Ponad 7000 publikacji medycznych!
Statystyki za 2021 rok:
odsłony: 8 805 378
Artykuły w Czytelni Medycznej o SARS-CoV-2/Covid-19

Poniżej zamieściliśmy fragment artykułu. Informacja nt. dostępu do pełnej treści artykułu
© Borgis - Postępy Nauk Medycznych 3/2011, s. 209-216
*Artur Pałasz, Marcin Kamiński
Rozwój trzustki i rola komórek macierzystych dojrzałego narządu w jego regeneracji
Pancreas development and the role of adult stem cells in regeneration
Department of Histology, Medical University of Silesia, Katowice, Poland
Head of Department: dr hab. med. Ryszard Wiaderkiewicz, prof. ndzw. SUM
Streszczenie
Morfogeneza trzustki jest procesem precyzyjnie regulowanym i koordynowanym zarówno przez indukcyjne szlaki sygnałowe (Hedgehog, Notch, Wnt), jak i przez liczne czynniki transkrypcyjne. Niektóre spośród nich, jak Pdx1, p48 i Mist1 są odpowiedzialne za różnicowanie trzustkowych komórek progenitorowych do fenotypu egzokrynowego, inne natomiast, takie jak Hex, Ngn3 i Pax4 promują wytwarzanie komórek endokrynowych. Dojrzała trzustka dysponuje względnie niewielką liczbą komórek macierzystych, które zdają się jednak odgrywać istotną rolę w regeneracji narządu w następstwie pankreatektomii lub jego patologicznych uszkodzeń. Owe niezróżnicowane komórki są obecne w strukturze wstawek, pęcherzyków wydzielniczych oraz wysp Langerhansa, ich nisze są generalnie zlokalizowane blisko błon podstawnych i naczyń krwionośnych. Pluripotencjalne trzustkowe komórki Oct-4 i Sox2-pozytywne charakteryzują się ekspresją tych samych antygenów powierzchniowych co hemopoetyczne komórki macierzyste i są zdolne do różnicowania do komórek mezodermalnych i endodermalnych. Populacja nestyno-pozytywnych komórek multipotencjalnych wywodzących się prawdopodobnie z przewodów wyprowadzających trzustki bierze również udział w formowaniu zarówno wewnątrz- jak i zewnątrzwydzielniczej części narządu.
Summary
Pancreatic morphogenesis is precisely regulated and coordinated by inductive signalling pathways; Hedgehog, Notch, Wnt as well as numerous transcription factors. Some of them e.g. Pdx1, p48 and Mist1 are responsible for progenitor cells differentiation into exocrine phenotype, others such as Hex, Ngn3 and Pax4 promote the formation of endocrine cells. Adult organ contains relatively small amount of stem cells which seem to play a significant role in regeneration following the pancreatectomy or pathological injury. These undifferentiated cells are present in the structure of intercalated ducts, exocrine acini and endocrine islets of Langerhans, their niches are generally located near a basement membranes and blood vessels. Pluripotential pancreatic Oct4- and Sox2-positive cells show the expression of the same surface antigens as hemopoietic stem cells and they are able to differentiate into both mesodermal and endodermal cells. A populations of multipotential nestin-positive cells probably derived from excretory ducts are also able to undergo formation of both endocrine and exocrine pancreatic components.



Introduction
The liver and the pancreas are organs that develop from two different groups of endodermal epithelium of the primitive foregut. The liver is formed from the segment of the hepatic recess, and proliferating cells penetrate the mesoderm of the transverse septum. A hepatic bud comprises the head part, which develops into the liver, and the caudal part, responsible for creating the gallbladder and bile ducts. Furthermore, the pancreas develops from two buds, dorsal and ventral, which are bulges of the proximal part of the duodenum. The ventral bud develops into the uncinate process and the inferior part of the pancreatic head, whereas the rest of this organ derives from the dorsal bud endoderm. In humans, the dorsal bud of the pancreas is visible in the fifth week of the development, penetrating into the dorsal mesentery of the stomach. The ventral bud appears slightly later and it is located in the area of the common bile duct. During the turn of the intestines, also of the duodenum, the ventral bud moves in the dorsal direction to finally merge with the dorsal bud and form the organ. In humans, pancreatic islets appear in the third month of the foetal development, whereas insulin secretion begins about the fifth month. The pancreatic and hepatic buds differentiate due to regulatory signals from the surrounding mesodermal cells.
The formation of embryonic stem cell (ES) culture requires their isolation from a blastocyst before implantation. A growth medium for culturing such types of cells should contain, beside standard foetal calf serum, nourishing cells (fibroblasts) and leukemia inhibitory factor (LIF). Under such conditions, ES cells form colonies composed of both undifferentiated and spontaneously differentiating cells. Self-renewable ES require binding LIF to a binary system of receptors, including a receptor specific for LIF and the gp130 receptor. The receptor activation initiates the JAK/STAT3 pathway, that is, one of pathways promoting cell proliferation. The activation of ERK by gp130 and other membranous receptors suppresses the effect of the LIF-STAT3 pathway. Thus, the maintenance of ES ability to divide results from the balance between the actions of different signalling pathways. The expression of the transcription factor Oct-3/4, a member of the POU family identified in embryonic neoplastic cells, and Nanog that plays a key role in blastocyst formation, is necessary for ES to maintain the ability to differentiate. The knowledge of the final formation of B cells from pluripotential precursors is hindered by the fact that they appear at relatively late stages of organogenesis. This brings up a question what molecular events regulate a sequential activation and inhibition of the adequate number of homeotic genes controlling the formation of endodermal cell line, and then, the generation of precursor exocrine cells, endocrine cells and finally, mature B cells (1).
Molecular mechanisms of pancreatic development
The suppression of mesodermal signalling pathways, Wnt and FGF4, which is predominant in the early phase of the foregut development, enables the induction of morphogenesis of both the liver and the pancreas, whereas the activation of the Wnt pathway in the hindgut prevents the initiation of this process (2). FGF released from the cardiac mesoderm of the ventral part of the foregut and BMP from mesenchymal cells of the transverse septum in a coordinated way, induce the molecular liver formation programme and at the same time block the pancreatic development programme (3, 4). Interestingly enough, the activation of the MAPK pathway as a reaction to FGF occurs earlier in cells located laterally inside the bud than in medially situated cells (5). At the time of foregut closing, ventrolateral endodermal cells, which move in the caudal direction in relation to the cardiac domain, cease to be affected by FGF and initiate the development of the ventral pancreatic bud (6) he dorsal part of the foregut, signals from the dorsal chord, activin and FGF, inhibit the Shh (Sonic hedgehog) pathway in endodermal cells, which activates the pancreatic development programme. It is worth emphasizing that all of the phenomena previously described occur in vertebrate embryos within a few hours only (7, 8).
Bipotential hepatic stem cells called hepatoblasts show the expression of genes characteristic for hepatocytes. They code serum proteins: albumin (alb1) and transthyretin (ttr). They undergo a differentiation into both hepatocytes and cholangiocytes (8). Tbx3 gene promotes the expansion of the hepatoblast population via p19ARFsuppression. At early stages, the pancreatic endoderm is characterised by the expression of genes for transcription factors, Pdx1 and Ptf1a (9). They play a key role in the pancreatic development (10, 11). Following the formation of hepatoblasts and pancreatic progenitor cells, they change their shape from cuboidal into columnar, and then they form a pseudostratified structure. This process is similar to the morphogenesis of the nervous epithelium, and in the foregut it is controlled by the transcription factor Hhex (6). The pancreatic epithelium spreads and penetrates into the stroma, where it forms the pancreatic bud, whereas, in the case of the hepatic epithelium, the basement membrane is disrupted and cells proliferate into the immediate surroundings of the stroma. Later morphological changes are controlled by the genes for transcription factors, Prox1, Hnf6/OC-1, Hnf6 and OC-1 regulating the synthesis of E-cadherin, thrombospondin-4, and Spp1, which participate in the process of the cell adhesion and migration (8). Neighbouring progenitor cells of both organs also receive regulatory signals from surrounding endothelial cells (12, 13). Interestingly enough, endothelial cells may also promote liver regeneration after its parenchymal damage, mainly via the HGF signalling pathway (14). Signalling factors produced by endothelial cells have not been identified yet. However, it has been established that sphingosine phosphate (present in blood) that is a substance of endothelial origin, promotes the development of the dorsal pancreatic bud (15). The cells of neural crests migrating into the developing pancreas are transformed into neurons and thus influence the number of B cells (16). The stimulating role of endothelial cells and neural crest cells illustrates the importance of cooperation of the mesenchymal stroma in the development of progenitor hepatic and pancreatic cells. In the liver and in pancreatic buds, the components of the Notch signalling pathway ensure the balance between hepatocytes and cholangiocytes formed from hepatoblasts as well as endo- and exocrine cells derived from pancreatic progenitor cells. The blockade of the Notch system initiates a preferential development of endocrine cells. The main role in this process is played by the Ngn3 transcription factor which is essential for the development of pancreatic islets and which has the features of a classic marker of pancreatic precursor endocrine cells. Ngn3 expression is considerably reduced at the moment of birth, and in the mature organ it is almost indeterminable. In the nascent endocrine cells, the expression of Notch ligands (delta, serrate, jagged) is considerably increased. These ligands react with the Notch receptor on the neighbouring cells and prevent them from differentiating in a similar direction. Multipotential Cpa-1-positive cells located in distal endings of the branched epithelium give rise to both pancreatic ducts and endocrine cells located along their branches. In mice, this process takes place about the 14th day of the embryonic development. At further stages, cells showing Cpa-1 expression are also differentiated into acinar cells of the exocrine part of the pancreas. This process occurs at the time when mature B cells are formed, which is mediated by the MafaA factor.

Powyżej zamieściliśmy fragment artykułu, do którego możesz uzyskać pełny dostęp.
Mam kod dostępu
  • Aby uzyskać płatny dostęp do pełnej treści powyższego artykułu albo wszystkich artykułów (w zależności od wybranej opcji), należy wprowadzić kod.
  • Wprowadzając kod, akceptują Państwo treść Regulaminu oraz potwierdzają zapoznanie się z nim.
  • Aby kupić kod proszę skorzystać z jednej z poniższych opcji.

Opcja #1

24

Wybieram
  • dostęp do tego artykułu
  • dostęp na 7 dni

uzyskany kod musi być wprowadzony na stronie artykułu, do którego został wykupiony

Opcja #2

59

Wybieram
  • dostęp do tego i pozostałych ponad 7000 artykułów
  • dostęp na 30 dni
  • najpopularniejsza opcja

Opcja #3

119

Wybieram
  • dostęp do tego i pozostałych ponad 7000 artykułów
  • dostęp na 90 dni
  • oszczędzasz 28 zł
Piśmiennictwo
1. Soria B, Bedoya FJ, Martin F: Gastrointestinal stem cells. I. Pancreatic stem cells. Am J Physiol Gastrointest Liver Physiol 2005 Aug; 289 (2): G177-G180.
2. McLin VA, Rankin SA, Zorn AM: Repression of Wnt/beta-catenin signaling in the anterior endoderm is essential for liver and pancreas development. Development 2007 Jun; 134 (12): 2207-17.
3. Deutsch G, Jung J, Zheng M et al.: A bipotential precursor population for pancreas and liver within the embryonic endoderm. Development 2001 Mar; 128 (6): 871-81.
4. Shin D, Shin CH, Tucker J et al.: Bmp and Fgf signaling are essential for liver specification in zebrafish. Development 2007 Jun; 134 (11): 2041-50.
5. Calmont A, Wandzioch E, Tremblay KD et al.: An FGF response pathway that mediates hepatic gene induction in embryonic endoderm cells. Dev Cell 2006 Sep; 11 (3): 339-48.
6. Bort R, Signore M, Tremblay K et al.: Hex homeobox gene controls the transition of the endoderm to a pseudostratified, cell emergent epithelium for liver bud development. Dev Biol 2006 Feb 1; 290 (1): 44-56.
7. Hebrok M, Kim SK, Melton DA: Notochord repression of endodermal Sonic hedgehog permits pancreas development. Genes Dev 1998 Jun 1; 12 (11): 1705-13.
8. Zaret KS: Genetic programming of liver and pancreas progenitors: lessons for stem-cell differentiation. Nat Rev Genet 2008 May; 9 (5): 329-40.
9. Gu G, Dubauskaite J, Melton DA: Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors. Development. 2002 May; 129 (10): 2447-57.
10. Kawaguchi Y, Cooper B, Gannon M et al.: The role of the transcriptional regulator Ptf1a in converting intestinal to pancreatic progenitors. Nat Genet 2002 Sep; 32 (1): 128-34.
11. Lin JW, Biankin AV, Horb ME et al.: Differential requirement for ptf1a in endocrine and exocrine lineages of developing pancreas. Dev Biol 2004 Jun; 270 (2): 474-86.
12. Matsumoto K, Yoshitomi H, Rossant J, Zaret KS: Liver organogenesis promoted by endothelial cells prior to vascular function. Science 2001 Oct; 294 (5542): 559-63.
13. Yoshitomi H, Zaret KS: Endothelial cell interactions initiate dorsal pancreas development by selectively inducing the transcription factor Ptf1a. Development 2004 Feb; 131 (4): 807-17.
14. Le Couter J, Moritz DR, Li B et al.: Angiogenesis-independent endothelial protection of liver: role of VEGFR-1. Science 2003 Feb; 299 (5608): 890-93.
15. Edsbagge J, Johansson JK, Esni F et al.: Vascular function and sphingosine-1-phosphate regulate development of the dorsal pancreatic mesenchyme. Development 2005 Mar; 132 (5): 1085-92.
16. Nekrep N, Wang J, Miyatsuka T, German MS: Signals from the neural crest regulate beta-cell mass in the pancreas. Development 2008 Jun; 135 (12): 2151-60.
17. Oliver-Krasinski JM, Stoffers DA: On the origin of the B cell. Genes Dev 2008 Aug; 22 (5): 1998-2021.
18. Sasaki H, Hogan BL: HNF-3β as a regulator of floor plate development. Cell 1994 Jan; 76 (1): 103-15.
19. Ang SL, Rossant J: HNF-3β is essential for node and notochord formation in mouse development. Cell 1994 Aug; 78 (4): 561-74.
20. Weinstein DC, Ruiz y Altaba A, Chen WS et al.: The winged-helix transcription factor HNF-3β is required for notochord development in the mouse embryo. Cell 1994 Aug; 78 (4): 575-88.
21. Fendrich V, Esni F, Garay MVR et al.: Hedgehog signaling is required for effective regeneration of exocrine pancreas. Gastroenterology 2008 Aug; 135 (2): 621-31.
22. Liu Y, MacDonald RJ, Swift GH: DNA binding transcriptional activation by a PDX1.PBX1b. MEIS2b trimer and cooperation with a pancreas-specific basic helix-loop-helix complex. J Biol Chem 2001 May; 276 (21): 17985-93.
23. Pin CL, Rukstalis JM, Johnson C, Konieczny SF: The bHLH transcription factor Mist1 is required to maintain exocrine pancreas cell organization and acinar cell identity. J Cell Biol 2001 Nov; 155 (4): 519-30.
24. Johnson CL, Kowalik AS, Rajakumar N, Pin CL: Mist1 is necessary for the establishment of granule organization in serous exocrine cells of the gastrointestinal tract. Mech Dev 2004 Mar; 1219 (3): 261-72.
25. Rovira M, Delaspre F, Massumi M et al.: Murine ES-derived pancreatic acinar cells recapitulate features of early pancreatic differentiation. Gastroenterology 2008 Oct; 135 (4): 1301-10.
26. Doyle MJ, Sussel L: Nkx2.2 regulates β-cell function in the mature islet. Diabetes 2007 Aug; 56 (8): 1999-2007.
27. Sander M, Sussel L, Conners J et al.: Homeobox gene Nkx6.1 lies downstream of NKx2.2 in the major pathway of β-cell formation in the pancreas. Development 2000 Dec; 127 (24): 5533-40.
28. Lioubinski O, Muller M, Wegner M, Sander M: Expression of Sox transcription factors in the developing mouse pancreas. Dev Dyn 2003 Jul; 227 (3): 402-08.
29. Mellitzer G, Bonne S, Luco RF et al.: IA1 is NGN3-dependent and essential for differentiation of the endocrine pancreas. EMBO J 2006 Mar; 25 (6): 1344-52.
30. Bjerknes M, Cheng H: Neurogenin 3 and the enteroendocrine cell lineage in the adult mouse small intestinal epithelium. Dev Biol 2006 Dec; 300 (2): 722-35.
31. Jensen JN, Rosenberg LC, Hencksher-Sorensen J, Serup P: Mutant neurogenin-3 in congenital malabsorptive diarrhea. N Engl J Med. 2007 Apr; 356 (17): 1781-82.
32. Kataoka K, Shioda S, Ando K et al.: Differentially expressed Maf family transcription factors, c-Maf and MafA, activate glucagon and insulin gene expression in pancreatic α- and β-cells. J Mol Endocrinol 2004 Feb; 32 (1): 9-20.
33. Artner I, Blanchi B, Raum JC et al.: MafB is required for islet β cell maturation. Proc Natl Acad Sci 2007 Mar; 104 (10): 3853-58.
34. Bonner-Weir S, Taneja M, Weir GC et al.: In vitro cultivation of human islets from expanded ductal tissue. Proc Natl Acad Sci USA 2000 Jul; 97 (14): 7999-8004.
35. Yatoh S, Dodge R, Akashi T et al.: Differentiation of affinity-purified human pancreatic duct cells to beta-cells. Diabets Res Clin Pract 2007 Jul; 56 (7): 1802-09.
36. Ramiya VK, Maraist M, Arfors KE et al.: Reversal of insulin-dependent diabetes using islets generated in vitro from pancreatic stem cells. Nat Med 2000 Mar; 6 (3): 278-82.
37. Desai BM, Oliver-Krasinski J, De Leon DD et al.: Preexisting pancreatic acinar cells contribute to acinar cell, but not islet β-cell regeneration. J Clin Invest 2007 Apr; 117 (4): 971-77.
38. Means AL, Meszoely IM, Suzuki K et al.: Pancreatic epithelial plasticity mediated by acinar cell transdifferentiation and generation of nestin-positive intermediates. Development 2005 Aug; 132 (16): 3767-76.
39. Zulewski H, Abraham EJ, Gerlach MJ et al.: Multipotential nestin-positive stem cells isolated from adult pancreatic islets differentiate ex vivo into pancreatic endocrine, exocrine and hepatic phenotypes. Diabetes 2001 Mar; 50 (3): 521-33.
40. Verstappen J, Katsaros C, Torensma R, Von den Hoff JW: A functional model of adult stem cells in epithelial tissues. Wound Rep Reg 2009 May-Jun; 17 (3): 296-305.
41. Weismann IL: Stem cells: units of development, units of regeneration and units of evolution. Cell 2000 Jan; 100 (1): 157-68.
42. Pittenger MF, Mackay AM, Beck SC et al.: Multilineage potential of adult human mesenchymal stem cells. Science 1999 Apr; 284 (5411): 143-47.
43. Lee OK, Kuo TK, Chen WM et al.: Isolation of multipotent mesenchymal stem cells from umbilical cord blood. Blood 2004 Mar; 103 (5): 1669-75.
44. Seeberger KL, Dufour JM, James Shapiro AM et al.: Expansion of mesenchymal stem cells from human pancreatic ductal epithelium. Lab Invest 2006 Feb; 86 (2): 141-53.
45. Karaoz E, Ayhan S, Gacar G et al.: Isolation and characterization of stem cells from pancreatic islet: pluripotency, differentiation potential and ultrastructural characteristics. Cytotherapy 2010 May; 12 (3): 288-302.
46. Zhao M, Amiel SA, Christie MR et al.: Evidence for the presence of stem cell-like progenitor cells in human adult pancreas. J Endocrinol 2007 Dec; 195 (3): 407-14.
47. Wang RN, Kloppel G, Bouwens L: Duct to islet-cell differentiation and islet growth in the pancreas of duct-ligated adult rats. Diabetologia 1995 Dec; 38 (12): 1405-11.
48. Hayashi KY, Tamaki H, Handa K et al.: Differentiation and proliferation of endocrine cells in the regenerating rat pancreas after 90% pancreatectomy. Arch Histol Cytol 2003 May; 66 (2): 163-74.
49. Ellis P, Fagan BM, Magness ST et al.: SOX2, a persistent marker of multipotential neural stem cells derived from embryonic stem cells, the embryo or the adult. Dev Neurosci 2004 Mar-Aug; 26 (2-4): 148-65.
50. Hong Y, Winkler C, Liu T et al.: Activation of the mouse Oct4 promoter in medaka embryonic stem cells and its use for ablation of spontaneous differentiation. Mech Dev 2004 Jul; 121 (7-8): 933-443.
51. Kuroda T, Tada M, Kubota H et al.: Octamer and Sox elements are required for transcriptional cis regulation of Nanog gene expression. Mol Cell Biol 2005 Mar; 25 (6): 2475-85.
52. Hough SR, Clements I, Welch PJ, Wiederholt KA: Differentiation of mouse embryonic stem cells after RNA interference-mediated silencing of OCT4 and Nanog. Stem Cells 2006 Jun; 24 (6): 1467-75.
53. Bortvin A, Eggan K, Skaletsky H et al.: Incomplete reactivation of Oct4-related genes in mouse embryos cloned from somatic nuclei. Development 2003 Apr; 130 (8): 1673-1680.
54. Avillion AA, Nicolis SK, Pevny LH et al.: Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev 2003 Jan; 17 (1): 126-40.
55. Cauffman G, Van de Velde H, Liebaers I, Van Steirteghem A: Oct-4 mRNA and protein expression during human preimplantation development. Mol Hum Reprod 2005 Mar; 11 (3): 173-81.
56. Elrick LJ, Docherty K: Phosphorylation-dependent nucleocytoplasmic shuttling of pancreatic duodenal homeobox-1. Diabetes 2001 Oct; 50 (10): 2244-52.
57. Lechner A, Leech CA, Abraham EJ et al.: Nestin-positive progenitor cells derived from adult human pancreatic islets of Langerhans contain side population (SP) cells defined by expression of the ABCG2(BCRP1) ATP-binding cassette transporter. Biochem Biophys Res Comun 2002 May; 293 (2): 670-74.
58. Ferber S, Halkin A, Cohen H et al.: Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia. Nature Med 2000 May; 6 (5): 568-72.
otrzymano: 2011-01-10
zaakceptowano do druku: 2011-02-22

Adres do korespondencji:
*Artur Pałasz
Department of Histology, Medical University of Silesia
ul. Medyków 18, 40-752 Katowice
tel.: (32) 208-83-77
e-mail: apalasz@sum.edu.pl

Postępy Nauk Medycznych 3/2011
Strona internetowa czasopisma Postępy Nauk Medycznych