson JF, Rastellini C, Chao C, Hellmich MR, Ko TC. BMP2 inhibits TGF-β-induced pancreatic stellate cell activation and extracellular matrix formation. Am J Physiol Gastrointest Liver Physiol. 2013;304:G804-G813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 54]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
208.  Zhou CH, Lin-Li XY, Wen-Tang DM, Dong Y, Li LY, Wang SF. Protective effects of edaravone on experimental chronic pancreatitis induced by dibutyltin dichloride in rats. Pancreatology. 2013;13:125-132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
209.  Niina Y, Ito T, Oono T, Nakamura T, Fujimori N, Igarashi H, Sakai Y, Takayanagi R. A sustained prostacyclin analog, ONO-1301, attenuates pancreatic fibrosis in experimental chronic pancreatitis induced by dibutyltin dichloride in rats. Pancreatology. 2014;14:201-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 13]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
210.  Mrazek AA, Porro LJ, Bhatia V, Falzon M, Spratt H, Zhou J, Chao C, Hellmich MR. Apigenin inhibits pancreatic stellate cell activity in pancreatitis. J Surg Res. 2015;196:8-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
211.  Tsang SW, Zhang H, Lin Z, Mu H, Bian ZX. Anti-fibrotic effect of trans-resveratrol on pancreatic stellate cells. Biomed Pharmacother. 2015;71:91-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
212.  Lin Z, Zheng LC, Zhang HJ, Tsang SW, Bian ZX. Anti-fibrotic effects of phenolic compounds on pancreatic stellate cells. BMC Complement Altern Med. 2015;15:259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 21]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
213.  Gundewar C, Ansari D, Tang L, Wang Y, Liang G, Rosendahl AH, Saleem MA, Andersson R. Antiproliferative effects of curcumin analog L49H37 in pancreatic stellate cells: a comparative study. Ann Gastroenterol. 2015;28:391-398.  [PubMed]  [DOI]  [Cited in This Article: ]
214.  Tsang SW, Zhang HJ, Chen YG, Auyeung KK, Bian ZX. Eruberin A, a Natural Flavanol Glycoside, Exerts Anti-Fibrotic Action on Pancreatic Stellate Cells. Cell Physiol Biochem. 2015;36:2433-2446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
215.  Bläuer M, Sand J, Laukkarinen J. Physiological and clinically attainable concentrations of 1,25-dihydroxyvitamin D3 suppress proliferation and extracellular matrix protein expression in mouse pancreatic stellate cells. Pancreatology. 2015;15:366-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
216.  Xiao W, Jiang W, Shen J, Yin G, Fan Y, Wu D, Qiu L, Yu G, Xing M, Hu G. Retinoic Acid Ameliorates Pancreatic Fibrosis and Inhibits the Activation of Pancreatic Stellate Cells in Mice with Experimental Chronic Pancreatitis via Suppressing the Wnt/β-Catenin Signaling Pathway. PLoS One. 2015;10:e0141462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 44]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
217.  Witteck L, Jaster R. Trametinib and dactolisib but not regorafenib exert antiproliferative effects on rat pancreatic stellate cells. Hepatobiliary Pancreat Dis Int. 2015;14:642-650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
218.  Ulmasov B, Neuschwander-Tetri BA, Lai J, Monastyrskiy V, Trisha Bhat, Yates MP, Oliva J, Prinsen MJ, Ruminski PG, Griggs DW. Inhibitors of Arg-Gly-Asp-Binding Integrins Reduce Development of Pancreatic Fibrosis in Mice. Cell Mol Gastroenterol Hepatol. 2016;2:499-518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
219.  Feldmann G, Dhara S, Fendrich V, Bedja D, Beaty R, Mullendore M, Karikari C, Alvarez H, Iacobuzio-Donahue C, Jimeno A. Blockade of hedgehog signaling inhibits pancreatic cancer invasion and metastases: a new paradigm for combination therapy in solid cancers. Cancer Res. 2007;67:2187-2196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 515]  [Cited by in F6Publishing: 516]  [Article Influence: 30.4]  [Reference Citation Analysis (0)]
220.  Diep CH, Munoz RM, Choudhary A, Von Hoff DD, Han H. Synergistic effect between erlotinib and MEK inhibitors in KRAS wild-type human pancreatic cancer cells. Clin Cancer Res. 2011;17:2744-2756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 60]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
221.  Froeling FE, Feig C, Chelala C, Dobson R, Mein CE, Tuveson DA, Clevers H, Hart IR, Kocher HM. Retinoic acid-induced pancreatic stellate cell quiescence reduces paracrine Wnt-β-catenin signaling to slow tumor progression. Gastroenterology. 2011;141:1486-1497, 1497.e1-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 326]  [Article Influence: 25.1]  [Reference Citation Analysis (0)]
222.  Chauhan VP, Martin JD, Liu H, Lacorre DA, Jain SR, Kozin SV, Stylianopoulos T, Mousa AS, Han X, Adstamongkonkul P. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat Commun. 2013;4:2516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 674]  [Cited by in F6Publishing: 720]  [Article Influence: 65.5]  [Reference Citation Analysis (0)]
223.  Sun XD, Liu XE, Huang DS. Curcumin reverses the epithelial-mesenchymal transition of pancreatic cancer cells by inhibiting the Hedgehog signaling pathway. Oncol Rep. 2013;29:2401-2407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 78]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
224.  Edderkaoui M, Lugea A, Hui H, Eibl G, Lu QY, Moro A, Lu X, Li G, Go VL, Pandol SJ. Ellagic acid and embelin affect key cellular components of pancreatic adenocarcinoma, cancer, and stellate cells. Nutr Cancer. 2013;65:1232-1244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 41]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
225.  Macha MA, Rachagani S, Gupta S, Pai P, Ponnusamy MP, Batra SK, Jain M. Guggulsterone decreases proliferation and metastatic behavior of pancreatic cancer cells by modulating JAK/STAT and Src/FAK signaling. Cancer Lett. 2013;341:166-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 66]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
226.  Kozono S, Ohuchida K, Eguchi D, Ikenaga N, Fujiwara K, Cui L, Mizumoto K, Tanaka M. Pirfenidone inhibits pancreatic cancer desmoplasia by regulating stellate cells. Cancer Res. 2013;73:2345-2356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 153]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
227.  Guan J, Zhang H, Wen Z, Gu Y, Cheng Y, Sun Y, Zhang T, Jia C, Lu Z, Chen J. Retinoic acid inhibits pancreatic cancer cell migration and EMT through the downregulation of IL-6 in cancer associated fibroblast cells. Cancer Lett. 2014;345:132-139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 90]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
228.  Gonzalez-Villasana V, Rodriguez-Aguayo C, Arumugam T, Cruz-Monserrate Z, Fuentes-Mattei E, Deng D, Hwang RF, Wang H, Ivan C, Garza RJ. Bisphosphonates inhibit stellate cell activity and enhance antitumor effects of nanoparticle albumin-bound paclitaxel in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 2014;13:2583-2594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 169]  [Reference Citation Analysis (0)]
229.  Pomianowska E, Sandnes D, Grzyb K, Schjølberg AR, Aasrum M, Tveteraas IH, Tjomsland V, Christoffersen T, Gladhaug IP. Inhibitory effects of prostaglandin E2 on collagen synthesis and cell proliferation in human stellate cells from pancreatic head adenocarcinoma. BMC Cancer. 2014;14:413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 22]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
230.  Gong J, Xie J, Bedolla R, Rivas P, Chakravarthy D, Freeman JW, Reddick R, Kopetz S, Peterson A, Wang H. Combined targeting of STAT3/NF-κB/COX-2/EP4 for effective management of pancreatic cancer. Clin Cancer Res. 2014;20:1259-1273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 60]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
231.  Yan HH, Jung KH, Son MK, Fang Z, Kim SJ, Ryu YL, Kim J, Kim MH, Hong SS. Crizotinib exhibits antitumor activity by targeting ALK signaling not c-MET in pancreatic cancer. Oncotarget. 2014;5:9150-9168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
232.  Zhang H, Zhou WC, Li X, Meng WB, Zhang L, Zhu XL, Zhu KX, Bai ZT, Yan J, Liu T. 5-Azacytidine suppresses the proliferation of pancreatic cancer cells by inhibiting the Wnt/β-catenin signaling pathway. Genet Mol Res. 2014;13:5064-5072.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 53]  [Reference Citation Analysis (0)]
233.  Wang S, Chen X, Tang M. MicroRNA-216a inhibits pancreatic cancer by directly targeting Janus kinase 2. Oncol Rep. 2014;32:2824-2830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 53]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
234.  Kumar V, Mondal G, Slavik P, Rachagani S, Batra SK, Mahato RI. Codelivery of small molecule hedgehog inhibitor and miRNA for treating pancreatic cancer. Mol Pharm. 2015;12:1289-1298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 67]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
235.  Petrova E, Matevossian A, Resh MD. Hedgehog acyltransferase as a target in pancreatic ductal adenocarcinoma. Oncogene. 2015;34:263-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 45]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
236.  Massó-Vallés D, Jauset T, Serrano E, Sodir NM, Pedersen K, Affara NI, Whitfield JR, Beaulieu ME, Evan GI, Elias L. Ibrutinib exerts potent antifibrotic and antitumor activities in mouse models of pancreatic adenocarcinoma. Cancer Res. 2015;75:1675-1681.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 91]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
237.  Zhou GX, Ding XL, Wu SB, Zhang HF, Cao W, Qu LS, Zhang H. Inhibition of 5-lipoxygenase triggers apoptosis in pancreatic cancer cells. Oncol Rep. 2015;33:661-668.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 32]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
238.  Lui GY, Kovacevic Z, V Menezes S, Kalinowski DS, Merlot AM, Sahni S, Richardson DR. Novel thiosemicarbazones regulate the signal transducer and activator of transcription 3 (STAT3) pathway: inhibition of constitutive and interleukin 6-induced activation by iron depletion. Mol Pharmacol. 2015;87:543-560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 34]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
239.  Khan S, Ebeling MC, Chauhan N, Thompson PA, Gara RK, Ganju A, Yallapu MM, Behrman SW, Zhao H, Zafar N. Ormeloxifene suppresses desmoplasia and enhances sensitivity of gemcitabine in pancreatic cancer. Cancer Res. 2015;75:2292-2304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 61]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
240.  Liu QQ, Chen K, Ye Q, Jiang XH, Sun YW. Oridonin inhibits pancreatic cancer cell migration and epithelial-mesenchymal transition by suppressing Wnt/β-catenin signaling pathway. Cancer Cell Int. 2016;16:57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 34]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
241.  Haqq J, Howells LM, Garcea G, Dennison AR. Targeting pancreatic cancer using a combination of gemcitabine with the omega-3 polyunsaturated fatty acid emulsion, Lipidem™. Mol Nutr Food Res. 2016;60:1437-1447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
242.  Ji T, Li S, Zhang Y, Lang J, Ding Y, Zhao X, Zhao R, Li Y, Shi J, Hao J. An MMP-2 Responsive Liposome Integrating Antifibrosis and Chemotherapeutic Drugs for Enhanced Drug Perfusion and Efficacy in Pancreatic Cancer. ACS Appl Mater Interfaces. 2016;8:3438-3445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 103]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
243.  Delitto D, Wallet SM, Hughes SJ. Targeting tumor tolerance: A new hope for pancreatic cancer therapy? Pharmacol Ther. 2016;166:9-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 29]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
244.  Heinemann V, Reni M, Ychou M, Richel DJ, Macarulla T, Ducreux M. Tumour-stroma interactions in pancreatic ductal adenocarcinoma: rationale and current evidence for new therapeutic strategies. Cancer Treat Rev. 2014;40:118-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 90]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]