You are on page 1of 13

Med Electron Microsc (2004) 37:3–15 © The Clinical Electron Microscopy Society of Japan 2004

DOI 10.1007/s00795-003-0230-3

SPECIAL REVIEW SERIES: Hepatic sinusoidal cells in liver physiology and pathology

Haruki Senoo

Structure and function of hepatic stellate cells

Received: June 12, 2003 / Accepted: June 16, 2003

Abstract Hepatic stellate cells (vitamin A-storing cells,


Introduction
lipocytes, interstitial cells, fat-storing cells, Ito cells) exist in
the space between parenchymal cells and sinusoidal endot-
helial cells of the hepatic lobule and store 80% of retinoids The hepatic parenchyma consists of parenchymal cells (epi-
in the whole body as retinyl palmitate in lipid droplets in thelial cells) and nonparenchymal cells associated with the
the cytoplasm. In physiological conditions, these cells sinusoids: endothelial cells, Kupffer cells, pit cells, dendritic
play pivotal roles in the regulation of retinoid homeostasis; cells, and stellate cells (vitamin A-storing cells, lipocytes,
they express specific receptors for retinol-binding protein interstitial cells, fat-storing cells, Ito cells) (Fig. 1).1–3 Endot-
(RBP), a binding protein specific for retinol, on their cell helial cells express lymphocyte co-stimulatory molecules4
surface, and take up the complex of retinol and RBP by and form the greater part of the extremely thin lining of the
receptor-mediated endocytosis. Hepatic stellate cells in sinusoids, which are larger than ordinary capillaries and
arctic animals such as polar bears and arctic foxes store 20– more irregular in shape. Kupffer cells are tissue macroph-
100 times the levels of retinoids found in humans or rats. ages and components of the diffuse mononuclear phagocyte
In pathological conditions such as liver fibrosis, hepatic stel- system. They are usually situated on the endothelium with
late cells lose retinoids, and synthesize a large amount of cellular processes extending between the underlying endot-
extracellular matrix (ECM) components including collagen, helial cells. The greater part of their irregular cell surface is
proteoglycan, and adhesive glycoproteins. Morphology of exposed to the blood in the lumen of the sinusoid. Dendritic
these cells also changes from the star-shaped stellate cells to cells capture and process antigens, migrate to lymphoid
that of fibroblasts or myofibroblasts. The three-dimensional organs, and secrete cytokines to initiate immune re-
structure of ECM components was found to regulate revers- sponses.5–7 The hepatic stellate cells2,3,8–10 that lie in the space
ibly the morphology, proliferation, and functions of the between sinusoidal endothelial cells and parenchymal cells
hepatic stellate cells. Molecular mechanisms in the revers- are considered to be derived from mesenchymal origin.
ible regulation of the stellate cells by ECM imply cell- Recently, stellate cells that store retinoids in their cyto-
surface integrin binding to ECM components, followed by plasm have been found in extrahepatic organs and charac-
signal transduction processes and then cytoskeleton assem- terized.11,12 The purpose of this review is to survey recent
bly. Stellate cells also exist in extrahepatic organs such as progress in studies of structure and function of the stellate
pancreas, lung, kidney, and intestine. Hepatic and extrahe- cells.
patic stellate cells form the stellate cell system.

Key words Hepatic stellate cells · Retinoids · Arctic


animals · Extracellular matrix · Collagen Regulation of retinoid homeostasis

Retinoids (vitamin A and its metabolites) (Fig. 2) are


known to regulate diverse cellular activities such as cell
proliferation, differentiation, morphogenesis, and tumori-
genesis.13,14 In physiological conditions, stellate cells store
80% of the total retinoids in the whole body as retinyl
H. Senoo (*) palmitate in lipid droplets in the cytoplasm and regulate
Department of Anatomy, Akita University School of Medicine, both transport and storage of retinoids (Fig. 3).15–18
1-1-1 Hondo, Akita 010-8543, Japan
Tel. ⫹81-18-884-6056; Fax ⫹81-18-834-7808 When [3H]retinol was injected via the portal vein, the
e-mail: senoo@ipc.akita-u.ac.jp largest amount of the labeled retinol was taken up by the
4

Fig. 1. Structure of a hepatic lobule. Hepatic cords of the lobule consist store 80% of retinoids of the whole body as retinyl palmitate in the
of parenchymal cells (PC). Endothelial cells (EC) form the thin lining lipid droplets in the cytoplasm. Arrows indicate a classical definition
of the sinusoids (S). Kupffer cells (KC) are tissue macrophages and of the perisinusoidal space of Disse between PCs and ECs; asterisks
belong to the monocyte-macrophage cell lineage. Stellate cells (HSC) indicate a new concept of the perisinusoidal space of Disse between
lie in the space between parenchymal cells and endothelial cells and PCs and the complex of HSCs and ECs

Fig. 2. Structural formulas of some naturally occurring retinoid and β-


carotene. Retinoid circulates in the plasma as retinol (1) that binds to
a specific carrier protein, retinol-binding protein. Retinoid is stored as
retinyl palmitate (2) in the hepatic stellate cells. 11-cis-Retinal (3)
exists in the retina with rhodopsin. All-trans-retinoic acid (4) binds to
nuclear retinoic acid receptors (RAR-α, RAR-β, RAR-γ), and 9-cis-
retinoic acid (5) binds to nuclear retinoid X receptors (RXR-α, RXR-
β, RXR-γ). These nuclear receptors regulate transcription of various
genes. 13-cis-Retinoic acid (6) can bind to RARs. β-Carotene (7) forms
two retinals and finally two retinols
5

Fig. 3. Major pathway for retinoid transport in the body. Dietary parenchymal cells and to some extent also by cells in other organs. In
retinyl esters (RE) are hydrolyzed to retinol (ROH) in the intestinal liver parenchymal cells, retinyl esters are rapidly hydrolyzed to retinol,
lumen before absorption by enterocytes, and carotenoids are absorbed which then binds to retinol-binding protein (RBP). A complex of
and then partially converted to retinol in the enterocytes. In the retinol–RBP is secreted and transported to hepatic stellate cells. Stel–
enterocytes, retinol reacts with fatty acid to form esters before incorpo- late cells store retinoids mainly as retinyl palmitate and secrete retinol–
ration into chylomicrons (CM). Chylomicrons then reach the general RBP directly into the blood. Most retinol–RBP in the bloodstream is
circulation by way of the intestinal lymph, and chylomicron remnants reversibly complexed with transthyretin (TTR). The uncomplexed
(CMR) are formed in blood capillaries. Chylomicron remnants, which retinol–RBP is presumably taken up in a variety of cells by cell-
contain almost all the absorbed retinol, are mainly cleared by the liver surface receptors specific for RBP

Fig. 4. Uptake and storage of retinol and retinol-binding protein


(RBP) by stellate cells. A complex of retinol and RBP circulates in the
blood. The complex binds specifically to the receptor expressed on the
cell surface of stellate cells (a), then reaches endosomes (d) through
pits (b) and vesicles (c). From the endosomes, retinol can take three
pathways: (1) retinol binds cellular retinol-binding protein (CRBP;
MW 14 000–17 500) and is esterified with palmitic acid in multivesicular
bodies (MVB) and stored in lipid droplets; (2) retinol is oxidized to
retinoic acid, which binds with cellular retinoic acid-binding protein
(CRABP; MW 16 000) or is transported and binds with nuclear retinoic
acid receptors (RAR for all-trans-retinoic acid, RXR for 9-cis-retinoic
acid); (3) retinol is transported from endosomes to rough-surface endo-
plasmic reticulum (rER), binds with RBP, and is secreted to the outside
of the cell through the Golgi apparatus (G) and secretory vesicles (sv).
(4) RBP and its receptor are recycled and reutilized
6

liver within 90 min after injection, although the labeled mate- Immunoelectron microscopic studies suggest that RBP
rial was detected in all organs examined.19 The radioactivity mediates the paracrine transfer of retinol from hepatic
of the retinol in the liver did not change until 6 days after the parenchymal cells to the stellate cells and that stellate
injection. These results were consistent with the reports that cells bind and internalize RBP by receptor-mediated
main storage site of retinoids in mammals is the liver.2,3 endocytosis.25–28 Recently, RBP receptor was cloned and
To examine the distribution of retinol in the liver, radio- characterized.29–31
activity per cell was determined after cell fractionation.19,20 Once inside the cell, free retinol has several fates, one of
Specific activity of [3H]retinol (per cell) was the highest in which is reformation of the complex with RBP and a return
the hepatic stellate cell fraction, both 90 min and 6 days to the bloodstream (Fig. 4).9,13,32 Thus, the hepatic stellate
after injection. These results strongly support earlier mor- cells are important for the regulation of homeostasis of
phological observations2,3 that the stellate cell is the storage retinoids.
site of retinoids in the liver and are not inconsistent with
reports on the retinol transfer from parenchymal cells to
stellate cells.21–23
The concentration of retinoids in the bloodstream is Hepatic stellate cells in arctic animals
regulated within the physiological range by these stellate
cells. By receptor-mediated endocytosis, the cells take up More than 50 years ago, Rodahl reported that animals in
retinol from the blood, where it circulates as a complex of the arctic area were able to store a large amount of retinoids
retinol and a specific binding protein called retinol-binding in the liver.33–35 To investigate the cellular and molecular
protein (RBP).15,18,24 mechanisms in transport and storage of retinoids in these

Fig. 5. Transmission electron micrographs of the livers of polar bears area (b, e), and central area (c, f) of the hepatic lobule. L, lipid droplet;
and arctic foxes. Electron micrographs of the livers of polar bears (a– SC, stellate cell; s, sinusoid; CV, central vein; pc, parenchymal cell
c) and arctic foxes (d–f) were taken in portal area (a, d), intermediate
7

arctic animals, we performed a study in the Svalbard within other organs such as kidney, spleen, lung, and
archipelago (situated at 80° N, 15° E).36–39 After obtaining jejunum. Top predators among arctic animals stored 6–
permission to hunt the animals from the district governor 23 µmole retinyl ester per gram liver, which is 20–100 times
of Svalbard, 11 arctic foxes (Alopex lagopus), 14 bearded the levels normally found in other animals, including
seals (Erignathus barbatus), 22 glaucous gulls (Larus humans. These results indicate that the hepatic stellate cells
hyperboreus), 5 fulmars (Fulmarus glacialis), 4 Brünnich’s in these animals have high ability for uptake and enough
guillemots (Uria lomvia), 6 ringed seals (Phoca hispida), 5 capacity for storage of retinoids.36–39
hooded seals (Cystophora cristata), 6 puffins (Fratercula As xenobiotics may reduce the threshold of retinoid
arctica), 5 Svalbard ptarmigans (Lagopus mutus toxicity and both retinoid and fat-soluble xenobiotics have
hyperboreus), and 7 Svalbard reindeer (Rangifer tarandus a tendency to accumulate in the food chain,40–46 we have
platyrhynchus) were caught in the period from August 1996 searched for signs of retinoid-related toxicity in these
to September 2001. Three polar bears (Ursus maritimus) animals.47–49 Kidney total retinol, which may be used as a
were shot in self-defense at Svalbard in February and biomarker for retinoid-related toxicity or excess, in polar
August 1998 in Ny Ålesund and Hornsund. We also bears and bearded seals was less than 1% of their liver
obtained 13 brown bears (Ursus arctos) from Jämtland, value, which is in the normal range for most animals. Arctic
Gävleborg, and Dalarna, 4 red foxes (Vulpes vulpes) foxes and glaucous gulls, however, had kidney levels of
from Västergötaland, and 8 grey gulls (Larus argentatus) about 9% and 42% of the liver values, respectively. This
from Skåne, Sweden. increased kidney concentration and decreased capacity for
Fresh organs, namely, the liver, kidney, spleen, lung, storage in hepatic stellate cells of total retinol is most likely
and jejunum, were examined by morphological methods a sign of retinoid toxicity that deserves attention. This
and high-performance liquid chromatography. Serum from observation is alarming as this has not been seen previously
each animal was analyzed with high-performance liquid in wild animals.
chromatography.
The arctic animals stored retinoids in hepatic stellate
cells (Figs. 5–7). Only a small amount of retinoids existed

Fig. 6. Gold chloride staining specifically demonstrates black-stained Fig. 7. Fluorescence micrographs demonstrate retinoid auto-
hepatic stellate cells of polar bears (a, b) and arctic foxes (c, d). Bars fluorescence in hepatic stellate cells of polar bears (a) and arctic
100 µm foxes (b). Bars 100 µm
8

Fig. 8. Phenotypic changes of the hepatic stellate cells. In physiological


conditions, the hepatic stellate cells (SC) store 80% of retinoids in the
whole body as retinyl palmitate in the lipid droplets (L) in the cyto-
plasm, whereas in pathological conditions, such as liver cirrhosis, these
cells lose retinoids, and synthesize a large amount of extracellular
matrix components. Morphology of the cells also changes from the
star-shaped stellate cells to that of the fibroblasts or myofibroblasts
(MYO) (passage of a). Inductive conditions to passage b imply the
reversibility of hepatic fibrosis

Production and degradation of ECM components by


hepatic stellate cells

In pathological conditions such as liver cirrhosis, the hepatic


stellate cells lose retinoids, proliferate vigorously, and
Fig. 9. Fluorescence autoradiograms of [3H]proline-labeled proteins.
synthesize and secrete a large amount of extracellular Stellate cells at confluency in the secondary passage were cultured in a
matrix (ECM) components such as collagen, proteoglycan, medium containing [3H]proline for 18 h. Collagenous proteins were
and glycoprotein. The structure of the cells also changes partially purified from the cell layer and medium with pepsin digestion
from star-shaped stellate cells to that of fibroblast-like after precipitation with ammonium sulfate. Sodium dodecyl sulfate-
polyacrylamide slab gel electrophoresis was performed before (a, b) or
cells or myofibroblasts50 with well-developed rough- after (c, d) treatment with purified bacterial collagenase and processed
surface endoplasmic reticulum and Golgi apparatus for fluorescence autoradiography. b and d show electrophoresis after
(Fig. 8).8–10,51 reduction with dithiothreitol (DTT). Arrows indicate the migration
positions of carrier rat collagen chains
To elucidate cell type or types responsible for collagen
metabolism among nonparenchymal cells in the liver, col-
lagen production by stellate cells and Kupffer and endothe-
lial cells was analyzed.19 Stellate cells were found to produce (Fig. 9, lanes c and d). Quantitation of these collagen bands
collagen on day 8 in primary culture, although collagen by densitometry indicated that the percentage of type I,
production was not induced at an earlier stage of culture type III, and type IV collagen was 88.2%, 10.4%, and 1.4%,
(day 2). Capability of collagen production by cells was re- respectively, which is consistent with an observation on
tained in the secondary culture, suggesting that the stellate collagen types in human alcoholic liver cirrhosis. The
cell is a candidate cell responsible for collagen production. dysregulation of collagen gene expression in hepatic stellate
Kupffer and endothelial cells produced little collagen either cells is a central pathogenetic step during the development
on day 2 or day 8 in primary culture under the conditions of hepatic fibrosis.52
employed. Long–Evans cinnamon-like colored (LEC) rats
Types of collagen produced by stellate cells in secondary spontaneously develop hepatocellular carcinoma with
culture were analyzed by fluorography after sodium cholangiofibrosis after chronic hepatitis. To investigate the
dodecyl sulfate-polyacrylamide gel electrophoresis (SDS- role of hepatic stellate cells in induction and suppression of
PAGE) under nonreducing and reducing conditions (Fig. hepatic fibrosis, the liver of LEC rats was morphologically
9).19 Type I collagen is the major component synthesized examined.53 The liver of LEC rats 1.5 years of age showed
(Fig. 9, lane a). Minor components include type III collagen, cholangiofibrosis and subcellular injury of hepatic paren-
which remained at the γ-region under nonreducing condi- chymal cells. However, no diffuse hepatic fibrosis was
tion (Fig. 9, lane a) but migrated to the α1-region after observed in the liver, and hepatic stellate cells around
reduction (Fig. 9, lane b), and type IV collagen, which re- the regions of cholangiofibrosis were negative for α-smooth
mained slightly below the origin under nonreducing condi- muscle actin, an indicator of “activated” hepatic stellate
tion (Fig. 9, lane a) but migrated to a region slightly lower cells. The area of lipid droplets of a stellate cell in the liver
than the β-region after reduction (Fig. 9, lane b). All these of LEC rats was 1.6 to 1.8 times as large as that of normal
bands were susceptible to purified bacterial collagenase Wistar rats. The hepatic stellate cells did not participate in
9

the accumulation of collagen fibers around themselves amount of collagen synthesized by stellate cells was more
when the cells contained a large amount of retinoid lipid on type I collagen than on type IV collagen. Thus, these
droplets, even though the development of hepatic lesions recent studies support the idea that ECM regulates pheno-
was in progress. Antagonistic relationship between the stor- types of the hepatic stellate cells such as collagen metabo-
age of retinoids and production of collagen in the stellate lism and storage of retinoids in lipid droplets in the
cells51 was strongly supported. cytoplasm. Responses of the hepatic stellate cells to
Matrix metalloproteinases (MMPs) and tissue inhibitor cytokines are also modulated by ECM.77 When transform-
of metalloproteinases (TIMP) were reported to be synthe- ing growth factor-β (TGF-β) was applied to hepatic stellate
sized by hepatic parenchymal and stellate cells.54–62 Recent cells cultured on type I or type IV collagen-coated culture
reports indicate a differential expression of MMP activity; dishes, collagen synthesis of the cells inoculated on type I
thus, the remodeling of ECM components is dependent collagen-coated dishes was stimulated. On the other hand,
on the substratum used for the culture of hepatic stellate there was no response to TGF-β in terms of collagen syn-
cells.63,64 Another report demonstrated that the hepatic thesis by the hepatic stellate cells inoculated on type IV
stellate cells were important for regeneration of the liver.65 collagen. Thus, reactions of the hepatic stellate cells to
These reports indicate that hepatic stellate cells play pivotal cytokines are modulated by ECM.
roles in remodeling and regeneration of the liver. Our studies and other works clearly show that ECM
can regulate morphology, proliferation, and functions of
hepatic stellate cells.
We reported recently that morphology, proliferation,
Reversible regulation of morphology, proliferation, and collagen synthesis of the stellate cells were reversibly
and function of the hepatic stellate cells by three- regulated by three-dimensional structure of ECM.78–85 The
dimensional structure of ECM cellular processes of the hepatic stellate cells were demon-

Tissues are not composed solely of cells. A substantial part


of their volume is intercellular space that is largely filled by
an intricate network of macromolecules constituting the
ECM. This matrix comprises a variety of polysaccharides
and proteins that are secreted locally and assembled into an
organized meshwork.66 ECM was considered to serve
mainly as a relatively inactive scaffolding to stabilize the
physical structure of tissues until recently. It is now clear,
however, that ECM plays a far more active complex role
in regulating the behavior of the cells that are contacted,
influencing their morphology, development, migration,
proliferation, and functions.67–70
We have reported that hepatic stellate cells proliferated
better and synthesized more collagen on type I collagen-
coated culture dishes than on polystyrene dishes.71 We also
demonstrated that the stellate cells formed a meshlike
structure, proliferated slowly, and synthesized a small
amount of collagen on a basement membrane gel prepared
from murine Engelbreth–Holm–Swarm (EHS) tumor,72 a
gel consisting largely of laminin, type IV collagen, heparan
sulfate proteoglycan, and nidogen.71
Other reports also indicate that ECM can regulate the
functions of stellate cells. The regulation of retinol esterifi-
cation activities, the central process in retinoid storage, was
evaluated in stellate cells cultured on type I or type IV
collagen-coated dishes in the presence of [3H]retinol.73 Up-
take of [3H]retinol into the cells and esterification into
retinyl palmitate was enhanced when the cells were cultured
on type IV collagen-coated dishes. The basement mem-
brane gel was reported to be able to maintain the differen-
tiated phenotype such as storage of lipids in cultured
stellate cells.74,75 Types of collagen synthesized by stellate Fig. 10. Motility of filopodia and lamellipodia in rat hepatic stellate
cells were also modulated by ECM.76 The cells synthesized cells inoculated on polystyrene surface and monitored by phase-
contrast video-enhanced microscopy. Photographs were taken at 29 s
mainly type IV collagen on type I collagen-coated culture
(2) and 55 s (3) after taking a picture of 1, and at 24 s (5) and 39 s (6),
dishes and synthesized equal amounts of type I and type IV respectively, after taking a picture of 4. Arrowheads indicate filopodia
collagen on type IV collagen-coated dishes. The total and asterisks indicate lamellipodia. Bars 20 µm
10

Fig. 12. Retraction of cellular processes by colchicine treatment. Rat


hepatic stellate cells cultured overnight on type I collagen gel were
monitored for 1 h after the addition of 1 µM colchicine. Phase-contrast
video-enhanced micrographs were taken 12 s (1), 15 min (2), 30 min (3),
45 min (4), and 60 min (5) after the addition of colchicine. Black arrow-
heads indicate the front of cellular process of the cell marked with white
arrowheads. Bars 100 µm

well, flattened with extensive stress fibers. The cell-surface


ruffling activity of filopodia and lamellipodia was prominent
(Fig. 10), reflecting weak adhesion to the substratum. All
filopodia remained dynamic throughout the 4-h recording
and extended and retracted repeatedly (Fig. 11). Within 1 h
Fig. 11. Extension of cellular processes in cultured rat stellate cells on after inoculating in or on type I collagen gel, the stellate
type I collagen gel. The cells were cultured on type I collagen gel and cells began to extend cellular processes (Fig. 11A–D), and
monitored by phase-contrast time-lapse video microscopy for up to 4 h. the cellular processes appeared to adhere to and extend
Photographs were taken 71 min (A), 2 h (B), 3 h (C), and 4 h (D) after
inoculation. Arrowheads indicate the front of elongating cellular along type I collagen fibers. After repeated extension and
processes. Bars 200 µm retraction of cellular processes, hepatic stellate cells dis-
played a number of long cellular processes with distal fine
branches by 4-h culture on type I collagen gel (Fig. 11D).
The cellular processes also extended in or on type III
strated to be extended and retracted according to the collagen gel, but not in type IV collagen-coated dishes
ECM, and were speculated to have important functions in or on Matrigel composed of the basement membrane
transport and storage of retinoids and transport of components.
metalloproteinases.86–90 These data also indicate that the The role of microtubule organization in maintenance of
stellate cells are not static, but dynamic, in the changeable the cellular process structure was demonstrated by video-
three-dimensional structure of ECM in the space between recording of the stellate cell culture after addition of colchi-
parenchymal cells and endothelial cells. cine or kinase inhibitors (Figs. 12, 13). In the presence of
The dynamic movement of cultured stellate cells was 1 µM colchicine, cellular processes that extended after over-
analyzed with video-enhanced optical microscopy.91 When night culture on type I collagen gel were time dependently
cultured on polystyrene surface, the stellate cells spread retracted (Fig. 12). The effects of colchicine were also dose
11

Fig. 13. Effects of protein tyrosine kinase inhibitor or herbimycin A (B) or 50 nM staurosporine (C), or with pho-
phosphatidylinositol 3-kinase inhibitor on extended cellular processes. sphatidylinositol 3-kinase inhibitor, 100 nM wortmannin (D). Bars
Rat hepatic stellate cells cultured overnight on type I collagen gel (A) 50 µm
were treated 1 h with protein tyrosine kinase inhibitor, 500 µg/ml

dependent at a concentration of 0.1 to 1.0 µM, and almost all


cells changed to a round shape within a few hours in the
presence of 1 µM colchicine. Virtually no effects were seen
after treatment with 1 µM γ-lumicolchicine as a control.
Cold treatment at 4°C, which is known to induce the degra-
dation of the cold-labile form of microtubules, also induced
the retraction of elongated cellular processes (Fig. 14)
within 3 h. The once extended cellular processes were also
partly retracted 1 h after treatment with 4 µg/ml cytochala-
sin B, as seen after treatment with 0.5 µM colchicine. The
effects of cytochalasin B at 4 µg/ml appeared to be weaker
than that of 1 µM colchicine, and a part of the cellular pro-
cesses still remained. However, almost all cells were
changed to round shapes after overnight treatment with
cytochalasin B.
Fibroblasts were also reported to change the phenotype
according to the three-dimensional structure of col-
lagen,83,92,93 and the integrin α2β1 was demonstrated to
recognize the three-dimensional structure of triple helical
collagen peptide.94

Stimulation of proliferation of hepatic stellate cells


and tissue formation of the liver by a long-acting
vitamin C derivative

A long-acting vitamin C derivative, l-ascorbic acid 2-


phosphate (Asc 2-P) (Fig. 15), was found to stimulate cell
proliferation, collagen accumulation, and tissue forma-
tion.95,96 On the basis of this discovery, Asc 2-P was added to
the medium in which hepatic stellate cells were cultured.71
The cells in the medium supplemented with Asc 2-P Fig. 14. Dual immunofluorescence staining of elongated cellular pro-
cesses of human hepatic stellate cells after culturing on type I collagen
stretched better than the cells in the control medium. Asc 2- gel. Cells were cultured overnight on type I collagen gel and then
P stimulated cell proliferation and collagen synthesis of the stained with FITC-labeled phalloidin for F-actin and with monoclonal
hepatic stellate cells and formation of the liver tissue-like antihuman tubulin-β and TRITC-labeled secondary antibody. A
Fluorescein isothiocyanate (FITC) image; B tetramethylrhodamine
structure in coculture of hepatic parenchymal cells and
isothiocyanate (TRITC) image. Bar 20 µm
fibroblasts (Fig. 16).97
12

Fig. 15. Molecular structure of l-ascorbic acid 2-phosphate,


magnesium salt

Extrahepatic stellate cells

Previous studies using fluorescence microscopy, transmis-


sion electron microscopy, and electron microscopic autora-
diography showed that cells that stored retinoids were
distributed in extrahepatic organs, namely, lung, digestive
tract, spleen, adrenal gland, testis, uterus, lymph node, thy-
mus, bone marrow, adventitia of the aorta, lamina propria
of the trachea, oral mucosa, and tonsil3,11,12 (and Wold et al.,
in manuscript). Morphology of these cells was similar to
that of fibroblasts. These cells emanate autofluorescence of
retinoids and contain lipid droplets in the cytoplasm. These
cells and hepatic stellate cells form the stellate cell system
that regulates homeostasis of retinoids in the whole body. Fig. 16. Stimulation of tissue formation of the liver by l-ascorbic acid
Extrahepatic stellate cells also can synthesize and secrete 2-phosphate. Hepatic parenchymal cells and tendon fibroblasts were
cultured with 0.2 mM l-ascorbic acid 2-phosphate for 30 days. B and C
ECM components. Pancreatic stellate cells, one type of ex- are higher magnification of a part of A. b, bile canaliculi; G, Golgi
trahepatic stellate cells, are now considered to be respon- apparatus; C, collagen fiber; m, microvilli; t, tight junction between
sible for the induction of chronic pancreatitis and pancreatic parenchymal cells. Arrow and arrowheads indicate basal and apical
surface of the fibroblast sheets, respectively. Bars 1 µm
fibrosis.98–103 These extrahepatic stellate cells are now to be
targets of the treatment of inflammation and organ fibrosis.

stellate cells by three-dimensional structure of the ECM


imply cell-surface integrin binding to the matrix compo-
Conclusions nents, followed by signal transduction processes and cytosk-
eleton assembly. Hepatic stellate cells play key roles in the
Hepatic stellate cells that lie in the space between parenchy- regeneration of the liver. The stellate cell system consists of
mal cells and endothelial cells play pivotal roles in the hepatic and extrahepatic stellate cells and regulates retinoid
regulation of homeostasis of retinoids in the whole body. homeostasis of the whole body.
Hepatic stellate cells in top predators of arctic animals store
20–100 times the levels normally found in other animals,
including humans. In pathological conditions such as liver
References
cirrhosis, their phenotype changes from that of star-shaped
stellate cells to that of fibroblasts or myofibroblasts. The
three-dimensional structure of the ECM can reversibly 1. Bloom W, Fawcett DW (1994) A textbook of histology, 12th edn.
Chapman & Hall, New York, pp 652–668
regulate the morphology, proliferation, and functions of the 2. Wake K (1971) “Sternzellen” in the liver: perisinusoidal cells with
stellate cells. Molecular mechanisms in the regulation of the special reference to storage of vitamin A. Am J Anat 132:429–462
13

3. Wake K (1980) Perisinusoidal stellate cells (fat-storing cells, 28. Senoo H, Stang E, Nilsson A, Kindberg GM, Berg T, Roos N,
interstitial cells, lipocytes), their related structure in and around Norum KR, Blomhoff R (1990) Internalization of retinol-binding
the liver sinusoids, and vitamin A-storing cells in extrahepatic protein in parenchymal and stellate cells of rat liver. J Lipid Res
organs. Int Rev Cytol 66:303–353 31:1229–1239
4. Kojima N, Sato M, Suzuki A, Sato T, Satoh S, Kato T, Senoo H 29. Båvik CO, Eriksson U, Allen RA, Peterson PA (1991)
(2001) Enhanced expression of B7-1, B7-2, and intercellular adhe- Identification and partial characterization of a retinal pigment
sion molecule 1 in sinusoidal endothelial cells by warm ischemia/ epithelial membrane receptor for plasma retinol-binding protein.
reperfusion injury in rat liver. Hepatology 34:751–757 J Biol Chem 266:14978–14985
5. Steiniger B, Klempnauer J, Wonigeit K (1984) Phenotype 30. Båvik C-O, Busch C, Eriksson U (1992) Characterization
and histological distribution of interstitial dendritic cells in the of a plasma retinol-binding protein membrane receptor expressed
rat pancreas, liver, heart, and kidney. Transplantation 38:169– in the retinal pigment epithelium. J Biol Chem 267:23035–23042
175 31. Båvik C-O, Lévy F, Hellman U, Wernstedt C, Eriksson U (1993)
6. Prickett TCR, McKenzie JL, Hart DNJ (1988) Characterization The retinal pigment epithelial membrane receptor for plasma
of interstitial dendritic cells in human liver. Transplantation retinol-binding protein. J Biol Chem 268:20540–20546
46:754–761 32. Andersen KB, Nilsson A, Blomhoff HK, Øyen TB, Gabrielsen
7. Banchereau J, Steinman RM (1998) Dendritic cells and the OS, Norum KR, Blomhoff R (1992) Direct mobilization of retinol
control of immunity. Nature (Lond) 392:245–252 from hepatic perisinusoidal stellate cells to plasma. J Biol Chem
8. Blomhoff R, Wake K (1991) Perisinusoidal stellate cells of the 267:1340–1344
liver: important roles in retinol metabolism and fibrosis. FASEB 33. Rodahl K (1949) Toxicity of polar bear liver. Nature (Lond)
J 5:271–277 164:530–531
9. Senoo H, Sato M, Imai K (1997) Hepatic stellate cells: from the 34. Rodahl K (1949) Vitamin sources in Arctic regions. Nor Polarinst
viewpoint of retinoid handling and function of the extracellular Skr 91:1–64
matrix. Acta Anat Nippon 72:79–94 35. Rodahl K, Moore T (1943) The vitamin A content and toxicity of
10. Sato M, Suzuki S, Senoo H (2003) Hepatic stellate cells: unique bear and seal liver. Biochem J 37:166–168
characteristics in cell biology and phenotype. Cell Struct Funct 36. Senoo H, Wake K, Imai K, Kojima N, Matano Y, Miura M,
28:105–112 Sato M, Roos N, Berg T, Norum KR, Blomhoff R (1999) Vitamin
11. Nagy NE, Holven KB, Roos N, Senoo H, Kojima N, Norum KR, A-storing system in mammals and birds in Arctic area: a study in
Blomhoff R (1997) Storage of vitamin A in extrahepatic stellate the Svalbard archipelago. Cells Hepatic Sinusoid 7:34–35
cells in normal rats. J Lipid Res 38:645–658 37. Senoo H, Imai K, Higashi N, Wake K, Kojima N, Miura M, Wold
12. Matano Y, Miura M, Kojima N, Sato M, Imai K, Senoo H (1999) HL, Moskaug JØ, Sato T, Sato M, Roos N, Berg T, Norum KR,
Hepatic stellate cells and extrahepatic stellate cells (extrahepatic Blomhoff R (2001) Transport and storage of vitamin A in arctic
vitamin A-storing cells). Cells Hepatic Sinusoid 7:26–27 animals. Cells Hepatic Sinusoid 8:207–209
13. Blomhoff R (1994) Vitamin A in health and disease. Dekker, 38. Higashi N, Senoo H (2003) Distribution of vitamin A-storing lipid
New York droplets in hepatic stellate cells in liver lobules: a comparative
14. Chawla A, Repa JJ, Evans RM, Mangelsdorf DJ (2001) Nuclear study. Anat Rec Part A 271:240–248
receptors and lipid physiology: opening the X-files. Science 39. Higashi N, Imai K, Sato M, Sato T, Kojima N, Miura M, Wold
294:1866–1870 HL, Moskaug JØ, Berg T, Norum KR, Roos N, Wake K,
15. Blomhoff R, Green MH, Berg T, Norum KR (1990) Transport Blomhoff R, Senoo H (2003) Intralobular distribution of vitamin
and storage of vitamin A. Science 250:399–404 A-storing lipid droplets in hepatic stellate cells with special refer-
16. Blomhoff R, Green MH, Green JB, Berg T, Norum KR (1992) ence to polar bear and arctic fox. Comp Hepatol (in press)
Vitamin A metabolism: new perspectives on absorption, trans- 40. Jarman WM, Simon M, Norstrom RJ, Burns SA, Bacon CA,
port, and storage. Physiol Rev 71:951–990 Simonelt BRT, Risenbrough RW (1992) Global distribution of
17. Blomhoff R, Green MH, Norum KR (1992) Vitamin A: physi- Tris(4-chlorophenyl)methanol in high trophic level birds and
ological and biochemical processing. Annu Rev Nutr 12:37–57 mammals. Environ Sci Technol 26:1770–1774
18. Senoo H (2000) Digestion, metabolism. In: Krinke GJ (ed) The 41. Barrie LA, Gregor D, Hargrave B, Lake R, Muir D, Shearer R,
digital handbook of experimental laboratory animals: the rat. Tracey N, Bidleman T (1992) Arctic contaminants: sources,
Academic Press, London, pp 359–383 occurrence and pathways. Sci Total Environ 122:1–74
19. Senoo H, Hata R, Nagai Y, Wake K (1984) Stellate cells (vitamin 42. Muir DCG, Wagemann R, Hargrave BT, Thomas DJ, Peakall
A-storing cells) are the primary site of collagen synthesis in non- DB, Norstrom RJ (1998) Arctic marine ecosystem contamination.
parenchymal cells in the liver. Biomed Res 5:451–458 Sci Total Environ 122:75–134
20. Senoo H, Hata R (1993) Isolation of perisinusoidal stellate cells 43. Holden C (1998) Polar bears and PCBs. Science 280:2053
(vitamin A-storing cells, fat-storing cells) of the liver. Connect 44. Wiig Ø, Derocher AE, Cronin MM, Skaare JU (1998) Female
Tissue 25:129–137 pseudohermaphrodite polar bears at Svalbard. J Wildl Dis
21. Gyøen T, Bjerkelund T, Blomhoff HK, Norum KR, Berg T, 34:792–796
Blomhoff R (1987) Liver takes up retinol-binding protein from 45. Skaare JU, Bernhoft A, Wiig Ø, Norum KR, Haug E, Eide DM,
plasma. J Biol Chem 262:10926–10930 Derocher AE (2001) Relationship between plasma levels of orga-
22. Senoo H, Smeland S, Malaba L, Bjerknes T, Stang E, Roos N, nochlorines, retinol and thyroid hormones from polar bears
Berg T, Norum KR, Blomhoff R (1993) Transfer of retinol-bind- (Ursus maritimus) at Svalbard. J Toxicol Environ Health 62:227–
ing protein from HepG2 human hepatoma cells to cocultured rat 241
stellate cells. Proc Natl Acad Sci USA 90:3616–3620 46. Dewailly E, Nantel A, Weber J-P, Meyer F (1989) High levels of
23. Malaba L, Smeland S, Senoo H, Norum KR, Berg T, Blomhoff R, PCBs in breast milk of Inuit women from arctic Quebec. Bull
Kindberg GM (1996) Retinol-binding protein and asialo- Environ Contam Toxicol 43:641–646
orosomucoid are taken up by different pathways in liver cells. 47. Sato M, Miura M, Kojima N, Higashi N, Imai K, Sato T, Wold
J Biol Chem 270:15686–15692 HL, Moskaug JØ, Blomhoff R, Wake K, Roos N, Berg T, Norum
24. Smeland S, Bjerknes T, Malaba L, Eskild W, Norum KR, KR, Senoo H (2001) Nuclear deviation in heptic parenchymal
Blomhoff R (1995) Tissue distribution of the receptor for plasma cells on sinusoidal surface in arctic animals. Cell Struct Funct
retinol-binding protein. Biochem J 305:419–424 26:71–77
25. Senoo H, Smeland S, Stang E, Roos N, Berg T, Norum KR, 48. Higashi N, Senoo H, Wold HL, Moskaug JØ, Blomhoff R (2003)
Blomhoff R (1993) Stellate cells take up retinol-binding protein. Destruction of Glisson’s sheath in the liver of polar fox. Comp
Cells Hepatic Sinusoid 4:423–425 Hepatol (in press)
26. Blomhoff R, Senoo H, Smeland S, Bjerknes T, Norum KR (1992) 49. Senoo H, Wake K, Wold HL, Higashi N, Imai K, Moskaug JØ,
Cellular uptake of vitamin A. J Nutr Sci Vitaminol 38:327–330 Kojima N, Miura M, Sato T, Sato M, Roos N, Berg T, Norum KR,
27. Senoo H, Stang E, Kindberg GM, Berg T, Roos N, Norum KR, Blomhoff R (2004) Decreased capacity for vitamin A storage in
Blomhoff R (1989) Uptake of retinol-binding protein in liver hepatic stellate cells in arctic animals. Comp Hepatol 3(suppl 1):
cells. Cells Hepatic Sinusoid 2:29–32 S18
14

50. Majno G (1979) The story of the myofibroblasts. Am J Surg 73. Davis BH, Vucic A (1989) Modulation of vitamin A metabolism
Pathol 6:535–542 during hepatic and intestinal culture. Biochim Biophys Acta
51. Senoo H, Wake K (1985) Suppression of experimental hepatic 1010:318–324
fibrosis by administration of vitamin A. Lab Invest 52:182– 74. Friedman SL, Roll FJ, Boyles J, Arenson DM, Bissell DM (1989)
194 Maintenance of differentiated phenotype of cultured rat hepatic
52. Davis BH, Chen A, Beno DWA (1996) Raf and mitogen- lipocytes by basement membrane matrix. J Biol Chem 264:10756–
activated protein kinase regulate stellate cell collagen gene ex- 1076275
pression. J Biol Chem 271:11039–11042 75. Friedman SL, Roll FJ, Boyles J, Bissell DM (1990) Autocrine
53. Imai K, Sato M, Kojima N, Miura M, Sato T, Sugiyama T, regulation of lipocyte matrix production: activation by extracellu-
Enomoto K, Senoo H (2000) Storage of lipid droplets in and lar matrix. Cells Hepatic Sinusoid 2:61–63
production of extracellular matrix by hepatic stellate cells (vita- 76. Davis BH, Pratt BM, Madri JA (1987) Retinol and extracellular
min A-storing cells) in Long-Evans cinnamon-like colored (LEC) collagen matrices modulate hepatic Ito cell collagen pheno-
rats. Anat Rec 258:338–348 type and cellular retinol binding protein levels. J Biol Chem
54. Lindsay CK, Thorgeirsson UP (1995) Localization of messenger 262:10280–10286
RNA for tissue inhibitor of metalloproteinases-1 and type IV 77. Davis BH (1988) Transforming growth factor-β responsiveness is
collagenases/gelatinases in monkey hepatocellular carcinomas. modulated by extracellular collagen matrix during hepatic Ito cell
Clin Exp Metastas 13:381–388 culture. J Cell Physiol 136:547–553
55. Montfort I, Perez-Tamayo R, Alvizouli AM, Tello E (1990) Col- 78. Senoo H, Imai K, Sato M, Kojima N, Miura M, Hata R (1996)
lagenase of hepatocytes and sinusoidal liver cells in the Three-dimensional structure of extracellular matrix reversibly
reversibility of experimental cirrhosis of the liver. Virchows Arch regulates morphology, proliferation and collagen metabolism of
B Cell Pathol 59:281–289 perisinusoidal stellate cells (vitamin A-storing cells). Cell Biol Int
56. Benyon RC, Arthur MJP (2001) Extracellular matrix degradation 20:501–512
and the role of hepatic stellate cells. Semin Liver Dis 21:373–384 79. Imai K, Sato M, Kojima N, Miura M, Matano Y, Senoo H (1997)
57. Arthur MJP (2002) Reversibility of liver fibrosis and cirrhosis Three-dimensional morphological changes in cultured hepatic
following treatment for hepatitis C. Gastroenterology 122:1525– stellate cells by type I collagen fibers. Cells Hepatic Sinusoid
1528 6:105–106
58. Poynard T, Mchutchison J, Manns M, Trepo C, Lindsay K, 80. Senoo H, Imai K, Matano Y, Sato M (1998) Molecular mecha-
Goodman Z, Ling M-H, Albrecht J (2002) Impact of pegylated nisms in the reversible regulation of morphology, proliferation
interferon alfa-2b and ribavirin on liver fibrosis in patients with and collagen metabolism in hepatic stellate cells by the three-
chronic hepatitis C. Gastroenterology 122:1303–1313 dimensional structure of the extracellular matrix. J Gastroenterol
59. Iredale JP, Benyon RC, Pickering J, McCullen M, Northrop M, Hepatol 13(suppl):S19–S32
Pawley S, Hovell C, Arthur MJP (1998) Mechanisms of spontane- 81. Imai K, Senoo H (1998) Morphology of sites of adhesion be-
ous resolution of rat liver fibrosis. Hepatic stellate cell apoptosis tween hepatic stellate cells (vitamin A-storing cells) and a three-
and reduced hepatic expression of metalloproteinase inhibitors. dimensional extracellular matrix. Anat Rec 250:430–437
J Clin Invest 102:538–549 82. Kojima N, Sato M, Imai K, Miura M, Matano Y, Senoo H (1998)
60. Pinzani M, Marra F (2001) Cytokine receptors and signaling in Hepatic stellate cells (vitamin A-storing cells) change their cy-
hepatic stellate cells. Semin Liver Dis 21:397–416 toskeleton structure by extracellular matrix components through
61. Friedman SL (2000) Molecular regulation of hepatic fibrosis, a signal transduction system. Histochem Cell Biol 110:121–128
an integrated cellular response to tissue injury. J Biol Chem 83. Kojima N, Sato M, Miura M, Imai K, Senoo H (1999) Alteration
275:2247–2250 in distribution of focal adhesion components by signaling inhibi-
62. Friedman SL, Arthur MJP (2002) Reversing hepatic fibrosis. Sci tors in hepatic stellate cells and fibroblasts cultured on type I
Med 8:194–205 collagen gel. Cells Hepatic Sinusoid 7:24–25
63. Li Y-L, Sato M, Kojima N, Miura M, Senoo H (1999) Regulatory 84. Imai K, Senoo H (2000) Morphology of sites of adhesion between
role of extracellular matrix components in expression of matrix extracellular matrix and hepatic stellate cells. Connect Tissue
metalloproteases in cultured hepatic stellate cells. Cell Struct 32:395–400
Funct 24:255–261 85. Imai K, Sato T, Senoo H (2000) Adhesion between cells and
64. Wang D-R, Sato M, Sato T, Kojima N, Imai K, Higashi N, Senoo extracellular matrix with special reference to hepatic stellate cell
H (2004) Regulation of matrix metallo-proteinase expression by adhesion to three-dimensional collagen fibers. Cell Struct Funct
extracellular matrix components in cultured hepatic stellate cells. 25:329–336
Comp Hepatol 3(suppl 1):S20 86. Sato M, Imai K, Kojima N, Miura M, Senoo H (1997) Long
65. Mabuchi A, Mullaney I, Sheard PA, Zimmermann A, Senoo H, cellular processes of hepatic stellate cells cultured on or in type I
Wheatley AM (2003) Role of hepatic stellate cells in the early collagen gel. Cells Hepatic Sinusoid 6:85–89
phase of liver regeneration in rat: formation of tight adhesion to 87. Sato M, Kojima N, Miura M, Imai K, Senoo H (1998) Induction of
parenchymal cells. Comp Hepatol 3(suppl 1):S29 cellular processes containing collagenase and retinoid by integrin-
66. Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter, P binding to interstitial collagen in hepatic stellate cell culture. Cell
(2002) Molecular biology of the cell, 4th edn. Garland, New York, Biol Int 22:115–125
pp 1090–1118 88. Sato M, Kojima N, Miura M, Imai K, Senoo H (1999) Intracellular
67. Senoo H, Hata R, Wake K, Nagai Y (1991) Isolation and serum signaling for process elongation in cultured hepatic stellate cells
free culture of stellate cells. Cells Hepatic Sinusoid 3:259–262 on type I collagen gel. Cells Hepatic Sinusoid 7:32–33
68. Hata R, Senoo H (1992) Extracellular matrix system regulates cell 89. Sato M, Sato T, Kojima N, Miura M, Imai K, Senoo H (2001)
growth, tissue formation, and cellular functions. Tissue Cult Res Induction of cellular process elongation mediated by microtu-
Commun 11:337–343 bule-associated protein 2 in hepatic stellate cells cultured on type
69. Senoo H, Hata R (1993) Tissue formation and extracellular I collagen gel. Cells Hepatic Sinusoid 8:205–206
matrix system: cellular devices for adhesion to extracellular 90. Sato M, Sato T, Kojima N, Imai K, Higashi N, Wang D-R, Senoo
matrix. Tissue Cult Res Commun 12:237–245 H (2004) 3-D structure of extracellular matrix regulates gene
70. Senoo H, Hata R (1994) Extracellular matrix regulates cell mor- expression in cultured stellate cells to induce process elongation.
phology, proliferation, and tissue formation. Acta Anat Nippon Comp Hepatol 3(suppl 1):S4
69:719–733 91. Miura M, Sato M, Toyoshima I, Senoo H (1997) Extension of
71. Senoo H, Hata R (1994) Extracellular matrix regulates and l- long cellular processes of hepatic stellate cells cultured on extra-
ascorbic acid 2-phosphate further modulates morphology, prolif- cellular type I collagen gel by microtubule assembly: observation
eration, and collagen synthesis of the perisinusoidal stellate cells. utilizing time-lapse video-microscopy. Cell Struct Funct 22:487–
Biochem Biophys Res Commun 200:999–1006 492
72. Kleinman HK, McGarvey ML, Hassell JR, Star VL, Cannon FB, 92. Cukierman E, Pankov R, Stevens DR, Yamada KM (2001) Tak-
Laurie GW, Martin GR (1986) Basement membrane complexes ing cell-matrix adhesion to the third dimension. Science 294:1708–
with biological activity. Biochemistry 25:312–318 1712
15

93. Grinnell F, Ho C-H, Tamariz E, Lee DJ, Skuta G (2003) 99. Bachem MG, Schneider E, Gross H, Weidenbach H, Schmid RM,
Dendritic fibroblasts in three-dimensional collagen matrices. Mol Menke A, Siech M, Beger H, Grnert A, Adler G (1998)
Biol Cell 14:384–395 Identification, culture, and characterization of pancreatic stellate
94. Emsley J, Knight CG, Farndale RW, Barnes MJ, Liddington RC cells in rats and humans. Gastroenterology 115:421–432
(2000) Structural basis of collagen recognition by integrin α2β1. 100. Wells RG, Crawford JM (1998) Pancreatic stellate cells. The new
Cell 101:47–56 stars of chronic pancreatitis? Gastroenterology 115:491–493
95. Hata R, Senoo H (1989) l-Ascorbic acid 2-phosphate stimulates 101. Masamune A, Kikuta K, Satoh M, Sakai Y, Satoh A,
collagen accumulation, cell proliferation, and formation of a Shimosegawa T (2002) Ligands of peroxisome proliferator-
three-dimensional tissue-like substance by skin fibroblast. J Cell activated receptor-γ block activation of pancreatic stellate cells.
Physiol 138:8–16 J Biol Chem 277:141–147
96. Kurata S, Senoo H, Hata R (1993) Transcriptional activation of 102. Bachem MG, Schmid-Kotsas A, Schuenemann M, Fundel M,
type I collagen genes by ascorbic acid 2-phosphate in human skin Adler G, Menke A, Siech M, Buck A, Gruenert A (2002) The
fibroblasts and its failure in cells from a patient with α2(I)-chain- increased deposition of connective tissue in pancreas carcinoma is
defective Ehlers–Danlos syndrome. Exp Cell Res 206:63–71 the result of a paracrine stimulation of pancreatic stellate cells by
97. Senoo H, Tsukada Y, Sato T, Hata R (1989) Co-culture of cancer cells. Mol Biol Cell 13:4a
fibroblasts and hepatic parenchymal cells induces metabolic 103. Bachem MG, Schmid-Kotsas A, Buck A, Siech M, Gruenert A
changes and formation of a three-dimensional structure. Cell Biol (2002) An TGFβ1-antisense oligonucleotide reduces autocrine
Int Rep 13:197–206 stimulated TGFβ1-, fibronectin- and collagen type I-synthesis of
98. Apte MV, Haber PS, Applegate TL, Norton ID, McCaughan cultured pancreatic stellate cells. Mol Biol Cell 13:345a
GW, Korsten MA, Pirola RC, Wilson JS (1998) Periacinar stellate
shaped cells in rat pancreas: identification, isolation, and culture.
Gut 43:128–133

You might also like