201
|
Liver development in zebrafish (Danio rerio). J Genet Genomics 2009; 36:325-34. [PMID: 19539242 DOI: 10.1016/s1673-8527(08)60121-6] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 04/17/2009] [Accepted: 04/17/2009] [Indexed: 12/17/2022]
Abstract
Liver is one of the largest internal organs in the body and its importance for metabolism, detoxification and homeostasis has been well established. In this review, we summarized recent progresses in studying liver initiation and development during embryogenesis using zebrafish as a model system. We mainly focused on topics related to the specification of hepatoblasts from endoderm, the formation and growth of liver bud, the differentiation of hepatocytes and bile duct cells from hepatoblasts, and finally the role of mesodermal signals in controlling liver development in zebrafish.
Collapse
|
202
|
Huppert SS, Magnuson MA. New complexity in differentiating stem cells toward hepatic and pancreatic fates. Sci Signal 2009; 2:pe50. [PMID: 19671927 DOI: 10.1126/scisignal.283pe50] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The differentiation of hepatic and pancreatic progenitor cells during embryogenesis is determined by inductive factors secreted by neighboring cells. These factors stimulate and repress the expression of key regulatory genes in progenitor cells, thereby establishing unique genetic programs that determine cell fate. The signaling network that controls liver and pancreas development is highly dynamic with respect to both concentration and timing of exposure to several key inductive factors. Not only do large changes occur within short time frames, multiple signaling pathways also converge on the same target genes. Given the intense effort under way to generate certain differentiated cell types from both embryonic and induced pluripotent stem cells, greater understanding of how different inductive signals interact with each other may be essential for the eventual success of such efforts.
Collapse
Affiliation(s)
- Stacey S Huppert
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | |
Collapse
|
203
|
Matsuura K, Katsumoto K, Fukuda K, Kume K, Kume S. Conserved origin of the ventral pancreas in chicken. Mech Dev 2009; 126:817-27. [PMID: 19651207 DOI: 10.1016/j.mod.2009.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 07/25/2009] [Accepted: 07/28/2009] [Indexed: 11/17/2022]
Abstract
To determine the origin of the ventral pancreas, a fate map of the ventral pancreas was constructed using DiI crystal or CM-DiI to mark regions of the early chick endoderm: this allowed correlations to be established between specific endoderm sites and the positions of their descendants. First, the region lateral to the 7- to 9-somite level, which has been reported to contribute to the ventral pancreas, was shown to contribute mainly to the intestine or the dorsal pancreas. At the 10 somite stage (ss), the ventral pre-pancreatic cells reside laterally at the 2-somite level, at the lateral boarder of the somite. At this stage, however, the fate of these cells has not yet segregated and they contribute to the ventral pancreas and to the intestine or bile duct. The ventral pancreas fate segregated at the 17 ss; the cells residing at the somite boarder at the 4-somite level at the 17 ss were revealed to contribute to the ventral pancreas. Interestingly, the dorsal and the ventral pancreatic buds are different in both origin and function. These two pancreatic buds begin to fuse at day 7 (HH 30) of embryonic development. However, whereas the dorsal pancreas gives rise to both Insulin-expressing endocrine and Amylase-expressing exocrine cells, the ventral pancreas gives rise to Amylase-expressing exocrine cells, but not insulin-expressing endocrine cells before day 7 (HH 30) of embryonic development.
Collapse
Affiliation(s)
- Kumi Matsuura
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto 860-0811, Japan
| | | | | | | | | |
Collapse
|
204
|
Spence JR, Lange AW, Lin SCJ, Kaestner KH, Lowy AM, Kim I, Whitsett JA, Wells JM. Sox17 regulates organ lineage segregation of ventral foregut progenitor cells. Dev Cell 2009; 17:62-74. [PMID: 19619492 PMCID: PMC2734336 DOI: 10.1016/j.devcel.2009.05.012] [Citation(s) in RCA: 225] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 03/28/2009] [Accepted: 05/04/2009] [Indexed: 11/29/2022]
Abstract
The ventral pancreas, biliary system, and liver arise from the posterior ventral foregut, but the cell-intrinsic pathway by which these organ lineages are separated is not known. Here we show that the extrahepatobiliary system shares a common origin with the ventral pancreas and not the liver, as previously thought. These pancreatobiliary progenitor cells coexpress the transcription factors PDX1 and SOX17 at E8.5 and their segregation into a PDX1+ ventral pancreas and a SOX17+ biliary primordium is Sox17-dependent. Deletion of Sox17 at E8.5 results in the loss of biliary structures and ectopic pancreatic tissue in the liver bud and common duct, while Sox17 overexpression suppresses pancreas development and promotes ectopic biliary-like tissue throughout the PDX1+ domain. Restricting SOX17+ biliary progenitor cells to the ventral region of the gut requires the notch effector Hes1. Our results highlight the role of Sox17 and Hes1 in patterning and morphogenetic segregation of ventral foregut lineages.
Collapse
Affiliation(s)
- Jason R. Spence
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039
| | - Alex W. Lange
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039
| | - Suh-Chin J. Lin
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039
| | - Klaus H. Kaestner
- School of Medicine Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6145
| | - Andrew M. Lowy
- Department of Surgery, UCSD Medical Center, La Jolla, CA 92093-0658
| | - Injune Kim
- Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea 306-701
| | - Jeffrey A. Whitsett
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039
| | - James M. Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039
| |
Collapse
|
205
|
Livigni A, Villegas SN, Oikonomopoulou I, Rahman A, Morrison GM, Brickman JM. Differentiation of embryonic stem cells into anterior definitive endoderm. CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2009; Chapter 1:Unit 1G.3. [PMID: 19585461 DOI: 10.1002/9780470151808.sc01g03s10] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Anterior definitive endoderm (ADE) is both an important embryonic signaling center and a unique multipotent precursor of liver, pancreas, and other visceral organs. Here we describe a method for the differentiation of mouse embryonic stem (ES) cells to endoderm with pronounced anterior character. ADE-containing cultures can be produced in vitro by suspension (aggregation or embryoid body) culture and in a serum-free adherent monolayer culture. Purified ES cell-derived ADE cells appear committed to endodermal fates and can undergo further differentiation in vitro towards liver and pancreas with enhanced efficiency.
Collapse
Affiliation(s)
- Alessandra Livigni
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | | | | |
Collapse
|
206
|
Wandzioch E, Zaret KS. Dynamic signaling network for the specification of embryonic pancreas and liver progenitors. Science 2009; 324:1707-10. [PMID: 19556507 PMCID: PMC2771431 DOI: 10.1126/science.1174497] [Citation(s) in RCA: 177] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Studies of the formation of pancreas and liver progenitors have focused on individual inductive signals and cellular responses. Here, we investigated how bone morphogenetic protein, transforming growth factor-beta (TGFbeta), and fibroblast growth factor signaling pathways converge on the earliest genes that elicit pancreas and liver induction in mouse embryos. The inductive network was found to be dynamic; it changed within hours. Different signals functioned in parallel to induce different early genes, and two permutations of signals induced liver progenitor domains, which revealed flexibility in cell programming. Also, the specification of pancreas and liver progenitors was restricted by the TGFbeta pathway. These findings may enhance progenitor cell specification from stem cells for biomedical purposes and can help explain incomplete programming in stem cell differentiation protocols.
Collapse
Affiliation(s)
- Ewa Wandzioch
- Cell and Developmental Biology Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | | |
Collapse
|
207
|
Séguin CA, Draper JS, Nagy A, Rossant J. Establishment of endoderm progenitors by SOX transcription factor expression in human embryonic stem cells. Cell Stem Cell 2009; 3:182-95. [PMID: 18682240 DOI: 10.1016/j.stem.2008.06.018] [Citation(s) in RCA: 166] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Revised: 05/06/2008] [Accepted: 06/19/2008] [Indexed: 12/25/2022]
Abstract
In this study, we explore endoderm cell fate regulation through the expression of lineage-determining transcription factors. We demonstrate that stable endoderm progenitors can be established from human ES cells by constitutive expression of SOX7 or SOX17, producing extraembryonic endoderm and definitive endoderm progenitors, respectively. In teratoma assays and growth factor-mediated differentiation, SOX7 cells appear restricted to the extraembryonic endoderm, and SOX17 cells demonstrate a mesendodermal phenotype in teratomas and the ability to undergo endoderm maturation in vitro in the absence of cytokine-mediated endoderm induction. These endoderm progenitor cells maintain a stable phenotype through many passages in culture, thereby providing new tools to explore the pathways of endoderm differentiation.
Collapse
Affiliation(s)
- Cheryle A Séguin
- The Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON M5G 1L7, Canada.
| | | | | | | |
Collapse
|
208
|
Motoyama H, Ogawa S, Kubo A, Miwa S, Nakayama J, Tagawa YI, Miyagawa S. In vitro reprogramming of adult hepatocytes into insulin-producing cells without viral vectors. Biochem Biophys Res Commun 2009; 385:123-8. [PMID: 19422803 DOI: 10.1016/j.bbrc.2009.04.146] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2009] [Accepted: 04/28/2009] [Indexed: 12/18/2022]
Abstract
The pancreas and the liver share the same endodermal origin. We have been studying whether mature hepatocytes can be induced to differentiate into pancreatic beta-cells by in vitro delivery of transcriptional factors using a non-viral approach. Here we showed that nucleofection allowed suitable transfection of primary hepatocytes employing various non-viral methods. We introduced either pancreatic and duodenal homeobox 1 (Pdx1) or neurogenin 3 (Ngn3), or both, into the mature cells using nucleofection. Co-expression of pdx1 and ngn3 using a bicistronic vector activated the transcription of various islet-related genes, and the transfected hepatocytes acquired the ability to synthesize and secrete insulin. Our results suggest that simultaneous expression of Pdx1 and Ngn3 is an excellent inducer of liver-to-pancreas reprogramming, and that reprogramming will occur even in mature somatic cells without the need for viral vectors. These findings are of considerable significance for further therapeutic development for various intractable diseases including diabetes.
Collapse
Affiliation(s)
- Hiroaki Motoyama
- Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | | | | | | | | | | | | |
Collapse
|
209
|
Kameda Y, Ito M, Nishimaki T, Gotoh N. FRS2alpha is required for the separation, migration, and survival of pharyngeal-endoderm derived organs including thyroid, ultimobranchial body, parathyroid, and thymus. Dev Dyn 2009; 238:503-13. [PMID: 19235715 DOI: 10.1002/dvdy.21867] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The docking protein FRS2alpha plays an important role in fibroblast growth factor (FGF)-induced intracellular signal transduction by linking FGF receptors (FGFRs) to a variety of intracellular signaling pathways. In FRS2alpha(2F/2F) mutant mice at embryonic day (E)18.5, in which the Shp2-binding sites of FRS2alpha were disrupted, the thyroid glands were aplastic or hypoplastic. C cells were absent or present in low numbers and rarely formed a compact mass of cells. Parathyroid glands were mostly connected to thymus tissues. At E10.5, the formations of pharyngeal pouches and thyroid primordium were normally initiated in the mutant mice. At E11.5 to E12.5, the thyroid primordium of wild-type embryos was located close to the aortic sac, and the epithelial buds of pharyngeal-derived organs, including the parathyroid gland, thymus and ultimobranchial body, were separated from the epithelium and began to migrate to their final destinations. In the FRS2alpha(2F/2F) mutants, however, the thyroid primordium became hypoplastic and the pharyngeal-derived organ primordia remained affiliated with the pharyngeal epithelium. At these stages, organ-specific differentiation markers (i.e., Nkx2-1/TTF1 for the thyroid lobe and ultimobranchial body; Pax8 for the thyroid lobe; parathormone (PTH), chromogranin A, P75(NTR), and S100 protein for the parathyroid gland; and p63 for the thymus) were normally expressed in the mutant tissues. Thus, the separation, migration, and survival of the pharyngeal organs were impaired in the FRS2alpha(2F/2F) mutants.
Collapse
Affiliation(s)
- Yoko Kameda
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan.
| | | | | | | |
Collapse
|
210
|
Borowiak M, Maehr R, Chen S, Chen AE, Tang W, Fox JO, Schreiber SL, Melton DA. Small molecules efficiently direct endodermal differentiation of mouse and human embryonic stem cells. Cell Stem Cell 2009; 4:348-58. [PMID: 19341624 PMCID: PMC4564293 DOI: 10.1016/j.stem.2009.01.014] [Citation(s) in RCA: 326] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 12/02/2008] [Accepted: 01/16/2009] [Indexed: 12/14/2022]
Abstract
An essential step for therapeutic and research applications of stem cells is the ability to differentiate them into specific cell types. Endodermal cell derivatives, including lung, liver, and pancreas, are of interest for regenerative medicine, but efforts to produce these cells have been met with only modest success. In a screen of 4000 compounds, two cell-permeable small molecules were indentified that direct differentiation of ESCs into the endodermal lineage. These compounds induce nearly 80% of ESCs to form definitive endoderm, a higher efficiency than that achieved by Activin A or Nodal, commonly used protein inducers of endoderm. The chemically induced endoderm expresses multiple endodermal markers, can participate in normal development when injected into developing embryos, and can form pancreatic progenitors. The application of small molecules to differentiate mouse and human ESCs into endoderm represents a step toward achieving a reproducible and efficient production of desired ESC derivatives.
Collapse
Affiliation(s)
- Malgorzata Borowiak
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - René Maehr
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Shuibing Chen
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Alice E. Chen
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Weiping Tang
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Julia O. Fox
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Stuart L. Schreiber
- Howard Hughes Medical Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, MA 02142, USA
- Department of Chemistry and Chemical Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Douglas A. Melton
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| |
Collapse
|
211
|
Katsumoto K, Fukuda K, Kimura W, Shimamura K, Yasugi S, Kume S. Origin of pancreatic precursors in the chick embryo and the mechanism of endoderm regionalization. Mech Dev 2009; 126:539-51. [PMID: 19341795 DOI: 10.1016/j.mod.2009.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2008] [Revised: 03/18/2009] [Accepted: 03/23/2009] [Indexed: 11/16/2022]
Abstract
To study the developmental origin of the pancreas we used DiI crystals to mark regions of the early chick endoderm: this allowed correlations to be established between specific endoderm sites and the positions of their descendants. Endodermal precursor cells for the stomach, pancreas and intestine were found to segregate immediately after completion of gastrulation. Transplantation experiments showed that region-specific endodermal fates are determined sequentially in the order stomach, intestine, and then pancreas. Non-pancreatic endoderm transplanted to the stomach region generated ectopic pancreas expressing both insulin and glucagon. These results imply that a pancreas-inducing signal is emitted from somitic mesoderm underlying the pre-pancreatic region, and this extends rostrally beyond the stomach endoderm region at the early somite stage. Transplantation experiments revealed that the endoderm responding to these pancreatic-inducing signals lies within the pre-pancreatic region and extends caudally beyond the region of the intestinal endoderm. The results indicate that pancreatic fate is determined in the area of overlap between these two regions.
Collapse
Affiliation(s)
- Keiichi Katsumoto
- Division of Stem Cell Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Honjo Kumamoto, Japan
| | | | | | | | | | | |
Collapse
|
212
|
Johannesson M, Ståhlberg A, Ameri J, Sand FW, Norrman K, Semb H. FGF4 and retinoic acid direct differentiation of hESCs into PDX1-expressing foregut endoderm in a time- and concentration-dependent manner. PLoS One 2009; 4:e4794. [PMID: 19277121 PMCID: PMC2651644 DOI: 10.1371/journal.pone.0004794] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Accepted: 02/10/2009] [Indexed: 01/27/2023] Open
Abstract
Background Retinoic acid (RA) and fibroblast growth factor 4 (FGF4) signaling control endoderm patterning and pancreas induction/expansion. Based on these findings, RA and FGFs, excluding FGF4, have frequently been used in differentiation protocols to direct differentiation of hESCs into endodermal and pancreatic cell types. In vivo, these signaling pathways act in a temporal and concentration-dependent manner. However, in vitro, the underlying basis for the time of addition of growth and differentiation factors (GDFs), including RA and FGFs, as well as the concentration is lacking. Thus, in order to develop robust and reliable differentiation protocols of ESCs into mature pancreatic cell types, including insulin-producing β cells, it will be important to mechanistically understand each specification step. This includes differentiation of mesendoderm/definitive endoderm into foregut endoderm- the origin of pancreatic endoderm. Methodology/Principal Findings Here, we provide data on the individual and combinatorial role of RA and FGF4 in directing differentiation of ActivinA (AA)-induced hESCs into PDX1-expressing cells. FGF4's ability to affect endoderm patterning and specification in vitro has so far not been tested. By testing out the optimal concentration and timing of addition of FGF4 and RA, we present a robust differentiation protocol that on average generates 32% PDX1+ cells. Furthermore, we show that RA is required for converting AA-induced hESCs into PDX1+ cells, and that part of the underlying mechanism involves FGF receptor signaling. Finally, further characterization of the PDX1+ cells suggests that they represent foregut endoderm not yet committed to pancreatic, posterior stomach, or duodenal endoderm. Conclusion/Significance In conclusion, we show that RA and FGF4 jointly direct differentiation of PDX1+ foregut endoderm in a robust and efficient manner. RA signaling mediated by the early induction of RARβ through AA/Wnt3a is required for PDX1 expression. Part of RA's activity is mediated by FGF signaling.
Collapse
MESH Headings
- Activins/pharmacology
- Cell Differentiation/drug effects
- Cells, Cultured/drug effects
- Cells, Cultured/metabolism
- Dose-Response Relationship, Drug
- Drug Synergism
- Embryonic Stem Cells/cytology
- Embryonic Stem Cells/drug effects
- Embryonic Stem Cells/metabolism
- Endoderm/cytology
- Endoderm/drug effects
- Endoderm/metabolism
- Fibroblast Growth Factor 4/physiology
- Gene Expression Regulation/drug effects
- Homeodomain Proteins/biosynthesis
- Homeodomain Proteins/genetics
- Humans
- Pancreas/cytology
- Pancreas/embryology
- Pyrroles/pharmacology
- RNA, Messenger/biosynthesis
- Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 2/biosynthesis
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/physiology
- Receptors, Retinoic Acid/biosynthesis
- Receptors, Retinoic Acid/genetics
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Time Factors
- Trans-Activators/biosynthesis
- Trans-Activators/genetics
- Tretinoin/pharmacology
- Up-Regulation/drug effects
- Wnt Proteins/physiology
- Wnt3 Protein
- Wnt3A Protein
Collapse
Affiliation(s)
- Martina Johannesson
- Lund Center for Stem Cell Biology and Cell Therapy, Lund University, Lund, Sweden
| | - Anders Ståhlberg
- Lund Center for Stem Cell Biology and Cell Therapy, Lund University, Lund, Sweden
- Department of Clinical Neuroscience and Rehabilitation, Institute of Neurosciences and Physiology, Sahlgrenska Academy at Göteborg University, Göteborg, Sweden
| | - Jacqueline Ameri
- Lund Center for Stem Cell Biology and Cell Therapy, Lund University, Lund, Sweden
| | | | - Karin Norrman
- Lund Center for Stem Cell Biology and Cell Therapy, Lund University, Lund, Sweden
| | - Henrik Semb
- Lund Center for Stem Cell Biology and Cell Therapy, Lund University, Lund, Sweden
- * E-mail:
| |
Collapse
|
213
|
Abstract
Prenatal alcohol exposure is the most common environmental factor leading to congenital birth defects in the United States. Although significant progress has been made in this field, the detailed molecular pathology of fetal alcohol syndrome (FAS) remains to be determined. Previously, we have shown that alcohol exposure perturbs hedgehog signal transduction in zebrafish embryos by inhibiting the post-translational cholesterol modification of Sonic hedgehog (Shh), leading to decreased levels of mature Shh ligand that is associated with the plasma membrane, and causing transient loss of Hh signaling, resulting in permanent FAS-related morphological abnormalities. In the present study, we further elucidate the mechanisms that regulate the intracellular transportation and secretion of Shh using the hepatic stellate cell line HSC8B. We have found that Shh is associated with caveolin-1 in the Golgi apparatus to form protein complexes and that these complexes are packaged as large punctuate structures (transport vesicles) that are transported to the plasma membrane in lipid raft microdomains. Alcohol exposure does not significantly interrupt translation of shh mRNA in endoplasmic reticulum (ER) or the trafficking of Shh from the ER to the Golgi apparatus. However, alcohol does prevent the entry of Shh into transport vesicles from the Golgi to the plasma membrane and specifically decreases the amount of caveolin-1/Shh complex found in lipid rafts, causing cytoplasmic accumulation of Shh and leading to a deficiency of Shh ligand secretion into the extracellular matrix.
Collapse
Affiliation(s)
- Hua Mao
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
214
|
Sumitran-Holgersson S, Nowak G, Thowfeequ S, Begum S, Joshi M, Jaksch M, Kjaeldgaard A, Jorns C, Ericzon BG, Tosh D. Generation of Hepatocyte-Like Cells from in Vitro Transdifferentiated Human Fetal Pancreas. Cell Transplant 2009; 18:183-93. [DOI: 10.3727/096368909788341333] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Although the appearance of hepatic foci in the pancreas has been described in animal experiments and in human pathology, evidence for the conversion of human pancreatic cells to liver cells is still lacking. We therefore investigated the developmental plasticity between human embryonic pancreatic cells and liver cells. Cells were isolated and expanded from 7–8-week-old human fetal pancreata (HFP) and were characterized for the absence and presence of pancreatic and hepatic markers. In vitro expanded HFP were treated with fibroblast growth factor 2 (FGF2) and dexamethasone (DX) to induce a liver phenotye in the cells. These treated cells in various passages were further studied for their capacity to be functional in hepatic parenchyma following retrorsine-induced injury in nude C57 black mice. Amylase- and EPCAM-positive-enriched cells isolated from HFP and treated with FGF2 and DX lost expression of pancreatic markers and gained a liver phenotype. Hepatic differentiation was based on the expression (both at the mRNA and protein level) of liver markers albumin and cytokeratin 19. When transplanted in vivo into nude mice treated with retrorsine, both cell types successfully engrafted and functionally differentiated into hepatic cells expressing human albumin, glycogen, dipeptidyl peptidase, and γ-glutamyltranspeptidase. These data indicate that human fetal pancreatic cells have a capacity to alter their gene expression profile in response to exogenous treatment with FGF2 and DX. It may be possible to generate an unlimited supply of hepatocytes in vitro for cell therapy.
Collapse
Affiliation(s)
- Suchitra Sumitran-Holgersson
- Division of Transplantation Surgery, Karolinska University Hospital-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Greg Nowak
- Division of Transplantation Surgery, Karolinska University Hospital-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Shifaan Thowfeequ
- Centre for Regenerative Medicine, Department of Biology & Biochemistry, University of Bath, Bath, UK
| | - Setara Begum
- Division of Transplantation Surgery, Karolinska University Hospital-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Meghnad Joshi
- Division of Transplantation Surgery, Karolinska University Hospital-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Marie Jaksch
- The Burnham Institute for Medical Research, La Jolla, CA, USA
| | - Anders Kjaeldgaard
- Division of Obstetrics and Gynecology, Karolinska University Hospital-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Carl Jorns
- Division of Transplantation Surgery, Karolinska University Hospital-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Bo-Göran Ericzon
- Division of Transplantation Surgery, Karolinska University Hospital-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - David Tosh
- Centre for Regenerative Medicine, Department of Biology & Biochemistry, University of Bath, Bath, UK
| |
Collapse
|
215
|
Eichenmüller M, Gruner I, Hagl B, Häberle B, Müller-Höcker J, von Schweinitz D, Kappler R. Blocking the hedgehog pathway inhibits hepatoblastoma growth. Hepatology 2009; 49:482-90. [PMID: 19177589 DOI: 10.1002/hep.22649] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
UNLABELLED Recent evidence has indicated that Hedgehog (Hh) signaling significantly contributes to liver development and regeneration and that activation of the pathway may contribute to growth of hepatocellular carcinoma (HCC) in adults. However, the role of Hh signaling in pediatric liver tumors remains to be elucidated. In this study, we show that Hh signaling is activated in hepatoblastoma (HB), the most common liver tumor in childhood, with most occurrences before the age of 3 years. The Hh target genes glioma-associated oncogene homolog 1 (GLI1) and Patched (PTCH1) showed increased transcript levels in 65% and 30% of HB samples, respectively, compared with normal liver tissues. Most interestingly, the gene encoding the hedgehog interacting protein (HHIP) is transcriptionally silenced by cytosine-phospho-guanosine (CpG) island promoter hypermethylation in 26% of HB cases and treatment with the DNA-demethylating agent 5-aza-2'-deoxycytidine partially restored HHIP expression. Blocking Hh signaling with the antagonist cyclopamine had a strong inhibitory effect on cell proliferation of HB cell lines with an activated pathway. We further demonstrate that this decrease in cell viability is caused by a massive induction of apoptosis, as shown by morphological changes and phosphatidylserine membrane asymmetry. In cyclopamine-exposed HB cells, caspase 3 and poly(adenosine diphosphate-ribose) polymerase proteins were specifically activated by their proteolytic cleavage. CONCLUSION This study demonstrates, for the first time, the frequent occurrence of GLI1 and PTCH1 overexpression and HHIP promoter methylation in early childhood HB, thus indicating a key role for Hh signaling activation in the malignant transformation of embryonal liver cells.
Collapse
Affiliation(s)
- Melanie Eichenmüller
- Department of Pediatric Surgery, Dr von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Federal Republic of Germany
| | | | | | | | | | | | | |
Collapse
|
216
|
Abstract
Tissue-resident stem cells or primitive progenitors play an integral role in homeostasis of most organ systems. Recent developments in methodologies to isolate and culture embryonic and somatic stem cells have many new applications poised for clinical and preclinical trials, which will enable the potential of regenerative medicine to be realized. Here, we overview the current progress in therapeutic applications of various stem cells and discuss technical and social hurdles that must be overcome for their potential to be realized.
Collapse
Affiliation(s)
- Ali M Riazi
- Department of Chemical Engineering, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
217
|
Abstract
Although endodermal organs including the liver, pancreas, and intestine are of significant therapeutic interest, the mechanism by which the endoderm is divided into organ domains during embryogenesis is not well understood. To better understand this process, global gene expression profiling was performed on early endodermal organ domains. This global analysis was followed up by dynamic immunofluorescence analysis of key transcription factors, uncovering novel expression patterns as well as cell surface proteins that allow prospective isolation of specific endodermal organ domains. Additionally, a repressive interaction between Cdx2 and Sox2 was found to occur at the prospective stomach-intestine border, with the hepatic and pancreatic domains forming at this boundary, and Hlxb9 was revealed to have graded expression along the dorsal-ventral axis. These results contribute to understanding the mechanism of endodermal organogenesis and should assist efforts to replicate this process using pluripotent stem cells.
Collapse
Affiliation(s)
- Richard I Sherwood
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | | | | |
Collapse
|
218
|
|
219
|
Semb H. Expandable endodermal progenitors: new tools to explore endoderm and its derivatives. Cell Stem Cell 2008; 3:355-6. [PMID: 18940723 DOI: 10.1016/j.stem.2008.09.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In the two recent issues of Cell Stem Cell, Rossant and colleagues (Seguin et al., 2008) produced expandable endoderm from hESCs by constitutive expression of Sox transcription factors, while Brickman and associates (Morrison et al., 2008) used a reporter gene strategy to isolate replicating anterior definitive endoderm from mESCs.
Collapse
Affiliation(s)
- Henrik Semb
- Stem Cell Center, Lund University, BMC B10, S-221 84 Lund, Sweden.
| |
Collapse
|
220
|
Abstract
Liver and pancreas progenitors develop from endoderm cells in the embryonic foregut. Shortly after their specification, liver and pancreas progenitors rapidly acquire markedly different cellular functions and regenerative capacities. These changes are elicited by inductive signals and genetic regulatory factors that are highly conserved among vertebrates. Interest in the development and regeneration of the organs has been fueled by the intense need for hepatocytes and pancreatic beta cells in the therapeutic treatment of liver failure and type I diabetes. Studies in diverse model organisms have revealed evolutionarily conserved inductive signals and transcription factor networks that elicit the differentiation of liver and pancreatic cells and provide guidance for how to promote hepatocyte and beta cell differentiation from diverse stem and progenitor cell types.
Collapse
Affiliation(s)
- Kenneth S Zaret
- Epigenetics and Progenitor Cells Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA.
| | | |
Collapse
|
221
|
Chung WS, Shin CH, Stainier DYR. Bmp2 signaling regulates the hepatic versus pancreatic fate decision. Dev Cell 2008; 15:738-48. [PMID: 19000838 PMCID: PMC2610857 DOI: 10.1016/j.devcel.2008.08.019] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2008] [Revised: 07/25/2008] [Accepted: 08/28/2008] [Indexed: 11/20/2022]
Abstract
Explant culture data have suggested that the liver and pancreas originate from common progenitors. We used single-cell-lineage tracing in zebrafish to investigate this question in vivo as well as to analyze the hepatic versus pancreatic fate decision. At early somite stages, endodermal cells located at least two cells away from the midline can give rise to both liver and pancreas. In contrast, endodermal cells closer to the midline give rise to pancreas and intestine, but not liver. Loss- and gain-of-function analyses show that Bmp2b, expressed in the lateral plate mesoderm, signals through Alk8 to induce endodermal cells to become liver. When Bmp2b was overexpressed, medially located endodermal cells, fated to become pancreas and intestine, contributed to the liver. These data provide in vivo evidence for the existence of bipotential hepatopancreatic progenitors and indicate that their fate is regulated by the medio-lateral patterning of the endodermal sheet, a process controlled by Bmp2b.
Collapse
Affiliation(s)
| | | | - Didier Y. R. Stainier
- Department of Biochemistry and Biophysics, Programs in Developmental Biology, Genetics and Human Genetics, the Liver Center and the Diabetes Center, University of California, San Francisco, 1550 4th Street, San Francisco, CA 94158, USA
| |
Collapse
|
222
|
Gittes GK. Developmental biology of the pancreas: a comprehensive review. Dev Biol 2008; 326:4-35. [PMID: 19013144 DOI: 10.1016/j.ydbio.2008.10.024] [Citation(s) in RCA: 317] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2008] [Revised: 10/09/2008] [Accepted: 10/13/2008] [Indexed: 02/06/2023]
Abstract
Pancreatic development represents a fascinating process in which two morphologically distinct tissue types must derive from one simple epithelium. These two tissue types, exocrine (including acinar cells, centro-acinar cells, and ducts) and endocrine cells serve disparate functions, and have entirely different morphology. In addition, the endocrine tissue must become disconnected from the epithelial lining during its development. The pancreatic development field has exploded in recent years, and numerous published reviews have dealt specifically with only recent findings, or specifically with certain aspects of pancreatic development. Here I wish to present a more comprehensive review of all aspects of pancreatic development, though still there is not a room for discussion of stem cell differentiation to pancreas, nor for discussion of post-natal regeneration phenomena, two important fields closely related to pancreatic development.
Collapse
Affiliation(s)
- George K Gittes
- Children's Hospital of Pittsburgh and the University of Pittsburgh School of Medicine, Department of Pediatric Surgery, 3705 Fifth Avenue, Pittsburgh, PA 15213, USA
| |
Collapse
|
223
|
Morrison GM, Oikonomopoulou I, Migueles RP, Soneji S, Livigni A, Enver T, Brickman JM. Anterior definitive endoderm from ESCs reveals a role for FGF signaling. Cell Stem Cell 2008; 3:402-15. [PMID: 18940732 DOI: 10.1016/j.stem.2008.07.021] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 06/01/2008] [Accepted: 07/22/2008] [Indexed: 01/12/2023]
Abstract
The use of embryonic stem cell (ESC) differentiation to generate functional hepatic or pancreatic progenitors and as a tool for developmental biology is limited by an inability to isolate in vitro equivalents of regionally specified anterior definitive endoderm (ADE). To address this, we devised a strategy using a fluorescent reporter gene under the transcriptional control of the anterior endoderm marker Hex alongside the definitive mesendoderm marker Cxcr4. Isolation of Hex(+)Cxcr4(+) differentiating ESCs yielded a population expressing ADE markers that both can be expanded and is competent to undergo differentiation toward liver and pancreatic fates. Hex reporter ESCs were also used to define conditions for ADE specification in serum-free adherent culture and revealed an unexpected role for FGF signaling in the generation of ADE. Our findings in defined monolayer differentiation suggest FGF signaling is an important regulator of early anterior mesendoderm differentiation rather than merely a mediator of morphogenetic movement.
Collapse
Affiliation(s)
- Gillian M Morrison
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, King's Buildings, West Mains Road, Edinburgh EH9 3JQ, UK.
| | | | | | | | | | | | | |
Collapse
|
224
|
Abstract
The major forms of diabetes are characterized by pancreatic islet beta-cell dysfunction and decreased beta-cell numbers, raising hope for cell replacement therapy. Although human islet transplantation is a cell-based therapy under clinical investigation for the treatment of type 1 diabetes, the limited availability of human cadaveric islets for transplantation will preclude its widespread therapeutic application. The result has been an intense focus on the development of alternate sources of beta cells, such as through the guided differentiation of stem or precursor cell populations or the transdifferentiation of more plentiful mature cell populations. Realizing the potential for cell-based therapies, however, requires a thorough understanding of pancreas development and beta-cell formation. Pancreas development is coordinated by a complex interplay of signaling pathways and transcription factors that determine early pancreatic specification as well as the later differentiation of exocrine and endocrine lineages. This review describes the current knowledge of these factors as they relate specifically to the emergence of endocrine beta cells from pancreatic endoderm. Current therapeutic efforts to generate insulin-producing beta-like cells from embryonic stem cells have already capitalized on recent advances in our understanding of the embryonic signals and transcription factors that dictate lineage specification and will most certainly be further enhanced by a continuing emphasis on the identification of novel factors and regulatory relationships.
Collapse
Affiliation(s)
- Jennifer M. Oliver-Krasinski
- Institute for Diabetes, Obesity and Metabolism and the Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Doris A. Stoffers
- Institute for Diabetes, Obesity and Metabolism and the Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
225
|
|
226
|
Vorobeychik M, Bloch K, Zemel R, Bachmetov L, Tur-Kaspa R, Vardi P. Immunohistochemical evaluation of hepatic oval cell activation and differentiation toward pancreatic beta-cell phenotype in streptozotocin-induced diabetic mice. J Mol Histol 2008; 39:463-8. [DOI: 10.1007/s10735-008-9185-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Accepted: 07/15/2008] [Indexed: 11/28/2022]
|
227
|
Hervieu V, Lombard-Bohas C, Dumortier J, Boillot O, Scoazec JY. Primary acinar cell carcinoma of the liver. Virchows Arch 2008; 452:337-41. [PMID: 18193278 DOI: 10.1007/s00428-007-0556-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Revised: 11/09/2007] [Accepted: 12/01/2007] [Indexed: 12/19/2022]
Abstract
We report a case of acinar cell carcinoma primary to the liver. The tumor was diagnosed in a 35-year-old woman complaining of abdominal pain and asthenia; serum alpha-fetoprotein (AFP) levels were increased at 6,000 IU/mL; imaging studies showed a hypervascular mass located in the left lobe of the liver. A left lobectomy was performed. The tumor had a heterogeneous appearance. In well-differentiated areas, tumor cells formed acinar structures, had a pyramidal shape and a highly eosinophilic, granular cytoplasm, PAS diastase resistant. In less-differentiated areas, tumor cells were endocrinelike. The immunohistochemical study showed that tumor cells expressed trypsin. Alpha-fetoprotein and alphal-antritrypsin were detected in about 30% of cells; HepPar1 was present in 15% of cells. Chromogranin A and synaptophysin were detected in rare cells. After surgery, serum AFP levels quickly returned to normal; no evidence of recurrence or metastasis was observed during follow-up. The final diagnosis, based on histological, immunohistochemical, and ultrastructural arguments, was extra-pancreatic acinar cell carcinoma, primary to the liver. This unusual lesion is likely to be the result of an abnormal differentiation pathway involving a transformed multipotential progenitor cell.
Collapse
Affiliation(s)
- Valérie Hervieu
- Service Central d'Anatomie et Cytologie Pathologiques, Hospices Civils de Lyon, Hôpital Edouard Herriot, 69437 Lyon Cedex 03, France
| | | | | | | | | |
Collapse
|
228
|
Jin CX, Li WL, Xu F, Geng ZH, He ZY, Su J, Tao XR, Ding XY, Wang X, Hu YP. Conversion of immortal liver progenitor cells into pancreatic endocrine progenitor cells by persistent expression of Pdx-1. J Cell Biochem 2008; 104:224-36. [PMID: 17979180 DOI: 10.1002/jcb.21617] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The conversion of expandable liver progenitor cells into pancreatic beta cells would provide a renewable cell source for diabetes cell therapy. Previously, we reported the establishment of liver epithelial progenitor cells (LEPCs). In this work, LEPCs were modified into EGFP/Pdx-1 LEPCs, cells with stable expression of both Pdx-1 and EGFP. Unlike previous work, with persistent expression of Pdx-1, EGFP/Pdx-1 LEPCs acquired the phenotype of pancreatic endocrine progenitor cells rather than giving rise to insulin-producing cells directly. EGFP/Pdx-1 LEPCs proliferated vigorously and expressed the crucial transcription factors involved in beta cell development, including Ngn3, NeuroD, Nkx2.2, Nkx6.1, Pax4, Pax6, Isl1, MafA and endogenous Pdx-1, but did not secrete insulin. When cultured in high glucose/low serum medium supplemented with cytokines, EGFP/Pdx-1 LEPCs stopped proliferating and gave rise to functional beta cells without any evidence of exocrine or other islet cell lineage differentiation. When transplanted into diabetic SCID mice, EGFP/Pdx-1 LEPCs ameliorated hyperglycemia by secreting insulin in a glucose regulated manner. Considering the limited availability of beta cells, we propose that our experiments will provide a framework for utilizing the immortal liver progenitor cells as a renewable cell source for the generation of functional pancreatic beta cells.
Collapse
Affiliation(s)
- Cai-Xia Jin
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Best M, Carroll M, Hanley NA, Piper Hanley K. Embryonic stem cells to beta-cells by understanding pancreas development. Mol Cell Endocrinol 2008; 288:86-94. [PMID: 18487011 DOI: 10.1016/j.mce.2008.03.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2007] [Revised: 03/18/2008] [Accepted: 03/18/2008] [Indexed: 12/25/2022]
Abstract
Insulin injections treat but do not cure Type 1 diabetes (T1DM). The success of islet transplantation suggests cell replacement therapies may offer a curative strategy. However, cadaver islets are of insufficient number for this to become a widespread treatment. To address this deficiency, the production of beta-cells from pluripotent stem cells offers an ambitious far-sighted opportunity. Recent progress in generating insulin-producing cells from embryonic stem cells has shown promise, highlighting the potential of trying to mimic normal developmental pathways. Here, we provide an overview of the current methodology that has been used to differentiate stem cells toward a beta-cell fate. Parallels are drawn with what is known about normal development, especially regarding the human pancreas.
Collapse
Affiliation(s)
- Marie Best
- Centre for Human Development, Stem Cells & Regeneration, UK
| | | | | | | |
Collapse
|
230
|
Piscaglia AC, Novi M, Campanale M, Gasbarrini A. Stem cell-based therapy in gastroenterology and hepatology. MINIM INVASIV THER 2008; 17:100-18. [PMID: 18465445 DOI: 10.1080/13645700801969980] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Protagonists of a new scientific era, stem cells are promising tools on which regenerative medicine relies for the treatment of human pathologies. Stem cells can be obtained from various sources, including embryos, fetal tissues, umbilical cord blood, and also terminally differentiated organs. Once forced to expand and differentiate into functional progenies, stem cells may become suitable for cell replacement and tissue engineering. The manipulation and/or stimulation of adult stem cells seems to be particularly promising, as it could improve the endogenous regenerative potential without risks of rejection and overcome the ethical and political issues related to embryonic stem cell research. Stem cells are already leaving the bench and reaching the bedside, despite an incomplete knowledge of the genetic control program driving their fate and plasticity. In gastroenterology and hepatology, the first attempts to translate stem cell basic research into novel therapeutic strategies have been made for the treatment of several disorders, such as inflammatory bowel diseases, diabetes mellitus, celiachy and acute or chronic hepatopaties. Nonetheless, critical aspects need to be further addressed, including the long-term safety, tolerability and efficacy of cell-based treatments, as well as their carcinogenic potential. Aim of this review is to summarize the state-of-the-arts on gastrointestinal and hepatic stem cells and on stem cell-based therapies in gastroenterology and hepatology, highlighting both the benefits and the potential risks of these new tools for the treatment and prevention of human diseases.
Collapse
Affiliation(s)
- Anna Chiara Piscaglia
- Gastrointestinal and Hepatic Stem Cell Research Group (G.H.S.C.) , Department of Internal Medicine and Gastroenterology, Gemelli Hospital, Catholic University of Rome, Rome, Italy
| | | | | | | |
Collapse
|
231
|
Tada M, Kanai F, Tanaka Y, Tateishi K, Ohta M, Asaoka Y, Seto M, Muroyama R, Fukai K, Imazeki F, Kawabe T, Yokosuka O, Omata M. Down-regulation of hedgehog-interacting protein through genetic and epigenetic alterations in human hepatocellular carcinoma. Clin Cancer Res 2008; 14:3768-3776. [PMID: 18559595 DOI: 10.1158/1078-0432.ccr-07-1181] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE Hedgehog (Hh) signaling is activated in several cancers. However, the mechanisms of Hh signaling activation in hepatocellular carcinoma (HCC) have not been fully elucidated. We analyzed the involvement of Hh-interacting protein (HHIP) gene, a negative regulator of Hh signaling, in HCC. EXPERIMENTAL DESIGN Glioma-associated oncogene homologue (Gli) reporter assay, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay, and quantitative real-time reverse transcription-PCR for the target genes of the Hh signals were performed in HHIP stably expressing hepatoma cells. Quantitative real-time PCR for HHIP was performed in hepatoma cells and 36 HCC tissues. The methylation status of hepatoma cells and HCC tissues was also analyzed by sodium bisulfite sequencing, demethylation assay, and quantitative real-time methylation-specific PCR. Loss of heterozygosity (LOH) analysis was also performed in HCC tissues. RESULTS HHIP overexpression induced significant reductions of Gli reporter activity, cell viability, and transcription of the target genes of the Hh signals. HHIP was hypermethylated and transcriptionally down-regulated in a subset of hepatoma cells. Treatment with a demethylating agent led to the HHIP DNA demethylation and restoration of HHIP transcription. HHIP transcription was also down-regulated in the majority of HCC tissues, and more than half of HCC tissues exhibited HHIP hypermethylation. The HHIP transcription level in HHIP-methylated HCC tissues was significantly lower than in HHIP-unmethylated HCC tissues. More than 30% of HCC tissues showed LOH at the HHIP locus. CONCLUSIONS The down-regulation of HHIP transcription is due to DNA hypermethylation and/or LOH, and Hh signal activation through the inactivation of HHIP may be implicated in the pathogenesis of human HCC.
Collapse
Affiliation(s)
- Motohisa Tada
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Kawasaki H, Mizuguchi T, Kikkawa Y, Oshima H, Sasaki Y, Tokino T, Kokai Y, Miyazaki JI, Katsuramaki T, Mitaka T, Hirata K. In vitro transformation of adult rat hepatic progenitor cells into pancreatic endocrine hormone-producing cells. JOURNAL OF HEPATO-BILIARY-PANCREATIC SURGERY 2008; 15:310-317. [PMID: 18535770 DOI: 10.1007/s00534-007-1252-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Accepted: 07/06/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND/PURPOSE Pancreatic and duodenal homeobox factor 1 (Pdx-1) plays an important role in initiating differentiation toward pancreatic endocrine cells. The transdifferentiation or transformation of hepatocytes into pancreatic endocrine cells could be feasible, due to their similar cellular origins. Our goal in this study was to see if small hepatocytes (SHs) could give rise to pancreatic endocrine cells via exogenous Pdx-1 gene expression. METHODS SHs were cultured for 10 days before adenovirus (Adt)-mediated Pdx-1 gene transfection. We performed western blot analysis for pancreatic transcription factors in the nuclei and reverse-transcription polymerase chain reaction (RT-PCR) for the gene expression of pancreatic endocrine hormones. Confocal laser microscanning analysis was used to observe the transformation of SHs. RESULTS Pancreatic transcription factors such as Pdx-1, Ngn3, NeuroD, Nkx2.2, and Pax6 were induced after Adt-Pdx-1 gene transfection. The mRNA expression of pancreatic endocrine hormones (insulin, glucagon, and somatostatin) was induced after the gene transfection. Pdx-1 was expressed in the nucleus, where the cells were positive for one or more of the hormones and cytokeratin (CK) 8. Some cells were positive for multiple hormones. The insulin level increased while the glucagon level decreased after the glucose loading test, depending on the glucose concentration. CONCLUSIONS SHs are transformed into functional pancreatic endocrine cells after Pdx-1 gene transfection.
Collapse
Affiliation(s)
- Hiroyuki Kawasaki
- First Department of Surgery, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido 060-8543, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Shiraki N, Umeda K, Sakashita N, Takeya M, Kume K, Kume S. Differentiation of mouse and human embryonic stem cells into hepatic lineages. Genes Cells 2008; 13:731-46. [PMID: 18513331 DOI: 10.1111/j.1365-2443.2008.01201.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We recently reported a novel method to induce embryonic stem (ES) cells differentiate into an endodermal fate, especially pancreatic, using a supporting cell line. Here we describe the modified culture condition with the addition and withdrawal of secreted growth factors could induce ES cells to selectively differentiate into a hepatic fate efficiently. The signaling of BMP and FGF that have been implicated in hepatic differentiation during normal embryonic development are shown to play pivotal roles in generating hepatic cells from the definitive endoderm derived from ES cells. Moreover, the expression of AFP, Albumin or a biliary molecular marker appeared sequentially thus suggested the differentiation of ES cells recapitulated normal developmental processes of liver. The ES cell-derived differentiated cells showed evidence of glycogen storage, secreted Albumin, exhibited drug metabolism activities and expressed a set of cytochrome or drug conjugate enzymes, drug transporters specifically expressed in mature hepatocytes. With the same procedure, human ES cells also gave rise to cells with mature hepatocytes' characteristics. In conclusion, this novel procedure for hepatic differentiation will be useful for elucidation of molecular mechanisms of hepatic fate decision at gut regionalization, and could represent an attractive approach for a surrogate cell source for pharmaceutical studies such as toxicology.
Collapse
Affiliation(s)
- Nobuaki Shiraki
- Divisions of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Kumamoto 860-0811, Japan
| | | | | | | | | | | |
Collapse
|
234
|
Goessling W, North TE, Lord AM, Ceol C, Lee S, Weidinger G, Bourque C, Strijbosch R, Haramis AP, Puder M, Clevers H, Moon RT, Zon LI. APC mutant zebrafish uncover a changing temporal requirement for wnt signaling in liver development. Dev Biol 2008; 320:161-74. [PMID: 18585699 DOI: 10.1016/j.ydbio.2008.05.526] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2008] [Revised: 04/21/2008] [Accepted: 05/02/2008] [Indexed: 12/13/2022]
Abstract
Developmental signaling pathways hold the keys to unlocking the promise of adult tissue regeneration, and to inhibiting carcinogenesis. Patients with mutations in the Adenomatous Polyposis Coli (APC) gene are at increased risk of developing hepatoblastoma, an embryonal form of liver cancer, suggesting that Wnt affects hepatic progenitor cells. To elucidate the role of APC loss and enhanced Wnt activity in liver development, we examined APC mutant and wnt inducible transgenic zebrafish. APC(+/-) embryos developed enlarged livers through biased induction of hepatic gene programs and increased proliferation. Conversely, APC(-/-) embryos formed no livers. Blastula transplantations determined that the effects of APC loss were cell autonomous. Induction of wnt modulators confirmed biphasic consequences of wnt activation: endodermal pattern formation and gene expression required suppression of wnt signaling in early somitogenesis; later, increased wnt activity altered endodermal fate by enhancing liver growth at the expense of pancreas formation; these effects persisted into the larval stage. In adult APC(+/-) zebrafish, increased wnt activity significantly accelerated liver regeneration after partial hepatectomy. Similarly, liver regeneration was significantly enhanced in APC(Min/+) mice, indicating the conserved effect of Wnt pathway activation in liver regeneration across vertebrate species. These studies reveal an important and time-dependent role for wnt signaling during liver development and regeneration.
Collapse
Affiliation(s)
- Wolfram Goessling
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, HHMI, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Hanley KP, Oakley F, Sugden S, Wilson DI, Mann DA, Hanley NA. Ectopic SOX9 mediates extracellular matrix deposition characteristic of organ fibrosis. J Biol Chem 2008; 283:14063-71. [PMID: 18296708 DOI: 10.1074/jbc.m707390200] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Appropriate temporospatial expression of the transcription factor SOX9 is important for normal development of a wide range of organs. Here, we show that when SOX9 is expressed ectopically, target genes become expressed that are associated with disease. Histone deacetylase inhibitors in clinical trials for cancer therapy induced SOX9 expression via enhanced recruitment of nuclear factor Y (NF-Y) to CCAAT elements in the SOX9 proximal promoter. The effect of histone deacetylase inhibitors could be elicited in cells that normally lack SOX9, such as hepatocytes. In human fetal hepatocytes, this aberrant induction of SOX9 protein caused ectopic expression of COL2A1 and COMP1 that encode extracellular matrix (ECM) components normally associated with chondrogenesis. Previously, ectopic expression of this "chondrogenic" profile has been implicated in vascular calcification. More broadly, inappropriate ECM deposition is a hallmark of fibrosis. We demonstrated that induction of SOX9 expression also occurred during activation of fibrogenic cells from the adult liver when the transcription factor was responsible for expression of the major component of fibrotic ECM, type 1 collagen. These combined data identify new aspects in the regulation of SOX9 expression. They support a role for SOX9 beyond normal development as a transcriptional regulator in the pathology of fibrosis.
Collapse
|
236
|
Abstract
In both Type 1 and 2 diabetes, insufficient numbers of insulin-producing beta-cells are a major cause of defective control of blood glucose and its complications. Restoration of damaged beta-cells by endocrine pancreas regeneration would be an ideal therapeutic option. The possibility of generating insulin-secreting cells with adult pancreatic stem or progenitor cells has been investigated extensively. The conversion of differentiated cells such as hepatocytes into beta-cells is being attempted using molecular insights into the transcriptional make-up of beta-cells. Additionally, the enhanced proliferation of beta-cells in vivo or in vitro is being pursued as a strategy for regenerative medicine for diabetes. Advances have also been made in directing the differentiation of embryonic stem cells into beta-cells. Although progress is encouraging, major gaps in our understanding of developmental biology of the pancreas and adult beta-cell dynamics remain to be bridged before a therapeutic application is made possible.
Collapse
Affiliation(s)
- Valeria Sordi
- Laboratory of Experimental Surgery, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | | | | |
Collapse
|
237
|
Minamiguchi H, Ishikawa F, Fleming PA, Yang S, Drake CJ, Wingard JR, Ogawa M. Transplanted human cord blood cells generate amylase-producing pancreatic acinar cells in engrafted mice. Pancreas 2008; 36:e30-5. [PMID: 18376297 DOI: 10.1097/mpa.0b013e3181584656] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Pancreatic acinar cells and hepatocytes arise from the same cell population located within the embryonic endoderm. It has been reported that a multipotent population of liver cells is capable of differentiating into pancreatic cells. Recent studies revealed that murine and human hematopoietic cells could generate hepatocytes in vivo. Based on this developmental proximity between liver and pancreatic acinar cells, we examined whether human cord blood (CB) cells can generate pancreatic cells in vivo using a murine xenograft model. METHODS We transplanted 1 x 10 CD34 human CB cells into "conditioned" newborn nonobese diabetic-severe combined immunodeficiency/beta-2 microglobulin-null mice via facial vein injection and, 3 to 4 months later, examined the pancreata from recipient mice showing high-level human multilineage hematopoietic engraftment in the bone marrow. RESULTS Reverse transcriptase-polymerase chain reaction and immunohistochemical analyses revealed human amylase mRNA and protein expression, respectively, in the pancreata from recipient mice. Using fluorescence in situ hybridization, we identified human alpha-satellite, DNA-positive cells with a morphology characteristic of pancreatic acinar cells. We also identified cells in paraffin sections of the pancreata that expressed amylase mRNA, had morphological characteristics of acinar cells, and contained human but not mouse centromeric DNA. CONCLUSION These findings establish that human umbilical CB cells are capable of generating pancreatic acinar cells via a nonfusion mechanism.
Collapse
Affiliation(s)
- Hitoshi Minamiguchi
- Department of Veterans Affairs Medical Center and Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | | | | | | | | | | | | |
Collapse
|
238
|
Omenetti A, Diehl AM. The adventures of sonic hedgehog in development and repair. II. Sonic hedgehog and liver development, inflammation, and cancer. Am J Physiol Gastrointest Liver Physiol 2008; 294:G595-8. [PMID: 18218671 DOI: 10.1152/ajpgi.00543.2007] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hedgehog (Hh) signaling modulates tissue remodeling by controlling the fate of Hh-responsive cells. Healthy adult livers exhibit little Hh activity. However, cells involved in adult liver repair, including myofibroblasts and progenitors, are capable of producing and responding to Hh ligands. During adult liver injury, Hh ligand production increases and populations of Hh-responsive cells expand. This process is accompanied by fibrosis. Ligand production and Hh-responsive cells diminish as fibrosis resolves and normal hepatic architecture is restored, but Hh signaling persists in hepatocellular carcinomas. These findings suggest that the Hh pathway mediates remodeling responses that are triggered by adult liver damage.
Collapse
Affiliation(s)
- Alessia Omenetti
- Duke University Medical Center, Division of Gastroenterology, Department of Medicine, Durham, NC 27710, USA
| | | |
Collapse
|
239
|
Histone deacetylase 3 (hdac3) is specifically required for liver development in zebrafish. Dev Biol 2008; 317:336-53. [PMID: 18367159 DOI: 10.1016/j.ydbio.2008.02.034] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 02/14/2008] [Accepted: 02/15/2008] [Indexed: 12/22/2022]
Abstract
Histone deacetylases (HDACs) are key transcription regulators that function by deacetylating histones/transcription factors and modifying chromatin structure. In this work, we showed that chemical inhibition of HDACs by valproic acid (VPA) led to impaired liver development in zebrafish mainly by inhibiting specification, budding, and differentiation. Formation of exocrine pancreas but not endocrine pancreas was also inhibited. The liver defects induced by VPA correlate with suppressed total HDAC enzymatic activity, but are independent of angiogenesis inhibition. Gene knockdown by morpholino demonstrated that hdac3 is specifically required for liver formation while hdac1 is more globally required for multiple development processes in zebrafish including liver/exocrine pancreas formation. Furthermore, overexpression of hdac3 but not hdac1 partially rescued VPA induced small liver. One mechanism by which hdac3 regulates zebrafish liver growth is through inhibiting growth differentiation factor 11 (gdf11), a unique target of hdac3 and a member of the transforming growth factor beta family. Simultaneous overexpression or morpholino knockdown showed that hdac3 and gdf11 function antagonistically in zebrafish liver development. These results revealed a novel and specific role of hdac3 in liver development and the distinct functions between hdac1 and hdac3 in zebrafish embryonic development.
Collapse
|
240
|
Racanicchi L, Basta G, Montanucci P, Guido L, Pensato A, Conti V, Calafiore R. Neonatal pig liver-derived progenitors for insulin-producing cells: an in vitro study. ACTA ACUST UNITED AC 2008; 13:2923-31. [PMID: 17916039 DOI: 10.1089/ten.2007.0098] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Beta (beta)-cell replacement represents an attractive approach for the possible cure of type 1 insulin-dependent diabetes mellitus (IDDM). In a search for potential sources of insulin-secreting cells for IDDM substitution therapy, we have focused on the neonatal pig liver, which is putatively enriched in multipotent stem cells. We then isolated cells measuring 10 to 15 microm in diameter, identified as small cells, characterized by a high proliferation rate and positive staining for immature liver and pancreatic endocrine cell markers (i.e., insulin and pancreatic duodenal homeobox). The ability of these cells to transdifferentiate into pancreatic beta-like cells under culture conditions with exendin-4 (Ex-4) or high glucose concentration was examined. We observed that insulin secretion was not physiological in basal conditions, although it became responsive to glucose after 5 days of exposure to Ex-4. This beta-cell-like phenotype remained physiologically stable, even after stimulus withdrawal. Based on these observations, we contend that the proposed cell and tissue model might offer several advantages as a candidate for substitution cell therapy in IDDM, because the neonatal pig liver seems enriched in cells, with a mixed pancreas-liver phenotype, that are easier to purify and grow in culture and are more functional than other beta-like cells upon in vitro single short-term stimulation challenge.
Collapse
Affiliation(s)
- Leda Racanicchi
- Department of Internal Medicine, Section of Internal Medicine and Endocrine and Metabolic Sciences, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | |
Collapse
|
241
|
Miki R, Tatsumi N, Matsumoto K, Yokouchi Y. New primary culture systems to study the differentiation and proliferation of mouse fetal hepatoblasts. Am J Physiol Gastrointest Liver Physiol 2008; 294:G529-39. [PMID: 18096607 DOI: 10.1152/ajpgi.00412.2007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatoblasts have the potential to differentiate into both hepatocytes and biliary epithelial cells through a differentiation program that has not been fully elucidated. With the aim to better define the mechanism of differentiation of hepatoblasts, we isolated hepatoblasts and established new culture systems. We isolated hepatoblasts from E12.5 fetal mouse liver by using E-cadherin. The E-cadherin+ cells expressed alpha-fetoprotein (AFP) and albumin (Alb) but not cytokeratin 19 (CK19). Transplantation of the E-cadherin+ cells into mice that had been subjected to liver injury or biliary epithelial injury led to differentiation of the cells into hepatocytes or biliary epithelial cells, respectively. In a low-cell-density culture system in the absence of additional growth factors, E-cadherin+ cells formed colonies of various sizes, largely comprising Alb-positive cells. Supplementation of the culture medium with hepatocyte growth factor and epidermal growth factor promoted proliferation of the cells. Thus the low-cell-density culture system should be useful to identify inductive factors that regulate the proliferation and differentiation of hepatoblasts. In a high-cell-density system in the presence of oncostatin M+dexamethasone, E14.5, but not E12.5, E-cadherin+ cells differentiated into mature hepatocytes, suggesting that unidentified factors are involved in hepatic maturation. Culture of E-cadherin+ cells derived from E12.5 or E14.5 liver under high-cell-density conditions should allow elucidation of the mechanism of hepatic differentiation in greater detail. These new culture systems should be of use to identify growth factors that induce hepatoblasts to proliferate or differentiate into hepatocytes and biliary epithelial cells.
Collapse
Affiliation(s)
- Rika Miki
- Division of Pattern Formation, Department of Organogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto, 860-0811, Japan
| | | | | | | |
Collapse
|
242
|
Kumagi T, Heathcote EJ. Primary biliary cirrhosis. Orphanet J Rare Dis 2008; 3:1. [PMID: 18215315 PMCID: PMC2266722 DOI: 10.1186/1750-1172-3-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Accepted: 01/23/2008] [Indexed: 12/15/2022] Open
Abstract
Primary biliary cirrhosis (PBC) is a chronic and slowly progressive cholestatic liver disease of autoimmune etiology characterized by injury of the intrahepatic bile ducts that may eventually lead to liver failure. Affected individuals are usually in their fifth to seventh decades of life at time of diagnosis, and 90% are women. Annual incidence is estimated between 0.7 and 49 cases per million-population and prevalence between 6.7 and 940 cases per million-population (depending on age and sex). The majority of patients are asymptomatic at diagnosis, however, some patients present with symptoms of fatigue and/or pruritus. Patients may even present with ascites, hepatic encephalopathy and/or esophageal variceal hemorrhage. PBC is associated with other autoimmune diseases such as Sjogren's syndrome, scleroderma, Raynaud's phenomenon and CREST syndrome and is regarded as an organ specific autoimmune disease. Genetic susceptibility as a predisposing factor for PBC has been suggested. Environmental factors may have potential causative role (infection, chemicals, smoking). Diagnosis is based on a combination of clinical features, abnormal liver biochemical pattern in a cholestatic picture persisting for more than six months and presence of detectable antimitochondrial antibodies (AMA) in serum. All AMA negative patients with cholestatic liver disease should be carefully evaluated with cholangiography and liver biopsy. Ursodeoxycholic acid (UDCA) is the only currently known medication that can slow the disease progression. Patients, particularly those who start UDCA treatment at early-stage disease and who respond in terms of improvement of the liver biochemistry, have a good prognosis. Liver transplantation is usually an option for patients with liver failure and the outcome is 70% survival at 7 years. Recently, animal models have been discovered that may provide a new insight into the pathogenesis of this disease and facilitate appreciation for novel treatment in PBC.
Collapse
Affiliation(s)
- Teru Kumagi
- Department of Medicine, Toronto Western Hospital (University Health Network/University of Toronto), Toronto, Ontario, Canada.
| | | |
Collapse
|
243
|
Transition to pancreatic cancer in response to carcinogen. Langenbecks Arch Surg 2008; 393:557-60. [DOI: 10.1007/s00423-007-0274-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Accepted: 11/12/2007] [Indexed: 01/04/2023]
|
244
|
Funa NS, Saldeen J, Akerblom B, Welsh M. Interdependent fibroblast growth factor and activin A signaling promotes the expression of endodermal genes in differentiating mouse embryonic stem cells expressing Src Homology 2-domain inactive Shb. Differentiation 2007; 76:443-53. [PMID: 18093225 DOI: 10.1111/j.1432-0436.2007.00249.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The mechanisms controlling endodermal development during stem cell differentiation have been only partly elucidated, although previous studies have suggested the participation of fibroblast growth factor (FGF) and activin A in these processes. Shb is a Src homology 2 (SH2) domain-containing adapter protein that has been implicated in FGF receptor 1 (FGFR1) signaling. To study the putative crosstalk between activin A and Shb-dependent FGF signaling in the differentiation of endoderm from embryonic stem (ES) cells, embryoid bodies (EBs) derived from mouse ES cells overexpressing wild-type Shb or Shb with a mutated SH2 domain (R522K-Shb) were cultured in the presence of activin A. We show that expression of R522K-Shb results in up-regulation of FGFR1 and FGF2 in EBs. Addition of activin A to the cultures enhances the expression of endodermal genes primarily in EBs expressing mutant Shb. Inhibition of FGF signaling by the addition of the FGFR1 inhibitor SU5402 completely counteracts the synergistic effects of R522K-Shb and activin A. In conclusion, the present results suggest that expression of R522K-Shb enhances certain signaling pathways downstream of FGF and that an interplay between FGF and activin A participates in ES cell differentiation to endoderm.
Collapse
Affiliation(s)
- Nina S Funa
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, PO Box 571, Husargatan 3, SE-751 23, Uppsala, Sweden
| | | | | | | |
Collapse
|
245
|
Xu J, Lu Y, Ding F, Zhan X, Zhu M, Wang Z. Reversal of diabetes in mice by intrahepatic injection of bone-derived GFP-murine mesenchymal stem cells infected with the recombinant retrovirus-carrying human insulin gene. World J Surg 2007; 31:1872-1882. [PMID: 17653584 DOI: 10.1007/s00268-007-9168-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The objective of this study was to assess the effect of intrahepatic injection of bone-derived green fluorescent protein (GFP)-transgenic murine mesenchymal stem cells (GFP-mMSCs) containing the human insulin(ins) gene in streptozotocin-induced diabetic mice. METHODS GFP-mMSCs were isolated from the bone marrow of GFP transgenic mice, expanded, and transfected with a recombinant retrovirus MSCV carrying the human insulin gene. C57BL/6J mice were made diabetic by an intraperitoneal administration of 160 mg/kg streptozotocin (STZ), followed by intrahepatic injection of transfected GFP-mMSCs. The variations in body weight and the blood glucose and serum insulin levels were determined after cell transplantation. GFP-mMSCs survival and human insulin expression in liver tissues were examined by fluorescent microscopy and immunohistochemistry. RESULTS The body weight in diabetic mice that received GFP-mMSCs harboring the human insulin gene was increased by 6% within 6 weeks after treatment, and the average blood glucose levels in these animals were 10.40 +/- 2.80 mmol/l (day 7) and 6.50 +/- 0.89 mmol/l (day 42), respectively, while the average values of blood glucose in diabetic animals without treatment were 26.80 +/- 2.49 mmol/l (day 7) and 25.40 +/- 4.10 mmol/l (day 42), showing a significant difference (p < 0.05). Moreover, secretion of human insulin of GFP-mMSCs in serum and animal liver was detected by radioimmunoassay (RIA) and immunohistochemistry (IHC). CONCLUSIONS Experimental diabetes could be relieved effectively for up to 6 weeks by intrahepatic transplantation of murine mesenchymal stem cells expressing human insulin. This study implies a novel approach of gene therapy for type I diabetes.
Collapse
Affiliation(s)
- Jian Xu
- Department of General Surgery, the Affiliated Hospital, Nantong University, Nantong, Jiangsu Province, China, 226001
| | - Yuhua Lu
- Department of General Surgery, the Affiliated Hospital, Nantong University, Nantong, Jiangsu Province, China, 226001
| | - Fei Ding
- Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China, 226001
| | - Xi Zhan
- Greenebaum Cancer Center, School of Medicine, University of Maryland, Baltimore, USA
| | - Mingyan Zhu
- Department of General Surgery, the Affiliated Hospital, Nantong University, Nantong, Jiangsu Province, China, 226001
| | - Zhiwei Wang
- Department of General Surgery, the Affiliated Hospital, Nantong University, Nantong, Jiangsu Province, China, 226001.
| |
Collapse
|
246
|
Thowfeequ S, Myatt EJ, Tosh D. Transdifferentiation in developmental biology, disease, and in therapy. Dev Dyn 2007; 236:3208-17. [PMID: 17948254 DOI: 10.1002/dvdy.21336] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Transdifferentiation (or metaplasia) refers to the conversion of one cell type to another. Because transdifferentiation normally occurs between cells that arise from the same region of the embryo, understanding the molecular and cellular events in cell type transformations may help to explain the mechanisms underlying normal development. Here we review examples of transdifferentiation in nature focusing on the possible role of cell type switching in metamorphosis and regeneration. We also examine transdifferentiation in mammals in relation to disease and the use of transdifferentiated cells in cellular therapy.
Collapse
Affiliation(s)
- Shifaan Thowfeequ
- Centre for Regenerative Medicine, Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, United Kingdom
| | | | | |
Collapse
|
247
|
Abstract
The circuitous cell signalling pathways of hepatocytes comprise several factors that operate to downgrade or even interrupt the transmission of a given signal. These down-regulating influences are essential to keep cell proliferation and cell survival in check and if impaired, can alter a delicate balance in favour of cell proliferation. Each signalling pathway that has been implicated in carcinogenesis is influenced by both oncogenic factors that promote tumour growth when activated as well as tumour suppressor proteins that have to be impaired to favour tumour growth. This summary of the Tumour Suppressors in Liver Carcinogenesis Symposium held at the 2007 EASL Annual Meeting discusses four pathways with pre-eminent tumour suppressor activity, each involved in hepatocarcinogenesis: p53, mTOR, beta-catenin and hedgehog.
Collapse
|
248
|
van den Brink GR. Hedgehog signaling in development and homeostasis of the gastrointestinal tract. Physiol Rev 2007; 87:1343-75. [PMID: 17928586 DOI: 10.1152/physrev.00054.2006] [Citation(s) in RCA: 210] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The Hedgehog family of secreted morphogenetic proteins acts through a complex evolutionary conserved signaling pathway to regulate patterning events during development and in the adult organism. In this review I discuss the role of Hedgehog signaling in the development, postnatal maintenance, and carcinogenesis of the gastrointestinal tract. Three mammalian hedgehog genes, sonic hedgehog (Shh), indian hedgehog (Ihh), and desert hedgehog (Dhh) have been identified. Shh and Ihh are important endodermal signals in the endodermal-mesodermal cross-talk that patterns the developing gut tube along different axes. Mutations in Shh, Ihh, and downstream signaling molecules lead to a variety of gross malformations of the murine gastrointestinal tract including esophageal atresia, tracheoesophageal fistula, annular pancreas, midgut malrotation, and duodenal and anal atresia. These congenital malformations are also found in varying constellations in humans, suggesting a possible role for defective Hedgehog signaling in these patients. In the adult, Hedgehog signaling regulates homeostasis in several endoderm-derived epithelia, for example, the stomach, intestine, and pancreas. Finally, growth of carcinomas of the proximal gastrointestinal tract such as esophageal, gastric, biliary duct, and pancreatic cancers may depend on Hedgehog signaling offering a potential avenue for novel therapy for these aggressive cancers.
Collapse
Affiliation(s)
- Gijs R van den Brink
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
249
|
Liang X, Ma L, Thai NL, Fung JJ, Qian S, Lu L. The role of liver-derived regulatory dendritic cells in prevention of type 1 diabetes. Immunology 2007; 120:251-60. [PMID: 17233742 PMCID: PMC2265858 DOI: 10.1111/j.1365-2567.2006.02496.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Development of type 1 diabetes has been attributed to T-cell-mediated autoimmunity, which is regulated by antigen-presenting cells. To study the role of liver-derived B220(+) regulatory dendritic cells (DCs) in the development of diabetes in non-obese diabetic (NOD) mice, we found that liver 220(+) DCs could easily be propagated from young NOD mice, but that such propagation was extremely difficult from mice older than 11 weeks, when insulitis began. This was not simply an age-related phenomenon, because liver B220(+) DCs were readily propagated from both young and old congenic non-obese diabetic-resistant (NOR) and normal BALB/c mice. It was therefore speculated that the development of diabetes might be associated with a lack of precursors of B220(+) DC in the liver in this animal model. Unfortunately, the specific marker for precursors of liver B220(+) DC has not been identified. An alternative approach to supplement liver B220(+) DCs by intravenous administration significantly inhibited the development of diabetes by inducing T-cell hyporesponsiveness via enhancement of their apoptotic death. Liver B220(+) DCs were capable of effectively presenting antigens but, unlike plasmacytoid DCs, did not express CD11c and were not interferon-alpha producers. These observations may throw new light on the aetiopathology of type 1 diabetes.
Collapse
Affiliation(s)
- Xiaoyan Liang
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of PittsburghPittsburgh, PA
| | - Linlin Ma
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of PittsburghPittsburgh, PA
| | - Ngoc L Thai
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of PittsburghPittsburgh, PA
| | - John J Fung
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of PittsburghPittsburgh, PA
- Department of General Surgery, Cleveland ClinicCleveland, OH, USA
| | - Shiguang Qian
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of PittsburghPittsburgh, PA
- Department of Immunology, Cleveland ClinicCleveland, OH, USA
| | - Lina Lu
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of PittsburghPittsburgh, PA
- Department of Immunology, Cleveland ClinicCleveland, OH, USA
| |
Collapse
|
250
|
Remacle C, Dumortier O, Bol V, Goosse K, Romanus P, Theys N, Bouckenooghe T, Reusens B. Intrauterine programming of the endocrine pancreas. Diabetes Obes Metab 2007; 9 Suppl 2:196-209. [PMID: 17919194 DOI: 10.1111/j.1463-1326.2007.00790.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Epidemiological studies have revealed strong relationships between poor foetal growth and subsequent development of the metabolic syndrome. Persisting effects of early malnutrition become translated into pathology, thereby determine chronic risk for developing glucose intolerance and diabetes. These epidemiological observations identify the phenomena of foetal programming without explaining the underlying mechanisms that establish the causal link. Animal models have been established and studies have demonstrated that reduction in the availability of nutrients during foetal development programs the endocrine pancreas and insulin-sensitive tissues. Whatever the type of foetal malnutrition, whether there are not enough calories or protein in food or after placental deficiency, malnourished pups are born with a defect in their beta-cell population that will never completely recover, and insulin-sensitive tissues will be definitively altered. Despite the similar endpoint, different cellular and physiological mechanisms are proposed. Hormones operative during foetal life like insulin itself, insulin-like growth factors and glucocorticoids, as well as specific molecules like taurine, or islet vascularization were implicated as possible factors amplifying the defect. The molecular mechanisms responsible for intrauterine programming of the beta cells are still elusive, but two hypotheses recently emerged: the first one implies programming of mitochondria and the second, epigenetic regulation.
Collapse
Affiliation(s)
- C Remacle
- Laboratory of Cell Biology, Institute of Life Sciences, Catholic University of Louvain, Louvain-la-Neuve, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|