151
|
Cano DA, Hebrok M, Zenker M. Pancreatic development and disease. Gastroenterology 2007; 132:745-62. [PMID: 17258745 DOI: 10.1053/j.gastro.2006.12.054] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Accepted: 12/20/2006] [Indexed: 12/22/2022]
Affiliation(s)
- David A Cano
- Diabetes Center, Department of Medicine, University of California San Francisco, 94143, USA
| | | | | |
Collapse
|
152
|
Abstract
Several key signalling pathways, such as Hedgehog, Notch, Wnt and BMP-TGFbeta-Activin (bone morphogenetic protein-transforming growth factor-beta-Activin), are involved in most processes essential to the proper development of an embryo. It is also becoming increasingly clear that these pathways can have a crucial role in tumorigenesis when reactivated in adult tissues through sporadic mutations or other mechanisms. We will focus here on the Hedgehog pathway, which is abnormally activated in most basal cell carcinomas, and discuss potential therapeutic opportunities offered by the progress made in understanding this signalling pathway.
Collapse
Affiliation(s)
- Lee L Rubin
- Harvard Stem Cell Institute, Biolabs, Room 150, 16 Divinity Avenue, Cambridge, Massachusetts 02138, USA.
| | | |
Collapse
|
153
|
Wells JM, Esni F, Boivin GP, Aronow BJ, Stuart W, Combs C, Sklenka A, Leach SD, Lowy AM. Wnt/beta-catenin signaling is required for development of the exocrine pancreas. BMC DEVELOPMENTAL BIOLOGY 2007; 7:4. [PMID: 17222338 PMCID: PMC1783845 DOI: 10.1186/1471-213x-7-4] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2006] [Accepted: 01/12/2007] [Indexed: 12/23/2022]
Abstract
Background β-catenin is an essential mediator of canonical Wnt signaling and a central component of the cadherin-catenin epithelial adhesion complex. Dysregulation of β-catenin expression has been described in pancreatic neoplasia. Newly published studies have suggested that β-catenin is critical for normal pancreatic development although these reports reached somewhat different conclusions. In addition, the molecular mechanisms by which loss of β-catenin affects pancreas development are not well understood. The goals of this study then were; 1] to further investigate the role of β-catenin in pancreatic development using a conditional knockout approach and 2] to identify possible mechanisms by which loss of β-catenin disrupts pancreatic development. A Pdx1-cre mouse line was used to delete a floxed β-catenin allele specifically in the developing pancreas, and embryonic pancreata were studied by immunohistochemistry and microarray analysis. Results Pdx1-cre floxed β-catenin animals were viable but demonstrated small body size and shortened median survival. The pancreata from knockout mice were hypoplastic and histologically demonstrated a striking paucity of exocrine pancreas, acinar to duct metaplasia, but generally intact pancreatic islets containing all lineages of endocrine cells. In animals with extensive acinar hypoplasia, putative hepatocyte transdifferention was occasionally observed. Obvious and uniform pancreatic hypoplasia was observed by embryonic day E16.5. Transcriptional profiling of Pdx1-cre floxed β-catenin embryonic pancreata at E14.5, before there was a morphological phenotype, revealed significant decreases in the β-catenin target gene N-myc, and the basic HLH transcription factor PTF1, and an increase of several pancreatic zymogens compared to control animals. By E16.5, there was a dramatic loss of exocrine markers and an increase in Hoxb4, which is normally expressed anterior to the pancreas. Conclusion We conclude that β-catenin expression is required for development of the exocrine pancreas, but is not required for development of the endocrine compartment. In contrast, β-catenin/Wnt signaling appears to be critical for proliferation of PTF1+ nascent acinar cells and may also function, in part, to maintain an undifferentiated state in exocrine/acinar cell precursors. Finally, β-catenin may be required to maintain positional identity of the pancreatic endoderm along the anterior-posterior axis. This data is consistent with the findings of frequent β-catenin mutations in carcinomas of acinar cell lineage seen in humans.
Collapse
Affiliation(s)
- James M Wells
- Department of Developmental Biology, Cincinnati Children's Hospital Research 45267, Cincinnati, OH 45267, USA
| | - Farzad Esni
- The Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | | | - Bruce J Aronow
- Department of Developmental Biology, Cincinnati Children's Hospital Research 45267, Cincinnati, OH 45267, USA
| | - William Stuart
- Department of Surgery, Division of Surgical Oncology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Chelsea Combs
- Department of Pathology and Laboratory Medicine, USA
| | | | - Steven D Leach
- The Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Andrew M Lowy
- Department of Surgery, Division of Surgical Oncology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
154
|
Chan KM, Raikwar SP, Zavazava N. Strategies for differentiating embryonic stem cells (ESC) into insulin-producing cells and development of non-invasive imaging techniques using bioluminescence. Immunol Res 2007; 39:261-270. [PMID: 17917070 DOI: 10.1007/s12026-007-0070-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/22/2023]
Abstract
Diabetes is a chronic autoimmune disease that affects 4-5% of the world's population. If the present trends continue, diabetes would soon become a major/leading health problem worldwide. Hence there is an urgent need to develop novel approaches for the treatment of diabetes. While transplantation of the pancreas or that of isolated pancreatic islets can lead to the cure of the disease in some patients, immunological complications and the chronic shortage of donors makes it impossible to adequately treat all patients. Interestingly, embryonic stem cells (ESC) have emerged as a possible source of pluripotent cells that can be coaxed into insulin-producing cells (IPCs) that can be used to treat diabetes. However, until appropriate protocols have been established, this new technology will be difficult to tap into. Our laboratory is interested in developing new strategies for harnessing the pluripotency of ESC and differentiating them into IPCs that are stable and will continue to produce insulin in vivo. A second aspect is the non-availability of non-invasive imaging protocols. We show here that transcriptionally targeted luciferase expression can be used successfully to non-invasively monitor the transplanted cells in vivo.
Collapse
Affiliation(s)
- Kun-Ming Chan
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine & VAMC Iowa City, University of Iowa, 200 Hawkins Dr., C51-F, Iowa City, IA 52241, USA.
| | | | | |
Collapse
|
155
|
diIorio P, Alexa K, Choe SK, Etheridge L, Sagerström CG. TALE-family homeodomain proteins regulate endodermal sonic hedgehog expression and pattern the anterior endoderm. Dev Biol 2006; 304:221-31. [PMID: 17289013 PMCID: PMC1868511 DOI: 10.1016/j.ydbio.2006.12.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Revised: 12/11/2006] [Accepted: 12/12/2006] [Indexed: 01/29/2023]
Abstract
sonic hedgehog (shh) is expressed in anterior endoderm, where it is required to repress pancreas gene expression and to pattern the endoderm, but the pathway controlling endodermal shh expression is unclear. We find that expression of meis3, a TALE class homeodomain gene, coincides with shh expression in the endoderm of zebrafish embryos. Using a dominant negative construct or anti-sense morpholino oligos (MOs) to disrupt meis3 function, we observe ectopic insulin expression in anterior endoderm. This phenotype is also observed when meis3 MOs are targeted to the endoderm, suggesting that meis3 acts within the endoderm to restrict insulin expression. We also find that meis3 is required for endodermal shh expression, indicating that meis3 acts upstream of shh to restrict insulin expression. Loss of pbx4, a TALE gene encoding a Meis cofactor, produces the same phenotype as loss of meis3, consistent with Meis3 acting in a complex with Pbx4 as reported in other systems. Lastly, we observe a progressive anterior displacement of endoderm-derived organs upon disruption of meis3 or pbx4, apparently as a result of underdevelopment of the pharyngeal region. Our data indicate that meis3 and pbx4 regulate shh expression in anterior endoderm, thereby influencing patterning and growth of the foregut.
Collapse
Affiliation(s)
- Phillip diIorio
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Kristen Alexa
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
| | - Seong-Kyu Choe
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
| | - Letitiah Etheridge
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
| | - Charles G. Sagerström
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
- *To whom correspondence should be addressed: Department of Biochemistry and Molecular Pharmacology, 364 Plantation Street/LRB 822, Worcester, MA 01605, Phone: (506) 856-8006, Fax: (508) 856-8007,
| |
Collapse
|
156
|
Simeone DM, Zhang L, Treutelaar MK, Zhang L, Graziano K, Logsdon CD, Burant CF. Islet hypertrophy following pancreatic disruption of Smad4 signaling. Am J Physiol Endocrinol Metab 2006; 291:E1305-16. [PMID: 16735447 DOI: 10.1152/ajpendo.00561.2005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
To investigate the role of transforming growth factor (TGF)-beta family signaling in the adult pancreas, a transgenic mouse (E-dnSmad4) was created that expresses a dominant-negative Smad4 protein driven by a fragment of the elastase promoter. Although E-dnSmad4 mice have normal growth, pancreas weight, and pancreatic exocrine and ductal histology, beginning at 4-6 wk of age, E-dnSmad4 mice show an age-dependent increase in the size of islets. In parallel, an expanded population of replicating cells expressing the E-dnSmad4 transgene is found in the stroma between the enlarged islets and pancreatic ducts. Despite the marked enlargement, E-dnSmad4 islets contain normal ratios and spatial organization of endocrine cell subtypes and have normal glucose homeostasis. Replication of cells derived from primary duct cultures of wild-type mice, but not E-dnSmad4 mice, was inhibited by the addition of TGF-beta family proteins, demonstrating a cell-autonomous effect of the transgene. These data show that, in the adult pancreas, TGF-beta family signaling plays a role in islet size by regulating the growth of a pluripotent progenitor cell residing in the periductal stroma of the pancreas.
Collapse
Affiliation(s)
- Diane M Simeone
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | |
Collapse
|
157
|
Cano DA, Sekine S, Hebrok M. Primary cilia deletion in pancreatic epithelial cells results in cyst formation and pancreatitis. Gastroenterology 2006; 131:1856-69. [PMID: 17123526 DOI: 10.1053/j.gastro.2006.10.050] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2006] [Accepted: 10/12/2006] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Defects in cilia formation or function have been implicated in several human genetic diseases, including polycystic kidney disease (PKD), Bardet-Biedl syndrome, and primary ciliary dyskinesia. Pancreatic lesions are found in approximately 10% of PKD patients, suggesting a connection between cilia defects and pancreatic pathologies. Here, we investigate the role of cilia in pancreas formation and function by analyzing mice that lack cilia in pancreatic cells. METHODS Using Cre/lox technology, we conditionally inactivated Kif3a, the gene encoding for a subunit of the kinesin-2 complex that is essential for cilia formation, in pancreatic epithelia. Kif3a mice were studied by immunohistochemical and biochemical methods to assess the morphology and differentiation status of pancreatic cells. RESULTS Tissue-specific loss of Kif3a in pancreatic cells resulted in severe pancreatic abnormalities including acinar-to-ductal metaplasia, fibrosis, and lipomatosis. Ductal metaplasia appears to be due to expansion of ductal cells rather than transdifferentiation of acinar cells. Cyst formation, aberrant ductal morphology, and extensive fibrosis associated with severe adhesion to adjacent organs were commonly observed in aged Kif3a mutant mice. Deletion of Kif3a using different pancreas-specific Cre strains suggests that these pancreatic phenotypes might be caused by the absence of cilia in ductal cells. Activation of transforming growth factor beta and Mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (MEK/ERK) pathways may play a role in these phenotypes. CONCLUSIONS These results demonstrate that the absence of cilia in pancreatic cells produces pancreatic lesions that resemble those found in patients with chronic pancreatitis or cystic fibrosis.
Collapse
Affiliation(s)
- David A Cano
- Diabetes Center, Department of Medicine, University of California San Francisco, California 94143, USA
| | | | | |
Collapse
|
158
|
Lee K, Jeong J, Tsai MJ, Tsai S, Lydon JP, DeMayo FJ. Molecular mechanisms involved in progesterone receptor regulation of uterine function. J Steroid Biochem Mol Biol 2006; 102:41-50. [PMID: 17067792 PMCID: PMC2562605 DOI: 10.1016/j.jsbmb.2006.09.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The ovarian steroid hormone progesterone is a major regulator of uterine function. The actions of this hormone is mediated through its cognate receptor, the progesterone receptor, Pgr. Ablation of the Pgr has shown that this receptor is critical for all female reproductive functions including the ability of the uterus to support and maintain the development of the implanting mouse embryo. High density DNA microarray analysis has identified direct and indirect targets of Pgr action. One of the targets of Pgr action is a member of the Hedgehog morphogen Indian Hedgehog, Ihh. Ihh and members of the Hh signaling cascade show a coordinate expression pattern in the mouse uterus during the preimplantation period of pregnancy. The expression of Ihh and its receptor Patched-1, Ptc1, as well as, down stream targets of Ihh-Ptch1 signaling, such as the orphan nuclear receptor COUP-TF II show that this morphogen pathway mediates communication between the uterine epithelial and stromal compartments. The members of the Ihh signaling axis may function to coordinate the proliferation, vascularization and differentiation of the uterine stroma during pregnancy. This analysis demonstrates that progesterone regulates uterine function in the mouse by coordinating the signals from the uterine epithelium to stroma in the preimplantation mouse uterus.
Collapse
Affiliation(s)
- K Lee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
159
|
Koizumi M, Nagai K, Kida A, Kami K, Ito D, Fujimoto K, Kawaguchi Y, Doi R. Forced expression of PDX-1 induces insulin production in intestinal epithelia. Surgery 2006; 140:273-80. [PMID: 16904980 DOI: 10.1016/j.surg.2006.06.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Revised: 05/12/2006] [Accepted: 06/17/2006] [Indexed: 02/07/2023]
Abstract
BACKGROUND In the developmental stage, pancreas derives from the endodermal cells where the transcription factor, pancreatic duodenal homeobox gene-1 (pdx-1) is expressed. In adulthood, pdx-1 expression is localized to pancreatic beta cells, which is necessary for maintenance of beta cell function. Recently, ectopic expression of pdx-1 in the liver successfully induced insulin production and ameliorated hyperglycemia. Our study was designed to investigate the effects of forced expression of pdx-1 in ileal epithelia by adenovirus-mediated gene transfer. METHODS The recombinant, replication-deficient adenovirus carrying the pdx-1 gene was constructed using the COS-TPC method. ICR mice were treated with intraperitoneal injection of 220 mg/kg streptozotocin (STZ). After determining the hyperglycemia, a loop of ileum was constructed and the adenovirus solutions (Ad-pdx-1 and Ad-lacZ 1 x 10(8) PFU/body) were injected into the lumen of the ileal loop. In this model, immunohistochemical or fluorescent analyses of PDX-1 and insulin in the adenovirus-infected ileal epithelia were carried out. Reverse transcription polymerase chain reaction of pdx-1 and other pancreatic markers were investigated. Blood glucose concentrations were measured by drawing blood from ocular veins. Immunoreactive insulin extracted from the adenovirus-infected ileum was measured. RESULTS Ad-pdx-1 induced ectopic PDX-1 expression in the ileum. The PDX-1 positive cells in the ileal epithelia were positive for insulin; mRNA of insulin-1, insulin-2 and pdx-1 were expressed in mice infected with Ad-pdx-1. Hyperglycemia was improved in STZ-treated mice infected with Ad-pdx-1. Immunoreactive insulin in the ileum extract was increased significantly in mice with Ad-pdx-1. CONCLUSIONS Gene transfer of PDX-1 in intestinal epithelia could be a promising strategy for diabetes mellitus by inducing ectopic insulin producing cells.
Collapse
|
160
|
Lee K, Jeong J, Kwak I, Yu CT, Lanske B, Soegiarto DW, Toftgard R, Tsai MJ, Tsai S, Lydon JP, DeMayo FJ. Indian hedgehog is a major mediator of progesterone signaling in the mouse uterus. Nat Genet 2006; 38:1204-9. [PMID: 16951680 DOI: 10.1038/ng1874] [Citation(s) in RCA: 220] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2006] [Accepted: 08/03/2006] [Indexed: 01/01/2023]
Abstract
The hedgehog family of morphogens are regulators of cell proliferation, differentiation and cell-cell communication. These morphogens have been shown to have important roles in organogenesis, spermatogenesis, stem cell maintenance and oncogenesis. Indian hedgehog (encoded by Ihh) has been shown to be expressed in the uterine epithelium under the control of the steroid hormone, progesterone. Although in vivo and in vitro studies have shown that progesterone achieves its effects on uterine function through epithelial-stromal cross-talk, molecular mediator(s) for this cellular communication pathway have not been elucidated. Using new experimental approaches that ablate Ihh specifically in Pgr-positive uterine cells of the mouse, we demonstrate that Ihh is an essential mediator of Pgr action in the uterus, and expression of this factor is critical in mediating the communication between the uterine epithelium and stroma required for embryo implantation.
Collapse
Affiliation(s)
- Kevin Lee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Asayesh A, Sharpe J, Watson RP, Hecksher-Sørensen J, Hastie ND, Hill RE, Ahlgren U. Spleen versus pancreas: strict control of organ interrelationship revealed by analyses of Bapx1-/- mice. Genes Dev 2006; 20:2208-13. [PMID: 16912273 PMCID: PMC1553204 DOI: 10.1101/gad.381906] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Accepted: 05/31/2006] [Indexed: 01/12/2023]
Abstract
During early stages of pancreatic development, the mesenchyme that contributes to the spleen overlies the dorsal pancreatic endoderm. Here, we show that interactions between splenic mesenchyme and pancreas proceed via a highly orchestrated morphogenetic program. Disruption of morphogenesis, as occurs in the Bapx1(Nkx3.2)(-/-) embryo, results in transformation of these tissues into well-organized, ectopic gut-like structures. Bapx1 plays a crucial organizing role effecting position and separation of the spleen and pancreas to prevent this metaplastic transformation. Similar transformations occur in organ cultures employing wild-type pancreatic endoderm and spleen mesenchyme, revealing the developmental plasticity of the pancreas and that precise spatial and temporal control of tissue interactions are required for development of both organs.
Collapse
Affiliation(s)
- Amir Asayesh
- Umeå Centre for Molecular Medicine, Umeå University, S-901 87, Umeå, Sweden
| | | | | | | | | | | | | |
Collapse
|
162
|
Varnat F, Heggeler BBT, Grisel P, Boucard N, Corthésy-Theulaz I, Wahli W, Desvergne B. PPARbeta/delta regulates paneth cell differentiation via controlling the hedgehog signaling pathway. Gastroenterology 2006; 131:538-53. [PMID: 16890607 DOI: 10.1053/j.gastro.2006.05.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Accepted: 04/27/2006] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS All 4 differentiated epithelial cell types found in the intestinal epithelium derive from the intestinal epithelial stem cells present in the crypt unit, in a process whose molecular clues are intensely scrutinized. Peroxisome proliferator-activated receptor beta (PPARbeta) is a nuclear hormone receptor activated by fatty acids and is highly expressed in the digestive tract. However, its function in intestinal epithelium homeostasis is understood poorly. METHODS To assess the role of PPARbeta in the small intestinal epithelium, we combined various cellular and molecular approaches in wild-type and PPARbeta-mutant mice. RESULTS We show that the expression of PPARbeta is particularly remarkable at the bottom of the crypt of the small intestine where Paneth cells reside. These cells, which have an important role in the innate immunity, are strikingly affected in PPARbeta-null mice. We then show that Indian hedgehog (Ihh) is a signal sent by mature Paneth cells to their precursors, negatively regulating their differentiation. Importantly, PPARbeta acts on Paneth cell homeostasis by down-regulating the expression of Ihh, an effect that can be mimicked by cyclopamine, a known inhibitor of the hedgehog signaling pathway. CONCLUSIONS We unraveled the Ihh-dependent regulatory loop that controls mature Paneth cell homeostasis and its modulation by PPARbeta. PPARbeta currently is being assessed as a drug target for metabolic diseases; these results reveal some important clues with respect to the signals controlling epithelial cell fate in the small intestine.
Collapse
Affiliation(s)
- Frédéric Varnat
- Center for Integrative Genomics, National Research Centre Frontiers in Genetics, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
163
|
Abstract
Annular pancreas is a rare congenital anomaly occurring in 1 of every 12,000 to 15,000 live births [Nerwich N, Shi E. Neonatal duodenal obstruction: a review of 30 consecutive cases. Pediatr Surg Int 1994;9:47-50]. It may remain asymptomatic throughout life, present in adulthood, or present in infancy as a high intestinal obstruction. Review of the literature demonstrates only 8 cases of familial annular pancreas and no case of twins exhibiting the disease. We will describe a case of identical (monochorionic diamniotic) female twins with neonatal presentation of duodenal obstruction and annular pancreas, and discuss data supportive of a genetic etiology.
Collapse
Affiliation(s)
- Melissa C Hulvat
- Stritch School of Medicine, Loyola University Medical Center, Maywood, IL, USA
| | | | | | | |
Collapse
|
164
|
Shao J, Zhang L, Gao J, Li Z, Chen Z. Aberrant expression of PTCH (patched gene) and Smo (smoothened gene) in human pancreatic cancerous tissues and its association with hyperglycemia. Pancreas 2006; 33:38-44. [PMID: 16804411 DOI: 10.1097/01.mpa.0000222319.59360.21] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To investigate the prevalence of PTCH (patched gene) and Smo (smoothened gene) expression in human pancreatic cancerous tissues and its association with clinical characteristics. METHODS A rabbit polyclonal antibody against PTCH was prepared through the immunization of prokaryotic recombinant PTCH1170-1433 protein. The PTCH and Smo expression in 39 resected pancreas specimens from 28 patients with pancreatic cancer, 6 with chronic pancreatitis (as control), and 5 with pancreatic pseudocyst (as control) were detected by reverse transcriptase polymerase chain reaction and immunohistochemistry. The relationships between their expressions and pathological characteristics such as tumor sizes, degree of differentiation, nodal status, distant metastasis, and the blood sugar level were analyzed. RESULTS The prevalence of PTCH and Smo expressions in cancerous tissues were 71.4% (20/28) and 53.6% (15/28), respectively, whereas no expression in the nontumor pancreas tissues was found. Both PTCH and Smo expressions correlated with the low levels of tumor tissue differentiation (P < 0.05) and PTCH and Smo expressions in islet cells of cancerous tissues associated with hyperglycemia. CONCLUSIONS Because aberrant expressions of PTCH and Smo were common in human pancreatic carcinoma tissues and were associated with the low-level differentiation of tumor tissue and hyperglycemia, this indicated that these molecules played a fundamental role in pancreas tumorigenesis and were regarded as new targets for diagnosis and treatment of human pancreatic cancer.
Collapse
MESH Headings
- Adult
- Aged
- Carcinoma, Islet Cell/genetics
- Carcinoma, Islet Cell/metabolism
- Carcinoma, Islet Cell/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Differentiation
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hyperglycemia/genetics
- Hyperglycemia/metabolism
- Hyperglycemia/pathology
- Immunohistochemistry
- Male
- Middle Aged
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Patched Receptors
- Patched-1 Receptor
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Smoothened Receptor
Collapse
Affiliation(s)
- Jianguo Shao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | | |
Collapse
|
165
|
Fukaya M, Isohata N, Ohta H, Aoyagi K, Ochiya T, Saeki N, Yanagihara K, Nakanishi Y, Taniguchi H, Sakamoto H, Shimoda T, Nimura Y, Yoshida T, Sasaki H. Hedgehog signal activation in gastric pit cell and in diffuse-type gastric cancer. Gastroenterology 2006; 131:14-29. [PMID: 16831586 DOI: 10.1053/j.gastro.2006.05.008] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2005] [Accepted: 03/16/2006] [Indexed: 12/29/2022]
Abstract
BACKGROUND & AIMS Sonic hedgehog (SHH) and Indian hedgehog (IHH) have cell-specific actions in some organs. Only SHH has been shown to regulate parietal cell differentiation. This study examined whether SHH, 2 other ligands IHH and Desert hedgehog, and receptors or downstream targets are expressed in normal gastric epithelium or in intestinal and diffuse-type gastric cancers. The effects of a Hedgehog (Hh) inhibitor, cyclopamine, were assessed in primary gastric epithelium cultures and gastric cancer cell lines. METHODS Reverse-transcription polymerase chain reaction and immunostaining compared expression and localization of Hh signaling molecules and phenotypic markers of pit, neck, and gland cells in situ and in cultured cells treated with cyclopamine. Bromodeoxyuridine staining assessed the effects of cyclopamine on proliferation. RESULTS Hh signaling molecules were expressed differentially in pit, neck, and gland cells. IHH co-expressed with most downstream targets in the pit. IHH, SHH, Patched (PTCH), Smoothened (SMO), and downstream targets were expressed more frequently and highly in the diffuse as compared with intestinal type cancers. In diffuse cancers, IHH was expressed in cells with an epithelial phenotype and SHH in cells with a mesenchymal phenotype. Cyclopamine reduced the number of cells with a pit phenotype but not a gland phenotype in primary cultures. Cyclopamine had particularly potent effects of inhibiting the growth of cell lines that expressed high levels of SMO. CONCLUSIONS Expression of IHH and downstream targets correlates with pit cells. IHH and SMO may be useful biomarkers of diffuse cancers that may show growth inhibition with Hh antagonists such as cyclopamine.
Collapse
Affiliation(s)
- Masahide Fukaya
- Genetics Division, National Cancer Center Research Institute, Tsukiji, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Georgia S, Soliz R, Li M, Zhang P, Bhushan A. p57 and Hes1 coordinate cell cycle exit with self-renewal of pancreatic progenitors. Dev Biol 2006; 298:22-31. [PMID: 16899237 DOI: 10.1016/j.ydbio.2006.05.036] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 05/19/2006] [Accepted: 05/24/2006] [Indexed: 01/31/2023]
Abstract
In developing organs, the regulation of cell proliferation and cell cycle exit is coordinated. How this coordination is achieved, however, is not clear. We show that the cyclin kinase inhibitor p57 regulates cell cycle exit of progenitors during the early stages of pancreas formation. In the absence of p57, the number of cycling progenitors increases, although expansion of progenitor population is prevented by apoptosis. We report that p57 is a direct target of transcriptional repression by Notch effector, Hes1. Inactivation of Hes1 results in the upregulation of p57 expression in progenitors, leading to cell cycle arrest, precocious differentiation and depletion of the progenitor pool. We present evidence that in p57/Hes1 double null embryos, the absence of apoptosis results in the expansion of the progenitor population. We propose that Hes1 and p57 not only coordinate cell cycle exit and self-renewal of pancreatic progenitors during an early stage in organogenesis to regulate the number of pancreatic progenitors, but could also constitute a surveillance system to eliminate cells with aberrant cell cycle characteristics.
Collapse
Affiliation(s)
- Senta Georgia
- Larry Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | |
Collapse
|
167
|
Monzo M, Moreno I, Artells R, Ibeas R, Navarro A, Moreno J, Hernandez R, Granell M, Pie J. Sonic hedgehog mRNA expression by real-time quantitative PCR in normal and tumor tissues from colorectal cancer patients. Cancer Lett 2006; 233:117-23. [PMID: 16473672 DOI: 10.1016/j.canlet.2005.03.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Revised: 03/01/2005] [Accepted: 03/04/2005] [Indexed: 11/16/2022]
Abstract
Shh is expressed in the early stages of embryogenesis and in the foregut development. Although Shh has been shown to be overexpressed in brain, pancreas, gastric and lung cancers, its role in the development of colorectal cancer has not been examined. We used real-time quantitative PCR to assess Shh mRNA expression levels in tumor and matched normal tissue from 57 colorectal cancer patients and correlated the results with patient clinicopathological characteristics. Shh expression levels were higher in tumor tissue than in normal tissue from the same patient (P=0.00001). Higher levels of Shh expression were associated with early stage disease (P=0.02). Shh overexpression may influence the development of colorectal cancer.
Collapse
Affiliation(s)
- Mariano Monzo
- Department of Anatomy, Faculty Medicine UB, Casanova 143, 08036 Barcelona, and Department of Medical Oncology, Hospital Municipal Badalona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Ta M, Choi Y, Atouf F, Park CH, Lumelsky N. The defined combination of growth factors controls generation of long-term-replicating islet progenitor-like cells from cultures of adult mouse pancreas. Stem Cells 2006; 24:1738-49. [PMID: 16556710 DOI: 10.1634/stemcells.2005-0367] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Application of pancreatic islet transplantation to treatment of diabetes is severely hampered by the inadequate islet supply. This problem could in principle be overcome by generating islet cells from adult pancreas in vitro. Although it is possible to obtain replicating cells from cultures of adult pancreas, these cells, when significantly expanded in vitro, progressively lose pancreatic-specific gene expression, including that of a "master" homeobox transcription factor Pdx1. Here we show for the first time that long-term proliferating islet progenitor-like cells (IPLCs) stably expressing high levels of Pdx1 and other genes that control early pancreatic development can be derived from cultures of adult mouse pancreas under serum-free defined culture conditions. Moreover, we show that cells derived thus can be maintained in continuous culture for at least 6 months without any substantial loss of early pancreatic phenotype. Upon growth factor withdrawal, the IPLCs organize into cell clusters and undergo endocrine differentiation of various degrees in a line-dependent manner. We propose that our experimental strategy will provide a framework for developing efficient approaches for ex vivo expansion of islet cell mass.
Collapse
Affiliation(s)
- Malancha Ta
- Islet and Autoimmunity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
169
|
Abstract
The hedgehog (Hh) family of genes, sonic hedgehog (Shh), Indian hedgehog (Ihh), and desert hedgehog (Dhh) encode signaling molecules that regulate multiple functions during organ development and in adult tissues. Altered hedgehog signaling has been implicated in disturbed organ development as well as in different degenerative and neoplastic human diseases. Hedgehog signaling plays an important role in determination the fate of the mesoderm of the gut tube, as well as in early pancreatic development, and islet cell function. Recently, it has been shown that deregulation of hedgehog signaling molecules contributes to the pathogenesis and progression of pancreatic cancer and of chronic pancreatitis. Inhibition of hedgehog signaling using hedgehog antagonists reduces pancreatic cancer cell growth in vitro and in vivo, thus holding promise of novel agents in the treatment of this devastating disease. In this review, we discuss the role of hedgehog signaling during pancreatic development, its role in the pathogenesis of both chronic pancreatitis and pancreatic cancer, and lastly, the implications of this newly available information with regards to treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Hany Kayed
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
170
|
Hruban RH, Adsay NV, Albores-Saavedra J, Anver MR, Biankin AV, Boivin GP, Furth EE, Furukawa T, Klein A, Klimstra DS, Kloppel G, Lauwers GY, Longnecker DS, Luttges J, Maitra A, Offerhaus GJA, Pérez-Gallego L, Redston M, Tuveson DA. Pathology of genetically engineered mouse models of pancreatic exocrine cancer: consensus report and recommendations. Cancer Res 2006; 66:95-106. [PMID: 16397221 DOI: 10.1158/0008-5472.can-05-2168] [Citation(s) in RCA: 310] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Several diverse genetically engineered mouse models of pancreatic exocrine neoplasia have been developed. These mouse models have a spectrum of pathologic changes; however, until now, there has been no uniform nomenclature to characterize these changes. An international workshop, sponsored by The National Cancer Institute and the University of Pennsylvania, was held from December 1 to 3, 2004 with the goal of establishing an internationally accepted uniform nomenclature for the pathology of genetically engineered mouse models of pancreatic exocrine neoplasia. The pancreatic pathology in 12 existing mouse models of pancreatic neoplasia was reviewed at this workshop, and a standardized nomenclature with definitions and associated images was developed. It is our intention that this nomenclature will standardize the reporting of genetically engineered mouse models of pancreatic exocrine neoplasia, that it will facilitate comparisons between genetically engineered mouse models and human pancreatic disease, and that it will be broad enough to accommodate newly emerging mouse models of pancreatic neoplasia.
Collapse
Affiliation(s)
- Ralph H Hruban
- Department of Pathology, The Sol Goldman Center for Pancreatic Cancer Research, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21231, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Bort R, Signore M, Tremblay K, Martinez Barbera JP, Zaret KS. Hex homeobox gene controls the transition of the endoderm to a pseudostratified, cell emergent epithelium for liver bud development. Dev Biol 2006; 290:44-56. [PMID: 16364283 DOI: 10.1016/j.ydbio.2005.11.006] [Citation(s) in RCA: 198] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 10/26/2005] [Accepted: 11/04/2005] [Indexed: 11/18/2022]
Abstract
Little is known about the mechanism by which embryonic liver, lung, and pancreas progenitor cells emerge from the endodermal epithelium to initiate organogenesis. Understanding this process and its genetic control provides insight into ontogeny, developmental abnormalities, and tissue regeneration. We find that shortly after hepatic endoderm cells are specified, they undergo a transition from a columnar, gut morphology to a pseudostratified morphology, with concomitant "interkinetic nuclear migration" (INM) during cell division. INM is a hallmark of pseudostratified epithelia and the process used by neural progenitors to emerge from the neural epithelium. We find that the transition of the hepatic endoderm, but not the neural epithelium, to a pseudostratified epithelium is dependent upon the cell-autonomous activity of the homeobox gene Hex. In the absence of Hex, hepatic endoderm cells survive but maintain a columnar, simple epithelial phenotype and ectopically express Shh and other genes characteristic of the midgut epithelium. Thus, Hex promotes endoderm organogenesis by promoting the transition to a pseudostratified epithelium, which in turn allows hepatoblasts to emerge into the stromal environment and continue differentiating.
Collapse
Affiliation(s)
- Roque Bort
- Cell and Developmental Biology Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | | | | | | | | |
Collapse
|
172
|
Embryonic stem cell-based therapy for the treatment of diabetes mellitus: a work in progress. Curr Opin Organ Transplant 2006. [DOI: 10.1097/01.mot.0000203883.87439.27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
173
|
Sicklick JK, Li YX, Jayaraman A, Kannangai R, Qi Y, Vivekanandan P, Ludlow JW, Owzar K, Chen W, Torbenson MS, Diehl AM. Dysregulation of the Hedgehog pathway in human hepatocarcinogenesis. Carcinogenesis 2005; 27:748-57. [PMID: 16339184 DOI: 10.1093/carcin/bgi292] [Citation(s) in RCA: 215] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hedgehog (Hh) pathway activation promotes tumors in several endodermally derived tissues, but its role in the pathogenesis of hepatocellular carcinoma (HCC) is unknown. Although normal hepatocytes lack Hh signaling, activation of the Hh pathway in endodermal progenitors is required for liver development. Thus, we hypothesized that hepatocarcinogenesis may involve regulation of Hh signaling. This pathway is activated when Hh ligand binds to its receptor, Patched (PTC). In an unoccupied state, PTC normally functions as a tumor suppressor that inhibits Smoothened (SMO), a proto-oncoprotein, from activating downstream components and transcription of target genes. Here we show that in HCCs, overexpression of the Smo proto-oncogene, as well as an increase in the stoichiometric ratio of Smo to Ptc mRNA levels, correlated with tumor size, a prognostic indicator in HCC biology. In one tumor we identified a novel Smo mutation in an evolutionarily conserved residue. We also demonstrated that HCC cell lines (HepG2 and Hep3B) expressed Hh pathway components and activated Hh transcriptional targets. In Hep3B cells, cyclopamine, an inhibitor of wild-type SMO, had no effect, but KAAD-cyclopamine, a blocker of oncogenic SMO, inhibited Hh signaling activity by 50%, decreased expression of the hepatocarcinogenic oncogene, c-myc, by 8-fold, and inhibited the growth rate of Hep3B cells by 94%. These data support our hypothesis that Hh signaling is dysregulated in human hepatocarcinogenesis. We demonstrate that overexpression and/or tumorigenic activation of the Smo proto-oncogene mediates c-myc overexpression which plays a critical role in hepatocarcinogenesis and suggests that Smo is a prognostic factor in HCC tumorigenesis.
Collapse
Affiliation(s)
- Jason K Sicklick
- Department of Surgery and Division of Surgical Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Lees C, Howie S, Sartor RB, Satsangi J. The hedgehog signalling pathway in the gastrointestinal tract: implications for development, homeostasis, and disease. Gastroenterology 2005; 129:1696-710. [PMID: 16285967 DOI: 10.1053/j.gastro.2005.05.010] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2005] [Accepted: 04/27/2005] [Indexed: 12/21/2022]
Abstract
The hedgehog signalling pathway is critical to normal mammalian gastrointestinal development. Through epithelial-mesenchymal interactions, hedgehog signalling ensures appropriate axial patterning of the embryonic gut. Congenital abnormalities, including malrotations, anorectal malformations, and tracheoesophageal fistula are associated with germ-line mutations/deletion of genes encoding hedgehog signalling components in man and present in genetically engineered animal models. In adults, there is evidence that the pathway plays a role in maintaining stem cell populations in the stomach and directing epithelial cell differentiation in the intestine. Recent data implicate hedgehog signalling in the formation and maintenance of a number of malignancies, including those of the upper gastrointestinal (GI) tract and pancreas, in which abrogation of the pathway offers a novel therapeutic approach in animal models. Most recently, evidence in vitro indicates that there is a recapitulation of embryonic hedgehog signalling in acute epithelial injury and chronic inflammation, a finding with key implications for inflammatory disorders of the intestine, such as inflammatory bowel diseases. This pathway may provide an important link between chronic inflammation and cancer. We summarize the available evidence demonstrating that this developmental pathway has continuing roles in adult homeostasis and is dysregulated in malignancy and inflammation of the gastrointestinal tract.
Collapse
Affiliation(s)
- Charlie Lees
- Gastrointestinal Unit, School of Molecular and Clinical Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.
| | | | | | | |
Collapse
|
175
|
Kim HJ, Schleiffarth JR, Jessurun J, Sumanas S, Petryk A, Lin S, Ekker SC. Wnt5 signaling in vertebrate pancreas development. BMC Biol 2005; 3:23. [PMID: 16246260 PMCID: PMC1276788 DOI: 10.1186/1741-7007-3-23] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2005] [Accepted: 10/24/2005] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Signaling by the Wnt family of secreted glycoproteins through their receptors, the frizzled (Fz) family of seven-pass transmembrane proteins, is critical for numerous cell fate and tissue polarity decisions during development. RESULTS We report a novel role of Wnt signaling in organogenesis using the formation of the islet during pancreatic development as a model tissue. We used the advantages of the zebrafish to visualize and document this process in living embryos and demonstrated that insulin-positive cells actively migrate to form an islet. We used morpholinos (MOs), sequence-specific translational inhibitors, and time-lapse imaging analysis to show that the Wnt-5 ligand and the Fz-2 receptor are required for proper insulin-cell migration in zebrafish. Histological analyses of islets in Wnt5a(-/-) mouse embryos showed that Wnt5a signaling is also critical for murine pancreatic insulin-cell migration. CONCLUSION Our results implicate a conserved role of a Wnt5/Fz2 signaling pathway in islet formation during pancreatic development. This study opens the door for further investigation into a role of Wnt signaling in vertebrate organ development and disease.
Collapse
Affiliation(s)
- Hyon J Kim
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
- Department of Molecular, Cellular, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Jack R Schleiffarth
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455 USA
| | - Jose Jessurun
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455 USA
| | - Saulius Sumanas
- Department of Molecular, Cellular, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Anna Petryk
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455 USA
| | - Shuo Lin
- Department of Molecular, Cellular, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Stephen C Ekker
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
| |
Collapse
|
176
|
Lu D, Ventura-Holman T, Li J, McMurray RW, Subauste JS, Maher JF. Abnormal glucose homeostasis and pancreatic islet function in mice with inactivation of the Fem1b gene. Mol Cell Biol 2005; 25:6570-7. [PMID: 16024793 PMCID: PMC1190348 DOI: 10.1128/mcb.25.15.6570-6577.2005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Type 2 diabetes mellitus is a disorder of glucose homeostasis involving complex gene and environmental interactions that are incompletely understood. Mammalian homologs of nematode sex determination genes have recently been implicated in glucose homeostasis and type 2 diabetes mellitus. These are the Hedgehog receptor Patched and Calpain-10, which have homology to the nematode tra-2 and tra-3 sex determination genes, respectively. Here, we have developed Fem1b knockout (Fem1b-KO) mice, with targeted inactivation of Fem1b, a homolog of the nematode fem-1 sex determination gene. We show that the Fem1b-KO mice display abnormal glucose tolerance and that this is due predominantly to defective glucose-stimulated insulin secretion. Arginine-stimulated insulin secretion is also affected. The Fem1b gene is expressed in pancreatic islets, within both beta cells and non-beta cells, and is highly expressed in INS-1E cells, a pancreatic beta-cell line. In conclusion, these data implicate Fem1b in pancreatic islet function and insulin secretion, strengthening evidence that a genetic pathway homologous to nematode sex determination may be involved in glucose homeostasis and suggesting novel genes and processes as potential candidates in the pathogenesis of diabetes mellitus.
Collapse
Affiliation(s)
- Deyin Lu
- Veterans Affairs Medical Center-Research Svc. (151), 1500 E. Woodrow Wilson Blvd., Jackson, MS 39216, USA
| | | | | | | | | | | |
Collapse
|
177
|
Neureiter D, Zopf S, Dimmler A, Stintzing S, Hahn EG, Kirchner T, Herold C, Ocker M. Different capabilities of morphological pattern formation and its association with the expression of differentiation markers in a xenograft model of human pancreatic cancer cell lines. Pancreatology 2005; 5:387-397. [PMID: 15980667 DOI: 10.1159/000086539] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2004] [Accepted: 03/28/2005] [Indexed: 12/11/2022]
Abstract
AIMS New concepts of tumorigenesis favor an unregulated process recapitulating different stages of embryogenic development with dysregulation of transition states. The aim of our study was to investigate the possibility of differentiation pathways of human pancreatic cancer cell lines in vivo. MATERIAL AND METHODS Different human pancreatic cancer cell lines (YAPC, DAN-G, CAPAN-1, PANC-1 and MIA PaCa-2) were implanted subcutaneously (3 x 10(6) cells) for 28 days in nude mice. Xenotransplants were characterized with histochemistry (HE, PAS), immunohistochemistry (cytokeratin (CK)7, CK8, CK18, CK19, CK20, vimentin, chromogranin A (Chr-A), alpha1-antichymotrypsin (alpha1-chym), beta-catenin, laminin-5, pancreatic and duodenal homeobox gene 1 (pdx-1), sonic hedgehog protein (shh), Patched (ptc)), Western blotting and real-time PCR (CK7, CK8, CK20, Chr-A, pdx-1, shh, ptc). RESULTS Depending on three major morphologic phenotypes of tumor cell xenotransplants (ductal (YAPC), ductal/solid (DAN-G, CAPAN-1), solid (PANC-1, MIA PaCa-2)), a decrease of CK7/CK19 was found, accompanied by an increase of CK8/18 and vimentin. Predominantly the CK7-positive ductal phenotype (YAPC and DAN-G) was associated with pdx-1 expression, whereas the CK8-positive solid phenotype was associated with shh/ptc expression on protein and mRNA level. Additionally, CK-20 expression was mainly linked to the ductal phenotype, co-localized with nuclear beta-catenin. The endocrine-exocrine transdifferentiation, as assessed by Chr-A and alpha1-chym, was on a constant low to moderate level in all xenotransplants. Finally, an intensive epithelial-mesenchymal interaction was observed by overexpression of laminin-5 at the invasion front. CONCLUSION The observed patterns of morphology and molecular differentiation in human pancreatic cancer xenografts indicate that these cancer cell lines have different capabilities of pattern formation in vivo associated with molecular differentiation markers, especially of embryonic pancreatic development.
Collapse
Affiliation(s)
- Daniel Neureiter
- Department of Pathology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
178
|
Abstract
Type 1 diabetes is caused by autoimmune destruction of pancreatic islet beta-cells. Management of this disease is burdensome both to the individual and society, costing over 100 billion US dollars annually. Shortage of pancreatic tissue, together with a lifetime requirement of immunosuppressive drugs to prevent rejection and recurrent disease, remain as major hurdles yet to be overcome prior to widespread applicability. Stem cells, with their potential of developing into pancreatic beta-cells, appear to be the best prospect for overcoming the islet shortage. Current investigation, however (both embryonic and adult stem cells), is still in the preliminary stage and several more years remain before they can potentially be used in the clinical setting. Procedures that reduce in vitro manipulation of cells and allow stem cells to develop into islets in vivo are crucial. Furthermore, the regeneration of existing islets is a distinct possibility. Simplistically, it might be hypothesized that down-regulation of autoimmunity may give the pancreas the breathing space to regenerate islets. Supplementation with factors known to induce beta-cell replication and neogenesis might further augment the regenerative processes. Clearly, islet-regeneration research will soon match the level of interest currently focused on in vitro stem cell-based approaches.
Collapse
Affiliation(s)
- Vijayakumar Ramiya
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA.
| | | |
Collapse
|
179
|
Kawahira H, Scheel DW, Smith SB, German MS, Hebrok M. Hedgehog signaling regulates expansion of pancreatic epithelial cells. Dev Biol 2005; 280:111-21. [PMID: 15766752 DOI: 10.1016/j.ydbio.2005.01.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2004] [Revised: 12/07/2004] [Accepted: 01/10/2005] [Indexed: 12/11/2022]
Abstract
Embryonic Hedgehog signaling is essential for proper tissue morphogenesis and organ formation along the developing gastrointestinal tract. Hedgehog ligands are expressed throughout the endodermal epithelium at early embryonic stages but excluded from the region that will form the pancreas. Ectopic activation of Hedgehog signaling at the onset of pancreas development has been shown to inhibit organ morphogenesis. In contrast, Hedgehog signaling components are found within pancreatic tissue during subsequent stages of development as well as in the mature organ, indicating that a certain level of pathway activation is required for normal organ development and function. Here, we ectopically activate the Hedgehog pathway midway through pancreas development via expression of either Sonic (Shh) or Indian Hedgehog (Ihh) under control of the human Pax4-promoter. Similar pancreatic defects are observed in both Pax4-Shh and Pax4-Ihh transgenic lines, suggesting that regulation of the overall level of Hedgehog activity is critical for proper pancreas development. We also show that Hedgehog signaling controls mesenchymal vs. epithelial tissue differentiation and that pathway activation impairs formation of epithelial progenitors. Thus, tight control of Hedgehog pathway activity throughout embryonic development ensures proper pancreas organogenesis.
Collapse
Affiliation(s)
- Hiroshi Kawahira
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
180
|
Perez SE, Cano DA, Dao-Pick T, Rougier JP, Werb Z, Hebrok M. Matrix metalloproteinases 2 and 9 are dispensable for pancreatic islet formation and function in vivo. Diabetes 2005; 54:694-701. [PMID: 15734845 PMCID: PMC2771170 DOI: 10.2337/diabetes.54.3.694] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Pancreatic islet formation is a highly regulated process that is initiated at the end of gestation in rodents. Endocrine precursor cells first form within the epithelium of duct-like structures and then delaminate from the epithelium, migrate, and cluster during the early stages of islet formation. The molecular mechanisms that regulate endocrine cell migration and islet formation are not well understood. Cell culture studies suggest that matrix metalloproteinases (MMPs) 2 and 9 are required for islet formation. To address whether MMP2 and MMP9 function are essential for endocrine cell migration and islet formation in vivo, we analyzed pancreas development in MMP2/MMP9 double-deficient mice. Our results show that islet architecture and function are unperturbed in these knockout mice, demonstrating that both MMP2 and MMP9 functions are dispensable for islet formation and endocrine cell differentiation. Our studies also show that a number of other MMPs are expressed at the time islet formation is initiated. This observation suggests that other MMPs may substitute for MMP2 and MMP9 loss in pancreatic tissue. However, islet formation is unaffected in transgenic mice with modified tissue inhibitor of metalloproteinase-1 (TIMP1) levels, suggesting that MMP activity may contribute little to islet morphogenesis in vivo.
Collapse
Affiliation(s)
- Sabina E. Perez
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California
| | - David A. Cano
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Trang Dao-Pick
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Jean-Phillipe Rougier
- Department of Anatomy, University of California San Francisco, San Francisco, California
| | - Zena Werb
- Department of Anatomy, University of California San Francisco, San Francisco, California
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
181
|
Abstract
Considerable progress has been made in the understanding of the sequential activation of signal transduction pathways and the expression of transcription factors during pancreas development. Much of this understanding has been obtained by analyses of the phenotypes of mice in which the expression of key genes has been disrupted (knockout mice). Knockout of the genes for Pdx1, Hlxb9, Isl1, or Hex results in an arrest of pancreas development at a very early stage (embryonic d 8-9). Disruption of genes encoding components of the Notch signaling pathway, e.g. Hes1 or neurogenin-3, abrogates development of the endocrine pancreas (islets of Langerhans). Disruption of transcription factor genes expressed more downstream in the developmental cascade (Beta2/NeuroD, Pax4, NKx2.2, and Nkx6.1) curtails the formation of insulin-producing beta-cells. An understanding of the importance of transcription factor genes during pancreas development has provided insights into the pathogenesis of diabetes, in which the mass of insulin-producing beta-cells is reduced.
Collapse
Affiliation(s)
- Joel F Habener
- Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Howard Hughes Medical Institute, Harvard Medical School, 55 Fruit Street, WEL320, Boston, Massachusetts 02114, USA.
| | | | | |
Collapse
|
182
|
Micallef SJ, Janes ME, Knezevic K, Davis RP, Elefanty AG, Stanley EG. Retinoic acid induces Pdx1-positive endoderm in differentiating mouse embryonic stem cells. Diabetes 2005; 54:301-5. [PMID: 15585742 DOI: 10.2337/diabetes.54.2.301] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We have generated an embryonic stem (ES) cell line in which sequences encoding green fluorescent protein (GFP) were targeted to the locus of the pancreatic-duodenal homeobox gene (Pdx1). Analysis of chimeric embryos derived from blastocyst injection of Pdx1(GFP/w) ES cells demonstrated that the pattern of GFP expression was consistent with that reported for the endogenous Pdx1 gene. By monitoring GFP expression during the course of ES cell differentiation, we have shown that retinoic acid (RA) can regulate the commitment of ES cells to form Pdx1(+) pancreatic endoderm. RA was most effective at inducing Pdx1 expression when added to cultures at day 4 of ES differentiation, a period corresponding to the end of gastrulation in the embryo. RT-PCR analysis showed that Pdx1-positive cells from day 8 cultures expressed the early endoderm markers Ptf1a, Foxa2, Hnf4alpha, Hnf1beta, and Hnf6, consistent with the notion that they corresponded to the early pancreatic endoderm present in the embryonic day 9.5 mouse embryo. These results demonstrate the utility of Pdx1(GFP/w) ES cells as a tool for monitoring the effects of factors that influence pancreatic differentiation from ES cells.
Collapse
Affiliation(s)
- Suzanne J Micallef
- Centre for Early Human Development, Monash Institute of Reproduction and Development, 27-31 Wright St., Clayton, Victoria 3168, Australia
| | | | | | | | | | | |
Collapse
|
183
|
Moore-Scott BA, Manley NR. Differential expression of Sonic hedgehog along the anterior–posterior axis regulates patterning of pharyngeal pouch endoderm and pharyngeal endoderm-derived organs. Dev Biol 2005; 278:323-35. [PMID: 15680353 DOI: 10.1016/j.ydbio.2004.10.027] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2004] [Revised: 09/20/2004] [Accepted: 10/25/2004] [Indexed: 12/01/2022]
Abstract
Previous studies have implicated Sonic hedgehog (Shh) as an important regulator of pharyngeal region development. Here we show that Shh is differentially expressed within the pharyngeal endoderm along the anterior-posterior axis. In Shh-/- mutants, the pharyngeal pouches and arches formed by E9.5 and marker expression showed that initial patterning was normal. However, by E10.5-E11.0, the first arch had atrophied and the first pouch was missing. Although small, the second, third, and fourth arches and pouches were present. The expression patterns of Fgf8, Pax1, and Bmp4 suggested that pouch identity was abnormal at E10.5 and that Shh is a negative regulator of these genes in the pouches. Despite the loss of pouch identity and an increase in mesenchymal cell death, arch identity markers were expressed normally. Our data show that a Shh-dependent patterning mechanism is required to maintain pouch patterning, independent or downstream of arch identity. Changes in the distribution of Bmp4 and Gcm2 in the third pouch endoderm and subsequent organ phenotypes in Shh-/- mutants suggested that exclusion of Shh from the third pouch is required for dorsal-ventral patterning and for parathyroid specification and organogenesis. Furthermore, this function for Shh may be opposed by Bmp4. Our data suggest that, as in the posterior gut endoderm, exclusion of Shh expression from developing primordia is required for the proper development of pharyngeal-derived organs.
Collapse
Affiliation(s)
- Billie A Moore-Scott
- Institute for Molecular Medicine and Genetics, Medical College of Georgia, Augusta, GA 30912, USA
| | | |
Collapse
|
184
|
Abstract
Type 1 diabetes is one of the more costly chronic diseases of children and adolescents throughout North America and Europe, exhibiting an average estimated prevalence rate of nearly 0.2%. It occurs in genetically predisposed individuals when the immune system attacks and destroys specifically the insulin-producing beta cells of the pancreatic islets of Langerhans. While routine insulin therapy can provide diabetic patients with their daily insulin requirements, non-compliance and undetected hyperglycemic excursions often lead to subsequent long-term microvascular and macrovascular complications. The only real cure for type 1 diabetes is replacement of the beta cell mass, currently being accomplished through ecto-pancreatic transplantation and islet implantation. Both of these procedures suffer from a chronic shortage of available donor tissue in comparison to the number of potential recipients. To circumvent this need, three alternative approaches are being intensively investigated: (1) the production of surrogate cells by genetically modifying non-endocrine cells to secrete insulin in response to glucose challenge; (2) the trans-differentiation of non-endocrine stem/progenitor cells or mature cells to glucose-responsive adult tissue; and (3) the regulated differentiation of islet stem/progenitor cells to produce large numbers of mature, functional islets. In recent years, each of these approaches has made impressive advances, leading to the most important question, 'how soon will this new science be available to the patient?' In the present review, we discuss some of the recent advances, focusing primarily on the differentiation of islet stem cells to functional endocrine pancreas that may form the basis for future treatment.
Collapse
Affiliation(s)
- Ammon B Peck
- Department of Pathology, Immumology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville 32610, USA.
| | | |
Collapse
|
185
|
Lemos B, Yunes JA, Vargas FR, Moreira MAM, Cardoso AA, Seuánez HN. Phylogenetic footprinting reveals extensive conservation of Sonic Hedgehog (SHH) regulatory elements. Genomics 2005; 84:511-23. [PMID: 15498458 DOI: 10.1016/j.ygeno.2004.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2003] [Accepted: 05/28/2004] [Indexed: 11/27/2022]
Abstract
Sonic Hedgehog (SHH) plays a fundamental role in numerous developmental processes including morphogenesis of limbs, nervous system, and teeth. Using a Bayesian alignment algorithm for phylogenetic footprinting we analyzed approximately 28 kb of noncoding DNA in the SHH locus of human and mouse. This showed that the length of conserved noncoding sequences (4196 nt) shared by these species was approximately 3 times larger than the SHH coding sequence (1386 nt). Most segments were located in introns (53%) or within 2-kb regions upstream (16%) or downstream (20%) of the first and last SHH codon. Even though regions more than 2 kb upstream or downstream of the first and last SHH codon represented 57% (16 kb) of the sequence compared, they accounted for only 11% (494 nt) of the total length of conserved noncoding segments. One region of 650 nt downstream of SHH was identified as a putative scaffold/matrix attachment region (SMAR). Human-mouse analysis was complemented by sequencing in apes, monkeys, rodents, and bats, thus further confirming the evolutionary conservation of some segments. Gel-shift assays indicated that conserved segments are targeted by nuclear proteins and showed differences between two cell types that expressed different levels of SHH, namely human endothelial cells and breast cancer cells. The relevance of these findings with respect to regulation of SHH expression during normal and pathologic development is discussed.
Collapse
Affiliation(s)
- Bernardo Lemos
- Department of Genetics, Universidade Federal do Rio de Janeiro, Brazil.
| | | | | | | | | | | |
Collapse
|
186
|
Saitsu H, Komada M, Suzuki M, Nakayama R, Motoyama J, Shiota K, Ishibashi M. Expression of the mouseFgf15 gene is directly initiated by Sonic hedgehog signaling in the diencephalon and midbrain. Dev Dyn 2005; 232:282-92. [PMID: 15614767 DOI: 10.1002/dvdy.20236] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Sonic hedgehog (Shh) is a secreted molecule that is thought to regulate tissue growth and patterning in vertebrate embryos. Although it has been reported that Gli transcription factors mediate Shh signaling to the nucleus, little is known about developmental target genes of Gli. In the previous genetic study, we showed that Shh is required for Fgf15 expression in the diencephalon and midbrain. Here, we examined whether Fgf15 is a direct target of Shh signaling through Gli. Shh was expressed in the midline cells and Fgf15 in the medial region of the diencephalon/midbrain by the seven-somite stage. The Fgf15 expression domain coincided with that of Gli1 and overlapped with that of Gli2 at this stage. Fgf15 expression in the diencephalon/midbrain was greatly reduced in the seven-somite Shh mutant embryos. Transgenic analysis showed that the 3.6-kb 5'-flanking region of the Fgf15 gene is sufficient for induction of Fgf15 in the medial/ventral diencephalon/midbrain. Luciferase assay showed that the 3.6-kb Fgf15 enhancer/promoter was activated by Gli2. A Gli-binding site was located 1 kb upstream of the transcription start site and was required for expression in the medial/ventral diencephalon/midbrain in transgenic embryos and for activation in luciferase assay. These findings indicate that Fgf15 is directly regulated by Shh signaling through Gli proteins.
Collapse
MESH Headings
- Amino Acid Motifs
- Animals
- Base Sequence
- Binding Sites
- Brain/metabolism
- Cell Line
- Cell Nucleus/metabolism
- Cloning, Molecular
- Diencephalon/embryology
- Enhancer Elements, Genetic
- Fibroblast Growth Factors/biosynthesis
- Gene Expression Regulation, Developmental
- Genes, Reporter
- Glutathione Transferase/metabolism
- Hedgehog Proteins
- In Situ Hybridization
- Kruppel-Like Transcription Factors
- Luciferases/metabolism
- Mesencephalon/embryology
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Transgenic
- Models, Genetic
- Molecular Sequence Data
- Mutagenesis
- Mutation
- Nucleic Acid Hybridization
- Oncogene Proteins/biosynthesis
- Plasmids/metabolism
- Promoter Regions, Genetic
- Protein Structure, Tertiary
- RNA/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Trans-Activators/metabolism
- Transcription Factors/biosynthesis
- Transcription, Genetic
- Transgenes
- Zinc Finger Protein GLI1
- Zinc Finger Protein Gli2
- beta-Galactosidase/metabolism
Collapse
Affiliation(s)
- Hirotomo Saitsu
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | | | |
Collapse
|
187
|
Zhang Q, Davenport JR, Croyle MJ, Haycraft CJ, Yoder BK. Disruption of IFT results in both exocrine and endocrine abnormalities in the pancreas of Tg737(orpk) mutant mice. J Transl Med 2005; 85:45-64. [PMID: 15580285 DOI: 10.1038/labinvest.3700207] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
While relatively ignored for years as vestigial, cilia have recently become the focus of intense interest as organelles that result in severe pathologies when disrupted. Here, we further establish a connection between cilia dysfunction and disease by showing that loss of polaris (Tg737), an intraflagellar transport (IFT) protein required for ciliogenesis, causes abnormalities in the exocrine and endocrine pancreas of the Tg737(orpk) mouse. Pathology is evident late in gestation as dilatations of the pancreatic ducts that continue to expand postnatally. Shortly after birth, the acini become disorganized, undergo apoptosis, and are largely ablated in late stage pathology. In addition, serum amylase levels are elevated and carboxypeptidase is abnormally activated within the pancreas. Ultrastructural analysis reveals that the acini undergo extensive vacuolization and have numerous 'halo-granules' similar to that seen in induced models of pancreatitis resulting from duct obstruction. Intriguingly, although the acini are severely affected in Tg737(orpk) mutants, cilia and Tg737 expression are restricted to the ducts and islets and are not detected on acinar cells. Analysis of the endocrine pancreas in Tg737(orpk) mutants revealed normal differentiation and distribution of cell types in the islets. However, after fasting, mutant blood glucose levels are significantly lower than controls and when challenged in glucose tolerance tests, Tg737(orpk) mutants exhibited defects in glucose uptake. These findings are interesting in light of the recently proposed role for polaris, the protein encoded by the Tg737 gene, in the hedgehog pathway and hedgehog signaling in insulin production and glucose homeostasis.
Collapse
Affiliation(s)
- Qihong Zhang
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
188
|
Abstract
Embryonic stem cells hold potential in the fields of regenerative medicine, developmental biology, tissue regeneration, disease pathogenicity, and drug discovery. Embryonic stem (ES) cell lines are now available in primates, including man, rhesus, and cynomologous monkeys. Monkey ES cells serve as invaluable clinically relevant models for studies that can't be conducted in humans because of practical or ethical limitations, or in rodents because of differences in physiology and anatomy. Here, we review the current status of nonhuman primate research with ES cells, beginning with a description of their isolation, characterization, and availability. Substantial limitations still plague the use of primate ES cells, such as their required growth on feeder layers, poor cloning efficiency, and restricted availability. The ability to produce homogenous populations of both undifferentiated as well as differentiated phenotypes is an important challenge, and genetic approaches to achieving these objectives are discussed. Finally, safety, efficiency, and feasibility issues relating to the transplantation of ES-derived cells are considered.
Collapse
Affiliation(s)
- Don P Wolf
- Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97006, USA.
| | | | | | | |
Collapse
|
189
|
Blyszczuk P, Wobus AM. Stem cells and pancreatic differentiation in vitro. J Biotechnol 2004; 113:3-13. [PMID: 15380643 DOI: 10.1016/j.jbiotec.2004.03.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2003] [Revised: 12/08/2003] [Accepted: 03/03/2004] [Indexed: 12/11/2022]
Abstract
Cell therapy using pancreatic islets would be a promising therapy to treat diabetes. But, because of the limited supply of human donor islets, other cellular sources have to be considered. Stem cells characterized by extensive proliferation and differentiation capacity may be a valuable source for the in vitro generation of islets. Insulin-producing cells derived from embryonic stem (ES) cells have been shown to reverse experimentally induced diabetes in animal models. However, the oncogenic properties of ES cells are critical in the context of clinical applications and efficient cell-lineage selection systems need to be established. Future studies have to demonstrate whether somatic stem cells residing in adult tissues, such as bone marrow, pancreatic ducts, intestine or liver may provide alternatives to generate functional pancreatic endocrine cells.
Collapse
Affiliation(s)
- Przemyslaw Blyszczuk
- In Vitro Differentiation Group, Institute of Plant Genetics and Crop Plant Research (IPK), Corrensstr. 3, D-06466 Gatersleben, Germany
| | | |
Collapse
|
190
|
Gutiérrez-Frías C, Sacedón R, Hernández-López C, Cejalvo T, Crompton T, Zapata AG, Varas A, Vicente A. Sonic hedgehog regulates early human thymocyte differentiation by counteracting the IL-7-induced development of CD34+ precursor cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:5046-53. [PMID: 15470048 DOI: 10.4049/jimmunol.173.8.5046] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The Hedgehog (Hh) family of signaling molecules normally functions in the development of numerous tissues by regulating cellular differentiation and proliferation. Recent results have demonstrated that the different components of the Hh signaling pathway are expressed in the human thymus. In this study, we investigate the potential role of Sonic hedgehog (Shh) in human intrathymic T cell maturation. Results show that the expression of the two components of the Hh receptor, Patched and Smoothened, is mostly restricted to CD34+ precursor cells that are committing to the T cell lineage. Shh significantly increased the viability of CD34+ T cell precursors modulating bcl-2 and bax protein expression, and also inhibited their proliferation. The treatment of chimeric human-mouse fetal thymus organ cultures with Shh resulted in an arrested thymocyte differentiation and an accumulation of CD34+ progenitor cells. This effect was mainly attributed to the ability of Shh to counteract the IL-7-induced proliferation and differentiation of CD34+ cells. Shh down-regulated in the precursor cell population the expression of IL-7R as well as stromal-derived factor-1 chemokine receptor, CXCR4, and inhibited IL-7-dependent STAT5 phosphorylation. Therefore, Shh may function as a maintenance factor for intrathymic CD34+ precursor cells.
Collapse
Affiliation(s)
- Cruz Gutiérrez-Frías
- Department of Cell Biology, Faculty of Biology, Complutense University, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
191
|
Lin JW, Biankin AV, Horb ME, Ghosh B, Prasad NB, Yee NS, Pack MA, Leach SD. Differential requirement for ptf1a in endocrine and exocrine lineages of developing zebrafish pancreas. Dev Biol 2004; 274:491-503. [PMID: 15570689 DOI: 10.1016/j.ydbio.2004.07.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mammalian studies have implicated important roles for the basic helix-loop-helix transcription factor Ptf1a-p48 in the development of both exocrine and endocrine pancreas. We have cloned the Ptf1a-p48 ortholog in Danio rerio. Early zebrafish ptf1a expression is observed in developing hindbrain and in endodermal pancreatic precursors. Analysis of ptf1a and insulin expression reveals a population of exocrine precursors that, throughout early development, are temporally and spatially segregated from endocrine elements. Morpholino-mediated knockdown of ptf1a confirms early divergence of these endocrine and exocrine lineages. Ptf1a morphants lack differentiated exocrine pancreas, but maintain normal differentiation and organization of the principal islet. In addition to the exocrine phenotype, ptf1a knockdown also reduces the prevalence of a small population of anterior endocrine cells normally found outside the principal islet. Together, these findings suggest the presence of distinct ptf1a-dependent and ptf1a-independent precursor populations in developing zebrafish pancreas.
Collapse
Affiliation(s)
- John W Lin
- Department of Surgery, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | |
Collapse
|
192
|
Watkins DN, Peacock CD. Hedgehog signalling in foregut malignancy. Biochem Pharmacol 2004; 68:1055-60. [PMID: 15313401 DOI: 10.1016/j.bcp.2004.04.025] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2004] [Accepted: 04/14/2004] [Indexed: 12/20/2022]
Abstract
Hedgehog (Hh) signalling mediates axial patterning and stem cell fate in development. This is mediated by Sonic, Desert and Indian Hedgehogs whose morphogen gradients determine the level of signalling in recipient tissues. Aberrant, cell autonomous, ligand-dependent Hh signalling has recently been demonstrated in small cell lung cancer (SCLC), as well as in upper gastrointestinal malignancies arising from pancreas, esophagus and stomach. These tumors lack mutations in the Hh receptor PATCHED, identifying a mechanism of pathway activation distinct from Gorlin's syndrome associated neural and skin tumors. We believe that this phenomenon represents a conserved mechanism for establishing niche-independent stem cell fates in cancer which is essential for malignant transformation and metastasis. Specific inhibition of Hh signalling by the naturally occurring plant alkaloid cyclopamine provides the opportunity for pharmacologic assessment of the role of Hh signalling in these tumors. Cyclopamine inhibits growth of SCLC and a wide range of foregut derived malignancies both in vitro and in vivo. This demonstrates an ongoing requirement for Hh signalling in these highly lethal and aggressive tumors. A novel therapeutic strategy is proposed using pharmacologic targeting of Hh dependent tumors with high potency pathway antagonists.
Collapse
Affiliation(s)
- D N Watkins
- School of Medicine, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA.
| | | |
Collapse
|
193
|
Wang J, Laurie GW. Organogenesis of the exocrine gland. Dev Biol 2004; 273:1-22. [PMID: 15302594 DOI: 10.1016/j.ydbio.2004.05.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2004] [Revised: 04/06/2004] [Accepted: 05/15/2004] [Indexed: 11/16/2022]
Abstract
Morphogenesis of exocrine glands is a complex stepwise process of epithelial ingrowth, ductal elongation, ductal branching, and alveolar or acinar differentiation. Emerging from an increasing number of mouse gene knockout, dominant-negative, and antisense models is the identification of a remarkable collection of cell adhesion molecules, growth factors, and their receptors whose time-dependent contributions to glandular organogenesis are essential. Many have cryptically overlapping and interdependent but noncompensatory roles. Discoidin domain receptor 1 tyrosine kinase (DDR1) and the ErbB1 receptor of amphiregulin are, for example, required for ductal branching and elongation. Each is in turn dependent on the Wnt family of morphogenic factors for autophosphorylation or transactivation, respectively. Here we review the current cast of exocrine glandular morphogens, as a foundation for a global or systems biology appreciation of the interweaving signaling pathways that underlie mammalian glandular morphogenesis.
Collapse
Affiliation(s)
- Jiahu Wang
- Department of Cell Biology, University of Virginia, Charlottesville 22908-0732, USA
| | | |
Collapse
|
194
|
Abstract
Pancreas regeneration after tissue damage is a key response to pancreatic injury, involving pancreatic duct progenitor cells and intra-islet precursor cells. Surgical removal of the pancreas, duct obstruction by cellophane wrapping and bone marrow-derived stem cell transplantation act as inductive stimuli, leading to pancreas regeneration. The exact role of growth and differentiation factors regulating pancreatic beta-cell mass remains unknown. Here, I will attempt to integrate recent findings and speculate on the factors that trigger this fascinating response, wherein the pancreas responds to a deficit in cell mass and undergoes new islet formation, leading to restoration of normal beta-cell mass. I will also discuss recent advances in regenerating endocrine pancreatic cells, which could affect stem cell-based approaches to treating diabetes mellitus.
Collapse
Affiliation(s)
- Anandwardhan A Hardikar
- National Institute of Diabetes and Digestive and Kidney Diseases, Bldg 50/Room 4128, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
195
|
Kayed H, Kleeff J, Keleg S, Guo J, Ketterer K, Berberat PO, Giese N, Esposito I, Giese T, Büchler MW, Friess H. Indian hedgehog signaling pathway: expression and regulation in pancreatic cancer. Int J Cancer 2004; 110:668-76. [PMID: 15146555 DOI: 10.1002/ijc.20194] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer is an aggressive malignancy that exhibits a number of genetic and epigenetic alterations. Indian hedgehog (Ihh) and its 2 signaling receptors, patched (Ptc) and smoothened (Smo), are involved in pancreatic development and regulation of beta-cell function as well as in certain human tumors. In the current study, we analyzed the expression, distribution and function of Ihh and its receptors in pancreatic cancer. Quantitative RT-PCR and immunohistochemistry were utilized to analyze the expression, localization and transcriptional regulation of Ihh, Ptc and Smo. The effects of inhibition and stimulation of the hedgehog signaling pathway on pancreatic cancer cell growth were examined by the MTT cell growth assay. By quantitative RT-PCR, Ihh, Ptc and Smo mRNA levels were increased 35-, 1.2- and 1.6-fold, respectively, in pancreatic cancer tissues in comparison to normal pancreatic tissues. By immunohistochemistry, Ihh, Ptc and Smo were expressed in the islet cells of normal and cancerous tissues and in pancreatic cancer cells. The growth of pancreatic cancer cells was dose-dependently inhibited by the hedgehog antagonist cyclopamine through G0/G1 arrest. In contrast, Ihh agonists exhibited no significant effect on pancreatic cancer cell growth. TGF-beta1 repressed Ihh transcription in a TGF-beta1-responsive pancreatic cancer cell line, but had no effect on the other tested cell lines. In conclusion, Ihh and its receptors Ptc and Smo are expressed in pancreatic cancer, and blockage of hedgehog signaling results in inhibition of pancreatic cancer cell growth, suggesting that aberrant activation of the Ihh signaling pathway contributes to tumor development in this malignancy.
Collapse
Affiliation(s)
- Hany Kayed
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Josten F, Fuss B, Feix M, Meissner T, Hoch M. Cooperation of JAK/STAT and Notch signaling in the Drosophila foregut. Dev Biol 2004; 267:181-9. [PMID: 14975725 DOI: 10.1016/j.ydbio.2003.11.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2003] [Revised: 11/07/2003] [Accepted: 11/16/2003] [Indexed: 10/26/2022]
Abstract
Temporal and spatial regulation of morphogenesis is pivotal to the formation of organs from simple epithelial tubes. In a genetic screen for novel genes controlling cell movement during posterior foregut development, we have identified and molecularly characterized two alleles of the domeless gene which encodes the Drosophila Janus kinase (JAK)/STAT receptor. We demonstrate that mutants for domeless or any other known component of the canonical JAK/STAT signaling pathway display a failure of coordinated cell movement during the development of the proventriculus, a multiply folded organ which is formed by stereotyped cell rearrangements in the posterior foregut. Whereas the JAK/STAT receptor is expressed in all proventricular precursor cells, expression of upd encoding its ligand and of STAT92E, the signal transducer of the pathway, is locally restricted to cells that invaginate during proventriculus development. We demonstrate by analyzing gene expression mediated by a model Notch response element and by studying the expression of the Notch target gene short stop, which encodes a cytoskeletal crosslinker protein, that JAK/STAT signaling is required for the activation of Notch-dependent gene expression in the foregut. Our results provide strong evidence that JAK/STAT and Notch signaling cooperate in the regulation of target genes that control epithelial morphogenesis in the foregut.
Collapse
Affiliation(s)
- Frank Josten
- Universität Bonn, Institut für Molekulare Physiologie und Entwicklungsbiologie, Abteilung für Molekulare Entwicklungsbiologie, D-53115 Bonn, Germany
| | | | | | | | | |
Collapse
|
197
|
Lin JW, Biankin AV, Horb ME, Ghosh B, Prasad NB, Yee NS, Pack MA, Leach SD. Differential requirement for ptf1a in endocrine and exocrine lineages of developing zebrafish pancreas. Dev Biol 2004; 270:474-486. [PMID: 15183727 DOI: 10.1016/j.ydbio.2004.02.023] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2003] [Revised: 01/20/2004] [Accepted: 02/25/2004] [Indexed: 11/15/2022]
Abstract
Mammalian studies have implicated important roles for the basic helix-loop-helix transcription factor Ptf1a-p48 in the development of both exocrine and endocrine pancreas. We have cloned the Ptf1a-p48 ortholog in Danio rerio. Early zebrafish ptf1a expression is observed in developing hindbrain and in endodermal pancreatic precursors. Analysis of ptf1a and insulin expression reveals a population of exocrine precursors that, throughout early development, are temporally and spatially segregated from endocrine elements. Morpholino-mediated knockdown of ptf1a confirms early divergence of these endocrine and exocrine lineages. Ptf1a morphants lack differentiated exocrine pancreas, but maintain normal differentiation and organization of the principal islet. In addition to the exocrine phenotype, ptf1a knockdown also reduces the prevalence of a small population of anterior endocrine cells normally found outside the principal islet. Together, these findings suggest the presence of distinct ptf1a-dependent and ptf1a-independent precursor populations in developing zebrafish pancreas.
Collapse
Affiliation(s)
- John W Lin
- Department of Surgery, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | |
Collapse
|
198
|
Fagman H, Grände M, Gritli-Linde A, Nilsson M. Genetic deletion of sonic hedgehog causes hemiagenesis and ectopic development of the thyroid in mouse. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:1865-72. [PMID: 15111333 PMCID: PMC1615667 DOI: 10.1016/s0002-9440(10)63745-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Thyroid dysgenesis encountered in 85% of patients with congenital hypothyroidism is a morphologically heterogeneous condition with primarily unknown pathogenesis. Here we identify sonic hedgehog (Shh) as a novel regulator of thyroid development. In Shh knockout mice the thyroid primordium is correctly specified in the pharyngeal endoderm, but budding and dislocation are slightly delayed. In late development the thyroid fails to form a bilobed gland. Instead a single thyroid mass is found unilaterally and mostly to the left of the midline. Thyroid-specific transcription factors (TTF-1 and TTF-2) and thyroglobulin are expressed indicating terminal differentiation. Strikingly, TTF-1- and TTF-2-positive cells aberrantly develop in the presumptive trachea of Shh-/- embryos. The ectopic tissue buds ventrolaterally into the adjacent mesenchyme, and less extensively into the tracheal lumen, forming follicle-like structures that accumulate thyroglobulin. Shh mRNA is not expressed in the thyroid precursor cells at any developmental stage. The results indicate that Shh signaling indirectly governs the symmetric bilobation of the thyroid during late organogenesis. Shh also seems to repress inappropriate thyroid differentiation in nonthyroid embryonic tissues. This study provides clues to the molecular mechanisms that might be dysregulated in thyroid hemiagenesis and development of ectopic thyroid tissue outside the thyroglossal duct.
Collapse
Affiliation(s)
- Henrik Fagman
- Institute of Anatomy and Cell Biology, The Sahlgrenska Academy at Göteborg University, Göteborg, Sweden.
| | | | | | | |
Collapse
|
199
|
Savastano S, Alessi S, Fantozzi O, Corrà S, Zonta L. MRI diagnosis of a periportal annular pancreas. ACTA ACUST UNITED AC 2004. [DOI: 10.1016/j.ejrex.2004.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
200
|
Zhu L, Tran T, Rukstalis JM, Sun P, Damsz B, Konieczny SF. Inhibition of Mist1 homodimer formation induces pancreatic acinar-to-ductal metaplasia. Mol Cell Biol 2004; 24:2673-81. [PMID: 15024058 PMCID: PMC371125 DOI: 10.1128/mcb.24.7.2673-2681.2004] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2003] [Revised: 12/19/2003] [Accepted: 01/06/2004] [Indexed: 12/27/2022] Open
Abstract
The pancreas consists of three main cell lineages (endocrine, exocrine, and duct) that develop from common primitive foregut precursors. The transcriptional network responsible for endocrine cell development has been studied extensively, but much less is known about the transcription factors that maintain the exocrine and duct cell lineages. One transcription factor that may be important to exocrine cell function is Mist1, a basic helix-loop-helix (bHLH) factor that is expressed in acinar cells. In order to perform a molecular characterization of this protein, we employed coimmunoprecipitation and bimolecular fluorescence complementation assays, coupled with electrophoretic mobility shift assay studies, to show that Mist1 exists in vivo as a homodimer complex. Analysis of transgenic mice expressing a dominant-negative Mist1 transgene (Mist1(mutant basic) [Mist1(MB)]) revealed the cell autonomous effect of inhibiting endogenous Mist1. Mist1(MB) cells become disorganized, exhibit a severe depletion of intercellular gap junctions, and express high levels of the glycoprotein clusterin, which has been shown to demarcate immature acinar cells. Inhibition of Mist1 transcriptional activity also leads to activation of duct-specific genes, such as cytokeratin 19 and cytokeratin 20, suggesting that alterations in the bHLH network produce a direct acinar-to-ductal phenotypic switch in mature cells. We propose that Mist1 is a key transcriptional regulator of exocrine pancreatic cells and that in the absence of functional Mist1, acinar cells do not maintain their normal identity.
Collapse
Affiliation(s)
- Liqin Zhu
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907-1392, USA
| | | | | | | | | | | |
Collapse
|