Pancreatic stellate cell

Last updated
Pancreatic stellate cell
Details
Location Pancreas
Identifiers
MeSH D058954
Anatomical terms of microanatomy

Pancreatic stellate cells (PaSCs) are classified as myofibroblast-like cells that are located in exocrine regions of the pancreas. [1] PaSCs are mediated by paracrine and autocrine stimuli and share similarities with the hepatic stellate cell. [1] Pancreatic stellate cell activation and expression of matrix molecules constitute the complex process that induces pancreatic fibrosis. [2] Synthesis, deposition, maturation and remodelling of the fibrous connective tissue can be protective, however when persistent it impedes regular pancreatic function. [2]

Contents

Structure

PaSCs are located within the peri-acinar spaces of the pancreas and extrude long cytoplasmic processes that surround the base of the acinus. [1] PaSCs compose 4% of the total cell mass in the gland [3] Stellate cells derive their name from their star shape and are located in other organs such as the kidney and lungs. [1] The cells are located in periductal and perivascular regions of the pancreas and contain vitamin A lipid droplets in their cytoplasm. [1] PaSCs engage in disease pathogenesis by transforming from a quiescent state into an activated state, which is also known as a “myofibroblastic” state. [1]

PaSCs express the intermediate filament proteins desmin and glial fibrillary acidic protein. [1] The expression of a diverse range of intermediate filament proteins enables the PaSC to harbour contractile abilities. [1] Cellular extensions also enable the cells to sense their environment. [1] Following inflammation or injury to the pancreas, quiescent PaSCs are activated to myofibroblast like cells, which expresses α- smooth muscle actin. [1] Several morphological changes take place including nuclear enlargement and increased growth of the endoplasmic reticulum network. [1] The activated PaSCs then grow in number, migrate and secrete extracellular matrix components such as type I collagen, chemokines and cytokines. [1]

Function

Quiescent PaSCs produce metalloproteinases such as MMP-2, MMP-9, and MMP-13 and their inhibitors, which assist in the turnover of the extracellular matrix (ECM). [4] As a result of regulating ECM turnover, PaSCs are involved in the maintenance of the modelling of normal tissue. [4] MMP-2 secreted by PaSCs, however, contributes to the development of pancreatic cancer. [5]

Fibrosis is a prominent feature of chronic pancreatitis and of the desmoplastic reaction linked with pancreatic cancer. [6] While the pathogenesis of fibrosis remains elusive, the activation of stellate cells contribute to pancreatic fibrosis. [7] Numerous soluble factors regulate PaSC activation, specifically IL-1, IL-6, TNF-α, TGF-B1 and activin 1. [5] The potential sources of these activating factors include platelets, macrophages, pancreatic acinar cells and endothelial cells in inflamed pancreas. [5] PaSCs, individually, are also capable of synthesising cytokines such as TGF-β1, activin A and IL-1. [5] The production of these factors indicates the presence of autocrine loops that perpetuate PaSC activation, promoting the development of fibrosis. [5]

Protein kinases such as MAPKs are primary mediators of activating signals initiated by the growth factors, angiotensin II and ethanol. [1] Other signalling pathways regulating PaSC activation include PI3K, RHO kinase and TGF-β/SMAD-related pathways. [1] Following activation, PaSCs migrate to areas of tissue damage and contract, phagocytose, and induce products that regulate the ECM through facilitating repair or by promoting fibrosis. [1] The migration of PaSCs is modulated by Indian hedgehog (IHH), a peptide that is involved in pancreatic development, patterning and differentiation. [8] Stellate cells express smoothened (Smo) and patched-1 (Ptch1) proteins, which are significant features of the hedgehog receptor system. [9] Indian Hedgehog binding results in relocation of the transcriptional of transcription factor Gli-1 into the nucleus, inducing chemokinetic migration of PaSCs. [9]

Following activation, PaSCs have two fates. [6] If there is sustained inflammation and injury, PaSC activation is perpetuated, resulting in the growth of pancreatic fibrosis. [6] The activation of P2 receptors induces intracellular calcium signalling which mediates the fibrogenic function of activated stellate cells. [10] However, if inflammation and injury is minor, PaSCs undergo an apoptotic fate and become quiescent, preventing the development of fibrosis. [6]

PaSCs also display ethanol inducible ADH activity. [7] The possibility that pancreatic stellate cells may be exposed to ethanol and acetaldehyde during ethanol consumption is likely, as the pancreas metabolise ethanol to acetaldehyde through the oxidative pathway. [7] PaSCs are activated upon exposure to ethanol and its metabolite acetaldehyde or to oxidant stress. [7] Ethanol at clinically relevant concentrations causes α-SMA expression and collagen production in PaSCs but produce a minimal effect on cell proliferation. [7]

Increased α-SMA expression in stellate cells exposed to ethanol suggests activation and transformation of the cells to a myofibroblast phenotype. [7] Incubation of PaSCs with ethanol in the presence of ADH inhibitor 4MP had inhibited the increase in collagen synthesis induced by ethanol. [7] The conversion of ethanol to acetaldehyde via ADH is a significant step in the ethanol induced activation of pancreatic stellate cells. [7]

Clinical significance

Pancreatitis

Pancreatitis is generally classified into two forms, acute and chronic. [11] In acute pancreatitis, necroinflammation of the organ occurs, while chronic pancreatitis is distinguished by the progressive loss of endocrine and exocrine function. [11] After pancreatic damage occurs, pathologic events such as interstitial oedema, necrosis of parenchymal cells, activation and proliferation of PaSCs take place. [1] Inflammation and parenchymal necrosis precede PaSC activation. [1] Activated PaSCs are located in areas of major necrosis and inflammation that harbour cytokines, growth factors and reactive oxygen species. [1] Inflammatory processes are essential in contributing towards the activation of stellate cells. [1] Therefore, both autocrine and paracrine mediators are involved pancreatic stellate cell activation. [1]

Copious amounts of α- SMA-expressing cells are present in fibrotic areas of pancreatic tissue sections in patients with chronic pancreatitis. [1] α-SMA-expressing cells in fibrotic areas yield MRNA encoding pro-collagen α1I, indicating that activated PaSCs are the predominant source of collagen in pancreatic fibrosis. [1] Activated PaSCs and other myo-fibroblast cells contribute to the formation of a provisional matrix at the injury site, which enables cell proliferation, migration and the assembly of new parenchymal cells. [1] In the majority of cases, activated PaSCs recede after the termination of the injurious agent, however repeated pancreatic damage can result in proliferation of PaSCs and eventual fibrosis. [1]

In humans, persistent injury to the pancreas is linked with chronic alcohol use, pancreatic duct obstruction and genetic. [1] Chronic damage leads to the sustained activation of the active PaSC phenotype. [1] Diminished production of MMPs by PaSCs also contributes to the fibrotic phenotype. [1] Other factors may also drive the persistent activated state of PaSCs in the event of pancreatitis. [1] For example, PaSCs express protease activated receptor-2 (PAR-2), which is cleaved by trypsin to become active. [1] Active PAR-2 then instigates PaSC growth and collagen synthesis. [1]

Cancer

Pancreatic adenocarcinomas are recognised by tumour desmoplasia, distinguished by an increase in the connective tissue that surrounds the neoplasm. [1] Activated PaSCs in the tumour desmoplasia of human pancreatic cancers express α-SMA and co-localise with MRNA encoding pro-collagen α1I. [1] These factors are significant contributors of the ECM proteins that compose the desmoplasia. [1] A symbiotic relationship exists between pancreatic adenocarcinoma cells and PaSCs, which leads to an overall increase in the rate of growth of the tumour. [1] For example, culture supernatants from human pancreatic tumour cell lines induce PaSC proliferation and the production of ECM proteins. [1]

Pancreatic tumour cells stimulate the proliferation of PaSCs through the secretion of PDGF, and induce PaSC production of ECM proteins by secreting TGF-β1 and FGF-2. [1] Pancreatic tumour cells and PaSCs operate in a symbiotic relationship in animal studies, however data from human pancreatic tumours is limited. [1] Connective tissue growth factor is involved in the pathogenesis of fibrotic diseases and is predominantly found in PaSCs through regulation by TGF-β. [1]

Pancreatic cancer cells also stimulate proliferation, ECM production and TIMP1 production in PaSCs. [5] The production of these factors is regulated by fibroblast growth factor 2, TGF-β1, and PDGF. [5] In addition to cytokine-mediated mechanisms, PaSCs also produce a tumour supportive micro-environment through the production of matri-cellular proteins. [5] The up regulation of matri-cellular proteins such as galectin-1, and tenascin-C is present in the stromal tissues of pancreatic cancer and chronic pancreatitis. [5] Matricellular proteins induce proliferation, migration and production of cytokines, ECM and angiogenic responses in PaSCs, which in turn induce cancer cell proliferation. [5] Matri-cellular proteins may therefore directly contribute to the development of pancreatic cancer through stimulating cancer cell activity. [5] The matricellular protein also facilitates a tumour supportive microenvironment through sustained fibrogenic stellate cell activity. [5]

A hypoxic environment in tumours influences pancreatic cancer progression. [12] An oxygen deficient environment concomitantly exists not only in cancer cells but also in surrounding pancreatic stellate cells. [12] The cellular response to hypoxia is mediated by the transcription factor HIF-1, which is a heterodimer protein composed of α and β subunits. [12] Hypoxia also stimulates nuclear expression of HIF-1α followed by the production of vascular endothelial growth factor (VEGF) in PaSCs. [12] The induction of HIF-α indicates that PaSCs serves as oxygen sensing cells within the pancreas. [12] PaSCs, endothelial cells and other cells involved in the development of pancreatic fibrosis therefore function in coordination with a low oxygen microenvironment. [12]

Ongoing research

Treatment of chronic pancreatitis and pancreatic cancer aims to target the major mechanisms involved in both their activation and proliferation. [1] For example, inhibition of the receptors PDGF, TGF-β and angiotensin II in addition to suppression of the intracellular signalling pathways downstream of these receptors is likely to be of therapeutic benefit. [1] In vitro experiments indicate that PaSCs influence the activation and proliferation process for mitogen activated protein kinase (MAPK) pathways, in particular ERK1/2, p38 kinase and JNK. [1] The inhibition of the majority of MAPK pathways leads to a reduction in the activation and proliferation of PaSCs. [1]

Anti-fibrosis treatment strategies targeting PaSCs include inhibition of the activation of quiescent PaSCs. [5] Agents such as angiotensin receptor blockers, serine protease inhibitors and adenine dinucleotide phosphate oxidase inhibit the activation and function of PaSCs. [5] Camostat mesilate, an oral protease inhibitor, that is used to treat patients with chronic pancreatitis inhibited the proliferation and MCP-1 production in PaSCs in vitro . [5] The success and effect of anti-fibrosis therapies in pancreatic cancer treatment, however, remains unclear. [5]

Rat PaSCs express COX-2 when stimulated with TGF beta 1 (TGF-β1) and other cytokines. [1] pharmacological inhibition of COX-2 and inhibition of TGF-β1 signalling pathway decreases the expression of COX-2, α-SMA and collagen I, indicating that COX-2 may be a therapeutic target for pancreatic cancer and chronic pancreatitis. [1] Strategies aimed at inducing PaSC transformation from an activated to a quiescent state and inducing PaSC apoptosis may also be used to treat pancreatic cancer and chronic pancreatitis. [1] For example, the administration of vitamin A induces culture activated rat PaSCs to trans-differentiate to a quiescent state, preventing the progression of pancreatic cancer and pancreatitis. [1]

History

While the discovery of hepatic stellate cells is attributed to Karl Wilhelm von Kupffer in 1876, who had termed them “stellate cells”, the original discovery is attributed to more than one research group. [4] The first documented observations of PaSCs were recorded by Watari et al. in 1982. [13] Watari observed the pancreas of vitamin A primed mice using fluorescence microscopy and electron microscopy. [9] Cells displaying fading blue-green fluorescence typical of vitamin A in the periacinar region of pancreas was observed. [9] Watari likened these cells to hepatic stellate cells. [9] The publication of two seminal research papers in 1998 outlining the isolation of these cells provided an in vitro method by which researchers may characterise PaSCs in both health and pathology. [3]

See also

Related Research Articles

<span class="mw-page-title-main">Pancreas</span> Organ of the digestive system and endocrine system of vertebrates

The pancreas is an organ of the digestive system and endocrine system of vertebrates. In humans, it is located in the abdomen behind the stomach and functions as a gland. The pancreas is a mixed or heterocrine gland, i.e., it has both an endocrine and a digestive exocrine function. 99% of the pancreas is exocrine and 1% is endocrine. As an endocrine gland, it functions mostly to regulate blood sugar levels, secreting the hormones insulin, glucagon, somatostatin and pancreatic polypeptide. As a part of the digestive system, it functions as an exocrine gland secreting pancreatic juice into the duodenum through the pancreatic duct. This juice contains bicarbonate, which neutralizes acid entering the duodenum from the stomach; and digestive enzymes, which break down carbohydrates, proteins and fats in food entering the duodenum from the stomach.

<span class="mw-page-title-main">Cytokine</span> Broad and loose category of small proteins important in cell signaling

Cytokines are a broad and loose category of small proteins important in cell signaling. Due to their size, cytokines cannot cross the lipid bilayer of cells to enter the cytoplasm and therefore typically exert their functions by interacting with specific cytokine receptors on the target cell surface. Cytokines have been shown to be involved in autocrine, paracrine and endocrine signaling as immunomodulating agents.

<span class="mw-page-title-main">Fibrosis</span> Excess connective tissue in healing

Fibrosis, also known as fibrotic scarring, is a pathological wound healing in which connective tissue replaces normal parenchymal tissue to the extent that it goes unchecked, leading to considerable tissue remodelling and the formation of permanent scar tissue.

<span class="mw-page-title-main">Chronic pancreatitis</span> Medical condition

Chronic pancreatitis is a long-standing inflammation of the pancreas that alters the organ's normal structure and functions. It can present as episodes of acute inflammation in a previously injured pancreas, or as chronic damage with persistent pain or malabsorption. It is a disease process characterized by irreversible damage to the pancreas as distinct from reversible changes in acute pancreatitis. Tobacco smoke and alcohol misuse are two of the most frequently implicated causes, and the two risk factors are thought to have a synergistic effect with regards to the development of chronic pancreatitis. Chronic pancreatitis is a risk factor for the development of pancreatic cancer.

Durotaxis is a form of cell migration in which cells are guided by rigidity gradients, which arise from differential structural properties of the extracellular matrix (ECM). Most normal cells migrate up rigidity gradients.

<span class="mw-page-title-main">Amphiregulin</span> Protein-coding gene in the species Homo sapiens

Amphiregulin, also known as AREG, is a protein synthesized as a transmembrane glycoprotein with 252 aminoacids and it is encoded by the AREG gene. in humans.

<span class="mw-page-title-main">Transforming growth factor beta</span> Cytokine

Transforming growth factor beta (TGF-β) is a multifunctional cytokine belonging to the transforming growth factor superfamily that includes three different mammalian isoforms and many other signaling proteins. TGFB proteins are produced by all white blood cell lineages.

<span class="mw-page-title-main">Mothers against decapentaplegic homolog 4</span> Mammalian protein found in Homo sapiens

SMAD4, also called SMAD family member 4, Mothers against decapentaplegic homolog 4, or DPC4 is a highly conserved protein present in all metazoans. It belongs to the SMAD family of transcription factor proteins, which act as mediators of TGF-β signal transduction. The TGFβ family of cytokines regulates critical processes during the lifecycle of metazoans, with important roles during embryo development, tissue homeostasis, regeneration, and immune regulation.

<span class="mw-page-title-main">TGF beta 1</span> Protein-coding gene in the species Homo sapiens

Transforming growth factor beta 1 or TGF-β1 is a polypeptide member of the transforming growth factor beta superfamily of cytokines. It is a secreted protein that performs many cellular functions, including the control of cell growth, cell proliferation, cell differentiation, and apoptosis. In humans, TGF-β1 is encoded by the TGFB1 gene.

<span class="mw-page-title-main">Myofibroblast</span>

A myofibroblast is a cell phenotype that was first described as being in a state between a fibroblast and a smooth muscle cell.

<span class="mw-page-title-main">Alveolar macrophage</span>

An alveolar macrophage, pulmonary macrophage, is a type of macrophage, a professional phagocyte, found in the airways and at the level of the alveoli in the lungs, but separated from their walls.

<span class="mw-page-title-main">CTGF</span> Protein-coding gene in the species Homo sapiens

CTGF, also known as CCN2 or connective tissue growth factor, is a matricellular protein of the CCN family of extracellular matrix-associated heparin-binding proteins. CTGF has important roles in many biological processes, including cell adhesion, migration, proliferation, angiogenesis, skeletal development, and tissue wound repair, and is critically involved in fibrotic disease and several forms of cancers.

<span class="mw-page-title-main">TGF alpha</span> Protein

Transforming growth factor alpha (TGF-α) is a protein that in humans is encoded by the TGFA gene. As a member of the epidermal growth factor (EGF) family, TGF-α is a mitogenic polypeptide. The protein becomes activated when binding to receptors capable of protein kinase activity for cellular signaling.

<span class="mw-page-title-main">Hepatic stellate cell</span>

Hepatic stellate cells (HSC), also known as perisinusoidal cells or Ito cells, are pericytes found in the perisinusoidal space of the liver, also known as the space of Disse. The stellate cell is the major cell type involved in liver fibrosis, which is the formation of scar tissue in response to liver damage, in addition these cells store and concentrate vitamin A.

<span class="mw-page-title-main">Integrin beta 6</span> Protein-coding gene in the species Homo sapiens

Integrin beta-6 is a protein that in humans is encoded by the ITGB6 gene. It is the β6 subunit of the integrin αvβ6. Integrins are αβ heterodimeric glycoproteins which span the cell’s membrane, integrating the outside and inside of the cell. Integrins bind to specific extracellular proteins in the extracellular matrix or on other cells and subsequently transduce signals intracellularly to affect cell behaviour. One α and one β subunit associate non-covalently to form 24 unique integrins found in mammals. While some β integrin subunits partner with multiple α subunits, β6 associates exclusively with the αv subunit. Thus, the function of ITGB6 is entirely associated with the integrin αvβ6.

<span class="mw-page-title-main">TNFRSF18</span> Protein-coding gene in the species Homo sapiens

Tumor necrosis factor receptor superfamily member 18 (TNFRSF18), also known as glucocorticoid-induced TNFR-related protein (GITR) or CD357. GITR is encoded and tnfrsf18 gene at chromosome 4 in mice. GITR is type I transmembrane protein and is described in 4 different isoforms. GITR human orthologue, also called activation-inducible TNFR family receptor (AITR), is encoded by the TNFRSF18 gene at chromosome 1.

<span class="mw-page-title-main">Dermatopontin</span> Protein-coding gene in the species Homo sapiens

Dermatopontin also known as tyrosine-rich acidic matrix protein (TRAMP) is a protein that in humans is encoded by the DPT gene. Dermatopontin is a 22-kDa protein of the noncollagenous extracellular matrix (ECM) estimated to comprise 12 mg/kg of wet dermis weight. To date, homologues have been identified in five different mammals and 12 different invertebrates with multiple functions. In vertebrates, the primary function of dermatopontin is a structural component of the ECM, cell adhesion, modulation of TGF-β activity and cellular quiescence). It also has pathological involvement in heart attacks and decreased expression in leiomyoma and fibrosis. In invertebrate, dermatopontin homologue plays a role in hemagglutination, cell-cell aggregation, and expression during parasite infection.

An inflammatory cytokine or proinflammatory cytokine is a type of signaling molecule that is secreted from immune cells like helper T cells (Th) and macrophages, and certain other cell types that promote inflammation. They include interleukin-1 (IL-1), IL-6, IL-12, and IL-18, tumor necrosis factor alpha (TNF-α), interferon gamma (IFNγ), and granulocyte-macrophage colony stimulating factor (GM-CSF) and play an important role in mediating the innate immune response. Inflammatory cytokines are predominantly produced by and involved in the upregulation of inflammatory reactions.

<span class="mw-page-title-main">Tumor microenvironment</span> Surroundings of tumors including nearby cells and blood vessels

The tumor microenvironment (TME) is a complex ecosystem surrounding a tumor, composed of a variety of non-cancerous cells including blood vessels, immune cells, fibroblasts, signaling molecules and the extracellular matrix (ECM). Mutual interaction between cancer cells and the different components of the TME support its growth and invasion in healthy tissues which correlates with tumor resistance to current treatments and poor prognosis. Tumors can influence the microenvironment by releasing extracellular signals, promoting tumor angiogenesis and inducing peripheral immune tolerance, while the immune cells in the microenvironment can affect the growth and evolution of cancerous cells.

A cancer-associated fibroblast (CAF) is a cell type within the tumor microenvironment that promotes tumorigenic features by initiating the remodelling of the extracellular matrix or by secreting cytokines. CAFs are a complex and abundant cell type within the tumour microenvironment; the number cannot decrease, as they are unable to undergo apoptosis.

References

  1. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 Omary MB, Lugea A, Lowe AW, Pandol SJ (January 2007). "The pancreatic stellate cell: a star on the rise in pancreatic diseases". J. Clin. Invest. 117 (1): 50–9. doi:10.1172/JCI30082. ISSN   0021-9738. PMC   1716214 . PMID   17200706.
  2. 1 2 Charo C, Holla V, Arumugam T, Hwang R, Yang P, Dubois RN, Menter DG, Logsdon CD, Ramachandran V (April 2013). "Prostaglandin E2 regulates pancreatic stellate cell activity via the EP4 receptor". Pancreas. 42 (3): 467–74. doi:10.1097/MPA.0b013e318264d0f8. ISSN   0885-3177. PMC   3600062 . PMID   23090667.
  3. 1 2 Apte MV, Haber PS, Applegate TL, Norton ID, McCaughan GW, Korsten MA, Pirola RC, Wilson JS (July 1998). "Periacinar stellate shaped cells in rat pancreas: identification, isolation, and culture". Gut. 43 (1): 128–33. doi:10.1136/gut.43.1.128. ISSN   0017-5749. PMC   1727174 . PMID   9771417.
  4. 1 2 3 Ferdek PE, Jakubowska MA (September 2017). "Biology of pancreatic stellate cells-more than just pancreatic cancer". Pflügers Arch. 469 (9): 1039–1050. doi:10.1007/s00424-017-1968-0. ISSN   0031-6768. PMC   5554282 . PMID   28382480.
  5. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 Masamune A, Watanabe T, Kikuta K, Shimosegawa T (November 2009). "Roles of pancreatic stellate cells in pancreatic inflammation and fibrosis". Clin. Gastroenterol. Hepatol. 7 (11 Suppl): S48–54. doi: 10.1016/j.cgh.2009.07.038 . ISSN   1542-3565. PMID   19896099.
  6. 1 2 3 4 Masamune A, Shimosegawa T (2009). "Signal transduction in pancreatic stellate cells". J. Gastroenterol. 44 (4): 249–60. doi: 10.1007/s00535-009-0013-2 . ISSN   0944-1174. PMID   19271115.
  7. 1 2 3 4 5 6 7 8 Apte MV, Phillips PA, Fahmy RG, Darby SJ, Rodgers SC, McCaughan GW, Korsten MA, Pirola RC, Naidoo D, Wilson JS (April 2000). "Does alcohol directly stimulate pancreatic fibrogenesis? Studies with rat pancreatic stellate cells". Gastroenterology. 118 (4): 780–94. doi:10.1016/S0016-5085(00)70148-X. ISSN   0016-5085. PMID   10734030.
  8. Shinozaki S, Ohnishi H, Hama K, Kita H, Yamamoto H, Osawa H, Sato K, Tamada K, Mashima H, Sugano K (July 2008). "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. 216 (1): 38–46. doi:10.1002/jcp.21372. ISSN   0021-9541. PMID   18286538. S2CID   11204851.
  9. 1 2 3 4 5 Apte MV, Pirola RC, Wilson JS (2012). "Pancreatic stellate cells: a starring role in normal and diseased pancreas". Front Physiol. 3: 344. doi: 10.3389/fphys.2012.00344 . ISSN   1664-042X. PMC   3428781 . PMID   22973234.
  10. Hennigs JK, Seiz O, Spiro J, Berna MJ, Baumann HJ, Klose H, Pace A (July 2011). "Molecular basis of P2-receptor-mediated calcium signaling in activated pancreatic stellate cells". Pancreas. 40 (5): 740–6. doi:10.1097/MPA.0b013e31821b5b68. ISSN   0885-3177. PMID   21654543. S2CID   21538837.
  11. 1 2 Mews P, Phillips P, Fahmy R, Korsten M, Pirola R, Wilson J, Apte M (April 2002). "Pancreatic stellate cells respond to inflammatory cytokines: potential role in chronic pancreatitis". Gut. 50 (4): 535–41. doi:10.1136/gut.50.4.535. ISSN   0017-5749. PMC   1773172 . PMID   11889076.
  12. 1 2 3 4 5 6 Xu Z, Vonlaufen A, Phillips PA, Fiala-Beer E, Zhang X, Yang L, Biankin AV, Goldstein D, Pirola RC, Wilson JS, Apte MV (November 2010). "Role of pancreatic stellate cells in pancreatic cancer metastasis". Am. J. Pathol. 177 (5): 2585–96. doi:10.2353/ajpath.2010.090899. ISSN   0002-9440. PMC   2966814 . PMID   20934972.
  13. Watari N, Hotta Y, Mabuchi Y (March 1982). "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. 58 (4–6): 837–58. doi: 10.2535/ofaj1936.58.4-6_837 . ISSN   0030-154X. PMID   7122019.