scholarly journals Evidence for the presence of stem cell-like progenitor cells in human adult pancreas

2007 ◽  
Vol 195 (3) ◽  
pp. 407-414 ◽  
Author(s):  
Min Zhao ◽  
Stephanie A Amiel ◽  
Michael R Christie ◽  
Paolo Muiesan ◽  
Parthi Srinivasan ◽  
...  

The origin of cells replacing ageing β-cells in adult life is unknown. This study assessed the expression of classic stem cell markers: Oct4, Sox2 and CD34 in islet-enriched fractions versus exocrine cell-enriched fractions from 25 adult human pancreases following human islet isolation. Expression of Oct4, Sox2 and CD34 mRNAs was found in all cell samples, with no significant differences between endocrine and exocrine cell fractions. Immunohistochemical staining for Oct4, Sox2, CD133, CD34, CK19, insulin and nestin on human pancreas sections showed that the majority of Oct4+ve cells were found in the walls of small ducts. Similar localisations were observed for Sox2+ve cells. The majority of Sox2+ve cells were found to co-express Oct4 proteins, but not vice versa. Cells positive for Oct4 and Sox2 appeared to be a unique cell population in the adult human pancreases without co-expression for CK19, CD34, CD133, insulin and nestin proteins. The numbers of Oct4+ve and Sox2+ve cells varied among donors and were ∼1–200 and 1–30 per 100 000 pancreatic cells respectively.

2011 ◽  
Vol 211 (2) ◽  
pp. 169-176 ◽  
Author(s):  
Michael G White ◽  
Hussain R Al-Turaifi ◽  
Graham N Holliman ◽  
Ali Aldibbiat ◽  
Aiman Mahmoud ◽  
...  

The source of new β-cells in adult human pancreas remains incompletely elucidated with recent studies on rodents providing evidence for neogenesis from progenitor cells in addition to self-replication. The aim of this study was to investigate the expression of pluripotency-associated stem cell markers in proliferative cultures derived from adult human pancreas. Human pancreatic tissue was obtained from deceased donors following ethical approval and relative consent. Islet-enriched fraction was separated from the retrieved organ by digestion and density gradient centrifugation. Dissociated cells were seeded in adherent culture forming proliferative ‘islet survivor cells’ (ISCs). These were characterised at fifth passage by RT-PCR, immunofluorescence staining, FACS, western blot and transfection studies with an OCT4 promoter-driven reporter. Nuclear expression of the pluripotency-associated stem cell marker complex OCT4/SOX2/NANOG was confirmed in ISCs. The phenotype constituted ∼8% of the overall population. OCT4 biosynthesis was confirmed by western blot and activation of an exogenous OCT4 promoter. Co-expression of pluripotency-associated markers has been confirmed in proliferative primary cells derived from adult human pancreas. Further studies are required to elucidate whether these cells possess functional stem cell characteristics and assess potential for differentiation into pancreatic cell lineages including new β-cells.


2020 ◽  
Author(s):  
Sandrina Martens ◽  
Mathias Van Bulck ◽  
Katarina Coolens ◽  
Hediel Madhloum ◽  
Farzad Esni ◽  
...  

SUMMARYObjectiveAn aggressive basal-like molecular subtype of pancreatic ductal adenocarcinoma (PDAC) exists, driven by ΔNp63. In other epithelia, ΔNp63+ basal cells have stem cell capacity and can be at the origin of tumors. In the pancreas, basal cells have not been identified.DesignWe assessed basal cell markers in human and mouse pancreas, chronic pancreatitis and PDAC, and developed a 3D imaging protocol (FLIP-IT) to study sizeable samples at single cell resolution. We generated organoid cultures of ducts from Sox9-eGFP reporter mice.ResultsIn normal human pancreas, rare ΔNp63+ cells exist in ducts that expand in chronic pancreatitis. ΔNp63+ cells express KRT19 and canonical basal markers (KRT5, KRT14 and S100A2) but lack markers of duct cells such as CA19.9 and SOX9. In addition, ΔNp63+ cells pertain to a niche of cells expressing gastrointestinal stem cell markers. 3D views of the ductal tree in formalin fixed paraffin embedded samples show that basal cells are localized on the basal membrane of medium to large ducts and expand as multilayer dome-like structures in chronic pancreatitis. In mice, ΔNp63 expression is induced when culturing organoids from Sox9-low ductal cells but could not be found in normal pancreas nor in models of pancreatitis or pancreatic cancer.ConclusionWe discovered a novel ductal cell population in normal human pancreas similar to basal cells in other tissues. Using FLIP-IT, we provide unprecedented 3D visualization of these cells in archival clinical specimens. ΔNp63+ cells may play an important role in pancreatic tissue regeneration and cancer.SUMMARY BOXWhat is already known about this subject?ΔNp63 has a central role in determining the basal-like subtype of pancreatic ductal adenocarcinoma (PDAC).Different to other tissues with basal cancers, the normal pancreas reportedly does not contain (ΔNp63-expressing) basal cells.Current protocols face severe limitations for marker-based identification and 3D imaging of individual (rare) cells in archival pancreatic samples.What are the new findings?We report a rare and atypical pancreatic duct cell that expresses ΔNp63, other basal cell markers and g.i. stem cell markers.The number of these basal cells increases in diseases such as chronic pancreatitis and pancreatic cancer.We provide an easy to implement protocol for 3D clearing and high-resolution imaging of sizeable samples of (fresh or FFPE) human pancreas or an entire mouse pancreas.Except after culturing medium to large ducts as organoids, we fail to detect basal cells in mouse experimental pancreatic models.How might it impact on clinical practice in the foreseeable future?Extrapolating from knowledge in other organs, basal cells in the pancreas may have a stem cell/progenitor role, including in diseases such as (basal) pancreatic cancer.Use of the 3D imaging protocol in archival clinical specimens will allow unprecedented insights in pancreatic histopathology.For above mentioned diseases, we caution for findings in experimental mouse models that may not (fully) recapitulate the etiopathogenesis.


Pancreas ◽  
2008 ◽  
Vol 36 (1) ◽  
pp. e1-e6 ◽  
Author(s):  
Jessy Lardon ◽  
Denis Corbeil ◽  
Wieland B. Huttner ◽  
Zhidong Ling ◽  
Luc Bouwens

2011 ◽  
Vol 54 (3) ◽  
pp. 501 ◽  
Author(s):  
SarasaBharati Arumugam ◽  
OmanaA Trentz ◽  
Devi Arikketh ◽  
Vijayalakshmi Senthinathan ◽  
Barry Rosario ◽  
...  

Reproduction ◽  
2014 ◽  
Vol 148 (4) ◽  
pp. 417-427 ◽  
Author(s):  
Eran Altman ◽  
Pamela Yango ◽  
Radwa Moustafa ◽  
James F Smith ◽  
Peter C Klatsky ◽  
...  

Autologous spermatogonial stem cell (SSC) transplantation is a potential therapeutic modality for patients with azoospermia following cancer treatment. For this promise to be realized, definitive membrane markers of prepubertal and adult human SSCs must be characterized in order to permit SSC isolation and subsequent expansion. This study further characterizes the markers of male gonocytes, prespermatogonia, and SSCs in humans. Human fetal, prepubertal, and adult testicular tissues were analyzed by confocal microscopy, fluorescence-activated cell sorting, and qRT-PCR for the expression of unique germ cell membrane markers. During male fetal development, THY1 and KIT (C-Kit) are transient markers of gonocytes but not in prespermatogonia and post-natal SSCs. Although KIT expression is detected in gonocytes, THY1 expression is also detected in the somatic component of the fetal testes in addition to gonocytes. In the third trimester of gestation, THY1 expression shifts exclusively to the somatic cells of the testes where it continues to be detected only in the somatic cells postnatally. In contrast, SSEA4 expression was only detected in the gonocytes, prespermatogonia, SSCs, and Sertoli cells of the fetal and prepubertal testes. After puberty, SSEA4 expression can only be detected in primitive spermatogonia. Thus, although THY1 and KIT are transient markers of gonocytes, SSEA4 is the only common membrane marker of gonocytes, prespermatogonia, and SSCs from fetal through adult human development. This finding is essential for the isolation of prepubertal and adult SSCs, which may someday permit fertility preservation and reversal of azoospermia following cancer treatment.


2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Fei Chen ◽  
Tuo Li ◽  
Yu Sun ◽  
Qinggui Liu ◽  
Tao Yang ◽  
...  

Abstract Background Stem cell-derived pancreatic β-like cells hold great promise for treating diabetes. Gallbladder belongs to the extrahepatic bile duct system and possesses stem-like cells. These stem cells could be expanded in vitro and have the potential of differentiating into hepatocytes, cholangiocytes, or pancreatic cells. As the gallbladder is highly available, gallbladder stem cells provide a new cell source of pancreatic β-like cells. In this study, we aimed to investigate an approach for the generation of pancreatic β-like cells from gallbladder stem cells (GSCs) without genetic modification. Methods A CK19CreERT;Rosa26R-GFP mouse was used to isolate CK19+ cells, which represented EpCAM+ stem cells in the gallbladder. They were cultured in the modified Kubota’s medium for expansion and further analyzed. Then, we developed a strategy to screen a combination of small molecules that can generate insulin-secreting cells from gallbladder stem cells. These cells were identified with markers of pancreatic cells. Finally, they were seeded into the cellulosic sponge and transplanted to the diabetic mice for functional examination in vivo. Results Gallbladder stem cells could be expanded for more than 15 passages. They expressed typical hepatic stem cell markers including CK19, EpCAM, Sox9, and albumin. By screening method, we found that adding Noggin, FR180204, and cyclopamine could efficiently induce gallbladder stem cells differentiating into insulin-secreting cells. These cells expressed Pdx1, Nkx6.1, and insulin but were negative for Gcg. After transplantation with the cellulosic sponge, they could ameliorate hyperglycemia in the diabetic mice. Conclusion This study provides a new approach which can generate insulin-secreting cells from the gallbladder without genetic modification. This offers an option for β cell therapy in treating type 1 diabetes.


2021 ◽  
Vol 12 ◽  
Author(s):  
Amanda Oakie ◽  
Maria Cristina Nostro

Restoring the number of glucose-responsive β-cells in patients living with diabetes is critical for achieving normoglycemia since functional β-cells are lost during the progression of both type 1 and 2 diabetes. Stem cell-derived β-cell replacement therapies offer an unprecedented opportunity to replace the lost β-cell mass, yet differentiation efficiencies and the final yield of insulin-expressing β-like cells are low when using established protocols. Driving cellular proliferation at targeted points during stem cell-derived pancreatic progenitor to β-like cell differentiation can serve as unique means to expand the final cell therapeutic product needed to restore insulin levels. Numerous studies have examined the effects of β-cell replication upon functionality, using primary islets in vitro and mouse models in vivo, yet studies that focus on proliferation in stem cell-derived pancreatic models are only just emerging in the field. This mini review will discuss the current literature on cell proliferation in pancreatic cells, with a focus on the proliferative state of stem cell-derived pancreatic progenitors and β-like cells during their differentiation and maturation. The benefits of inducing proliferation to increase the final number of β-like cells will be compared against limitations associated with driving replication, such as the blunted capacity of proliferating β-like cells to maintain optimal β-cell function. Potential strategies that may bypass the challenges induced by the up-regulation of cell cycle-associated factors during β-cell differentiation will be proposed.


2011 ◽  
Author(s):  
Moon Nian Lim ◽  
Umapathy Thiageswari ◽  
Othman Ainoon ◽  
P. J. N. Baharuddin ◽  
R. A. Jamal ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document