Neurogenins

Last updated
neurogenin 1
Identifiers
Symbol NEUROG1
Alt. symbolsNEUROD3
NCBI gene 4762
HGNC 7764
OMIM 601726
RefSeq NM_006161
UniProt Q92886
Other data
Locus Chr. 5 q23-q31
Search for
Structures Swiss-model
Domains InterPro
neurogenin 2
Identifiers
Symbol NEUROG2
NCBI gene 63973
HGNC 13805
OMIM 606624
RefSeq NM_024019
UniProt Q9H2A3
Other data
Locus Chr. 4 q25
Search for
Structures Swiss-model
Domains InterPro
neurogenin 3
Identifiers
Symbol NEUROG3
NCBI gene 50674
HGNC 13806
OMIM 604882
RefSeq NM_020999
UniProt Q9Y4Z2
Other data
Locus Chr. 10 q21.3
Search for
Structures Swiss-model
Domains InterPro

Neurogenins, often abbreviated as Ngn, are a family of bHLH transcription factors involved in specifying neuronal differentiation. The family consisting of Neurogenin-1, Neurogenin-2, and Neurogenin-3, plays a fundamental role in specifying neural precursor cells and regulating the differentiation of neurons during embryonic development. It is one of many gene families related to the atonal gene in Drosophila. Other positive regulators of neuronal differentiation also expressed during early neural development include NeuroD and ASCL1. [1]

Contents

Function

Neurogenins primarily govern the transition of neural progenitor cells to neurons by activating specific downstream genes associated with neuronal differentiation. Their involvement spans various stages of neurogenesis, including the determination of neural progenitor identity, cell cycle exit, and the acquisition of neuronal characteristics. Notably, Neurogenins influence the specification of different neuronal subtypes, contributing to the diverse array of neurons within the central and peripheral nervous systems. [2]

In neural crest cells, the neurogenin family is essential for neurogenesis in the developing dorsal root ganglia and development of the sensory lineage. [3] [4]

Regulation of neurogenic cascades

The activity of Neurogenins is intricately regulated by molecular pathways and environmental cues. Interaction with other transcription factors, such as proneural factors and Notch signaling, further refines the neurogenic cascades. Their spatiotemporal expression patterns and cross-regulation contribute to the exquisite precision required for proper neural development. [5] [6]

Clinical implications

Neurogenins holds significant implications for clinical research, particularly in the context of neurodevelopmental disorders and neurological regeneration. [7] Aberrations in the expression or regulation of Neurogenins have been linked to conditions such as autism spectrum disorders and neurodegenerative diseases. [8] Ongoing research continues to explore the therapeutic potential of manipulating Neurogenin activity for neural repair and regeneration.

Neurogenin-1

Neurogenin 1 (Ngn1) is a Class-A basic-helix-loop-helix (bHLH) transcription factor that acts as a regulator for neuronal differentiation, and acts by binding to enhancer regulatory elements on genes that encode transcriptional regulators of neurogenesis. In order for Ngn1 to bind with high fidelity with genomic DNA, it must dimerize with another bHLH protein. [9] Ngn1 is a proneural gene because its expression is seen prior to neural lineage determination, indicating it plays a role in neuronal differentiation. [1]

Neuronal differentiation

In E14 rats, when Ngn1 is present in the cerebral cortex, it binds to the CBP/p300/Smad1 transcriptional co-activator complex, which recruits it to the enhancer box upstream of the gene in the promoter for neuronal genes. Binding of Ngn1, to the enhancer box, induces the transcription factor NeuroD to bind to its own enhancer boxes, inducing the genes involved in neuronal differentiation. [10]

Regulation by BMP

Bone-morphogenetic-protein (BMP) signaling is responsible for the expression of the transcriptional co-activators CBP, p300, and Smad1. [10] In the presence of Ngn1, BMPs promote neuronal differentiation in stem cells through binding of all endogenous CBP/p300/Smad1 to Ngn1, and being recruited toward the neuronal promoters, causing neuronal differentiation. [10] In the embryonic forebrain, Ngn1 is associated with dorsal patterning and cell fate specification, with the patterning molecules and proneural proteins establishing the spatial domains of both proneural and homeodomain protein expression. This is critical for the initiation of neurogenesis. [11]

Regulation by LIF

In the presence of Ngn1, the leukemia inhibitory factor (LIF) pathway is inhibited by Ngn1 blocking STAT activation. Normally, the STAT binding site promotes GFAP transcription through binding the STAT1/3 complex, which is activated through the LIF pathway. [10]

Glial differentiation

Along with supporting neuronal differentiation, when expressed in embryonic neural tissue, Ngn1 also acts to inhibit glial differentiation. [12] In the absence of Ngn1, the CBP/p300/Smad1 transcriptional co-activator complex is recruited to and binds to activated STAT1/3, which in turn causes the expression of GFAP, causing glial differentiation. In the presence of Ngn1, inhibition of gliogenesis occurs through Ngn1 binding to the CBP/p300/Smad1 transcriptional co-activator complex, recruiting it away from STAT1/3. [10]

Regulation by BMP

In cases of low levels of Ngn1, BMPs promote glial differentiation. Since Ngn1 is the limiting factor, CBP/p300/Smad1 is able to interact with STAT1/3 and induce gliogenesis. [10]

Regulation by Notch

Activation of the notch pathway, causes the inhibition of proneural bHLH genes, such as Ngn1, which allows for the CBP/p300/Smad1 to interact with STAT1/3 and induce gliogenesis. [10] Along with the embryonic rat, it was also seen in zebrafish that the repression of Ngn1 by Notch, promotes glial lineage in neural crest and central nervous system formation through the inhibition of neuronal differentiation. [1] [13] In addition to the Notch pathway activating the transcriptional factors involved in the promotion of gliogenesis, it is possible that these same factors are involved in the inhibition of other fates.

Regulation by LIF

In the absence of Ngn1, the LIF pathway is able to activate STAT1/3, which allows for the promotion of GFAP transcription via the STAT binding site. The promotion of GFAP transcription induced glial differentiation. [10]

Neurogenin-2

Neurogenin 2 (Ngn2) is a bHLH transcription factor involved in both neurogenesis and neural specification. This protein binds to enhancer-box regulatory elements on the promoters of many genes related to neurogenesis and neural specification. For sufficient DNA binding, Ngn2 must form a dimer with an enhancer protein. [14]

Neurogenesis and glial inhibition

Ngn2 is a transcription factor that both increases expression of proneural genes and drives neural fate by inhibiting expression of glial genes in neural progenitor cells (NPCs). This was observed in mice lacking Ngn2 and mash-1 (another proneural bHLH transcription factor), which have more glia in the cortex and decreased capacity to generate neurons. Olig2 expression in what will become NPCs precedes Ngn2 and promotes its expression. [10] During the switch from neural progenitor fate to glial fate, Ngn2 is downregulated and Nkx2.2, which inhibits proneural genes, is upregulated. [15] Glial fate switch was reduced by inhibiting Nkx2.2 and Olig2 in neural progenitors while allowing the expression of Ngn2. The ability of Olig2 to induce expression of Ngn2 is reduced when Nkx2.2 is expressed. [16]

Neural specification

Mice lacking Ngn2 have fewer motor neurons and ventral interneurons, indicating that Ngn2 plays a role in specification of these neurons. [17]

Pan-neuronal fate

Heterodimerized Ngn2/enhancer protein complex can bind to enhancer boxes to promote transcription of genes related to a non-specified neuronal fate. [17]

V2 interneuron fate

When an enhancer box of a promoter that has been bound by the Ngn-2/enhancer protein complex is also bound by a dimer of the adaptor nuclear LIM interactor (NLI) bound to two LIM homeobox protein 3 (Lhx3), genes related to V2 interneuron identity are expressed. [17]

Motor neuron fate

A dimer of the adaptor NLI bound to two islet 1 (Isl1) proteins and each Isl1 is bound by Lhx3 is called the LIM-homeodomain (LIM-HD) transcription complex. When an enhancer box of a promoter that has been bound by the heterodimerized Ngn2/E-protein complex, the LIM-HD transcription complex is able to bind to drive expression of genes related to motor neuron fate, but only if Ngn2 has been properly phosphorylated. [17]

Ngn2 has two serines, S231 and S234, which can be phosphorylated by glycogen synthase kinase 3 (Gsk3). Phosphorylation of Ngn2 enables interaction with LIM-homeodomain proteins, leading to ventral neural fate and motor neuron specification. [18] The importance of this phosphorylation was determined by using mice that express a mutated form of Ngn2 protein which has the serines from the previously mentioned phosphorylation sites mutated into alanines, which cannot be phosphorylated. These mutant mice have a decreased number of motor neurons and an increased number of V2 interneurons, suggesting that phosphorylation is necessary for driving expression of genes related to motor neuron fate but not V2 interneuron fate and non-specified neural fate. [17]

Neurogenin-3

Neurogenin 3 (Ngn3) is another member of the bHLH family of transcription factors. Ngn3 functions in the differentiation of endocrine pancreas cells. Although its key function is in the pancreas, intestinal cells and neural cells express Ngn3 as well. Several studies have highlighted the importance of Ngn3 for differentiation of endocrine cells. In mice, Ngn3 is present in cells as the pancreas begins to bud and glucagon cells are formed. There are several pathways that Ngn3 works through. [19] [20] [21] [22]

Ngn3 is a crucial component in pancreatic development and plays a supporting role in intestinal as well as neuronal cell development. Studies have demonstrated that knockout of Ngn3 in mice leads to death shortly after birth possibly due to after effects of severe diabetes. [19] Further studies are taking place to investigate Ngn3's possible role as a treatment for diabetes and regeneration of cells in the pancreas. [19] [21]

Neurogenin 3 (NGN3) is expressed by 2-10% of acinar and duct cells in the histologically normal adult human pancreas. NGN3+ cells isolated from cultured exocrine tissue by coexpressed cell surface glycoprotein CD133 have a transcriptome consistent with exocrine dedifferentiation, a phenotype that resembles endocrine progenitor cells during development, and a capacity for endocrine differentiation in vitro. [23] Human [24] and rodent [25] [26] [27] [28] [29] [30] [31] [32] [33] exocrine cells have been reprogrammed into cells with an islet cell-like phenotype following direct expression of NGN3 or manipulation that leads to its expression.

Phases of pancreatic development

The development of the pancreas is broken up into three phases, primary phase, secondary phase, and tertiary phase. Ngn3 is active in the primary and secondary phase. In the primary phase Ngn3 assists in α cell differentiation and in the secondary phase another wave of Ngn3 assists in differentiation of β cells, pancreatic polypeptide cells, and δ cells. Differentiation is marked as complete after the secondary phase. Ngn3 allows for the commitment of pancreatic progenitor cells to become an endocrine multipotent pro-precursor. [19]

Modulation via notch pathway

The Notch pathway is one of the key modulators of Ngn3. The binding of Delta and Serrate, activation ligands for the Notch pathway, activates the Notch surface molecule. This allows the Notch intracellular domain to activate RBK-Jκ to translocate into the nucleus. This complex then activates hairy and enhancer of split (HES)-type proteins, which are inhibitors of Ngn3. The cells that allow the Notch/RBK-Jκ complex to enter are the ones that will not be differentiated into pancreatic cells because Ngn3 is suppressed. It is important to mention that Ngn3 has three HES1 binding sites adjacent to the TATA box sequence that allow for the regulation of this transcription factor. [19]

Downstream targets of Ngn3

NeuroD

Ngn3 can also activate the neurogenic differentiation factor 1(NeuroD1) like most of its other family members through the enhancer boxes present in its structure. Being that NeuroD1 is expressed along with Ngn3 in differentiating cells, it is considered one of the transcription factors downstream targets. [19]

Pax4

Another important target is paired box gene 4 (Pax4), which plays a major role in β cell and δ cell differentiation. Ngn3 works hand-in-hand with HNF1α to activate the Pax4 promoter to induce specific cell differentiation. [19]

Nkx2.2

Another transcription factor that may be a downstream target of Ngn3 is Nkx2.2 because it is often coexpressed with it. Studies have shown that disrupting Nkx2.2 expression results in problems with α and β cell differentiation. [20] [21]

Related Research Articles

In vertebrates, a neuroblast or primitive nerve cell is a postmitotic cell that does not divide further, and which will develop into a neuron after a migration phase. In invertebrates such as Drosophila, neuroblasts are neural progenitor cells which divide asymmetrically to produce a neuroblast, and a daughter cell of varying potency depending on the type of neuroblast. Vertebrate neuroblasts differentiate from radial glial cells and are committed to becoming neurons. Neural stem cells, which only divide symmetrically to produce more neural stem cells, transition gradually into radial glial cells. Radial glial cells, also called radial glial progenitor cells, divide asymmetrically to produce a neuroblast and another radial glial cell that will re-enter the cell cycle.

Neuroepithelial cells, or neuroectodermal cells, form the wall of the closed neural tube in early embryonic development. The neuroepithelial cells span the thickness of the tube's wall, connecting with the pial surface and with the ventricular or lumenal surface. They are joined at the lumen of the tube by junctional complexes, where they form a pseudostratified layer of epithelium called neuroepithelium.

Neural stem cells (NSCs) are self-renewing, multipotent cells that firstly generate the radial glial progenitor cells that generate the neurons and glia of the nervous system of all animals during embryonic development. Some neural progenitor stem cells persist in highly restricted regions in the adult vertebrate brain and continue to produce neurons throughout life. Differences in the size of the central nervous system are among the most important distinctions between the species and thus mutations in the genes that regulate the size of the neural stem cell compartment are among the most important drivers of vertebrate evolution.

Neuropoiesis is the process by which neural stem cells differentiate to form mature neurons, astrocytes, and oligodendrocytes in the adult mammal. This process is also referred to as adult neurogenesis.

<span class="mw-page-title-main">Subgranular zone</span>

The subgranular zone (SGZ) is a brain region in the hippocampus where adult neurogenesis occurs. The other major site of adult neurogenesis is the subventricular zone (SVZ) in the brain.

<span class="mw-page-title-main">NUMB (gene)</span> Protein-coding gene in the species Homo sapiens

Protein numb homolog is a protein that in humans is encoded by the NUMB gene. The protein encoded by this gene plays a role in the determination of cell fates during development. The encoded protein, whose degradation is induced in a proteasome-dependent manner by MDM2, is a membrane-bound protein that has been shown to associate with EPS15, LNX1, and NOTCH1. Four transcript variants encoding different isoforms have been found for this gene.

<span class="mw-page-title-main">Protein BTG2</span> Protein-coding gene in the species Homo sapiens

Protein BTG2 also known as BTG family member 2 or NGF-inducible anti-proliferative protein PC3 or NGF-inducible protein TIS21, is a protein that in humans is encoded by the BTG2 gene and in other mammals by the homologous Btg2 gene. This protein controls cell cycle progression and proneural genes expression by acting as a transcription coregulator that enhances or inhibits the activity of transcription factors.

<span class="mw-page-title-main">HES1</span> Protein-coding gene in the species Homo sapiens

Transcription factor HES1 is a protein that is encoded by the Hes1 gene, and is the mammalian homolog of the hairy gene in Drosophila. HES1 is one of the seven members of the Hes gene family (HES1-7). Hes genes code nuclear proteins that suppress transcription.

<span class="mw-page-title-main">ASCL1</span> Protein-coding gene in humans

Achaete-scute homolog 1 is a protein that in humans is encoded by the ASCL1 gene. Because it was discovered subsequent to studies on its homolog in Drosophila, the Achaete-scute complex, it was originally named MASH-1 for mammalian achaete scute homolog-1.

<span class="mw-page-title-main">OLIG2</span> Protein-coding gene in the species Homo sapiens

Oligodendrocyte transcription factor (OLIG2) is a basic helix-loop-helix (bHLH) transcription factor encoded by the OLIG2 gene. The protein is of 329 amino acids in length, 32 kDa in size and contains one basic helix-loop-helix DNA-binding domain. It is one of the three members of the bHLH family. The other two members are OLIG1 and OLIG3. The expression of OLIG2 is mostly restricted in central nervous system, where it acts as both an anti-neurigenic and a neurigenic factor at different stages of development. OLIG2 is well known for determining motor neuron and oligodendrocyte differentiation, as well as its role in sustaining replication in early development. It is mainly involved in diseases such as brain tumor and Down syndrome.

<span class="mw-page-title-main">ATOH1</span> Protein-coding gene in the species Homo sapiens

Protein atonal homolog 1 is a protein that in humans is encoded by the ATOH1 gene.

<span class="mw-page-title-main">PTF1A</span> Protein-coding gene in the species Homo sapiens

Pancreas transcription factor 1 subunit alpha is a protein that in humans is encoded by the PTF1A gene.

Gliogenesis is the generation of non-neuronal glia populations derived from multipotent neural stem cells.

<span class="mw-page-title-main">Neurogenin-2</span> Protein-coding gene in the species Homo sapiens

Neurogenin-2 is a protein that in humans is encoded by the NEUROG2 gene.

<span class="mw-page-title-main">Neurogenin-3</span> Mammalian protein found in Homo sapiens

Neurogenin-3 (NGN3) is a protein that in humans is encoded by the Neurog3 gene.

<span class="mw-page-title-main">Eomesodermin</span> Protein-coding gene in the species Homo sapiens

Eomesodermin also known as T-box brain protein 2 (Tbr2) is a protein that in humans is encoded by the EOMES gene.

Epigenetic regulation of neurogenesis is the role that epigenetics plays in the regulation of neurogenesis.

Proneural genes encode transcription factors of the basic helix-loop-helix (bHLH) class which are responsible for the development of neuroectodermal progenitor cells. Proneural genes have multiple functions in neural development. They integrate positional information and contribute to the specification of progenitor-cell identity. From the same ectodermal cell types, neural or epidermal cells can develop based on interactions between proneural and neurogenic genes. Neurogenic genes are so called because loss of function mutants show an increase number of developed neural precursors. On the other hand, proneural genes mutants fail to develop neural precursor cells.

Neurogenesis is the process by which nervous system cells, the neurons, are produced by neural stem cells (NSCs). In short, it is brain growth in relation to its organization. This occurs in all species of animals except the porifera (sponges) and placozoans. Types of NSCs include neuroepithelial cells (NECs), radial glial cells (RGCs), basal progenitors (BPs), intermediate neuronal precursors (INPs), subventricular zone astrocytes, and subgranular zone radial astrocytes, among others.

<span class="mw-page-title-main">Pancreatic progenitor cell</span>

Pancreatic progenitor cells are multipotent stem cells originating from the developing fore-gut endoderm which have the ability to differentiate into the lineage specific progenitors responsible for the developing pancreas.

References

  1. 1 2 3 Kageyama R, Ishibashi M, Takebayashi K, Tomita K (December 1997). "bHLH transcription factors and mammalian neuronal differentiation". The International Journal of Biochemistry & Cell Biology. 29 (12): 1389–1399. doi:10.1016/S1357-2725(97)89968-2. PMID   9570134.
  2. Hulme AJ, Maksour S, St-Clair Glover M, Miellet S, Dottori M (January 2022). "Making neurons, made easy: The use of Neurogenin-2 in neuronal differentiation". Stem Cell Reports. 17 (1): 14–34. doi:10.1016/j.stemcr.2021.11.015. PMC   8758946 . PMID   34971564.
  3. Rao MS, Jacobson M (2005). Developmental Neurobiology. New York: Kluwer Academic/Plenum. ISBN   0-306-48330-0.
  4. Ma Q, Fode C, Guillemot F, Anderson DJ (July 1999). "Neurogenin1 and neurogenin2 control two distinct waves of neurogenesis in developing dorsal root ganglia". Genes & Development. 13 (13): 1717–1728. doi:10.1101/gad.13.13.1717. PMC   316844 . PMID   10398684.
  5. Hodge RD, Hevner RF (August 2011). "Expression and actions of transcription factors in adult hippocampal neurogenesis". Developmental Neurobiology. 71 (8): 680–689. doi:10.1002/dneu.20882. PMC   3134120 . PMID   21412988.
  6. Rea J, Menci V, Tollis P, Santini T, Armaos A, Garone MG, et al. (July 2020). "HOTAIRM1 regulates neuronal differentiation by modulating NEUROGENIN 2 and the downstream neurogenic cascade". Cell Death & Disease. 11 (7): 527. doi:10.1038/s41419-020-02738-w. PMC   7359305 . PMID   32661334.
  7. Chen ZA, Wang JL, Liu RT, Ren JP, Wen LQ, Chen XJ, et al. (July 2009). "Liquiritin potentiate neurite outgrowth induced by nerve growth factor in PC12 cells". Cytotechnology. 60 (1–3): 125–132. doi:10.1007/s10616-009-9226-8. PMC   2780551 . PMID   19789989.
  8. Christensen EL, Beasley A, Radchuk J, Mielko ZE, Preston E, Stuckett S, et al. (June 2020). "ngn-1/neurogenin Activates Transcription of Multiple Terminal Selector Transcription Factors in the Caenorhabditis elegans Nervous System". G3. 10 (6): 1949–1962. doi:10.1534/g3.120.401126. PMC   7263688 . PMID   32273286.
  9. Universal protein resource accession number Q92886 for "Neurogenin-1" at UniProt.
  10. 1 2 3 4 5 6 7 8 9 Morrison SJ (May 2001). "Neuronal differentiation: proneural genes inhibit gliogenesis". Current Biology. 11 (9): R349–R351. Bibcode:2001CBio...11.R349M. doi: 10.1016/S0960-9822(01)00191-9 . PMID   11369245. S2CID   15885798.
  11. Rowitch DH, Kriegstein AR (November 2010). "Developmental genetics of vertebrate glial-cell specification". Nature. 468 (7321): 214–222. Bibcode:2010Natur.468..214R. doi:10.1038/nature09611. PMID   21068830. S2CID   573477.
  12. "Neurogenin-1 Products: R&D Systems". www.rndsystems.com. Archived from the original on 2014-01-16.
  13. Gammill LS, Bronner-Fraser M (October 2003). "Neural crest specification: migrating into genomics". Nature Reviews. Neuroscience. 4 (10): 795–805. doi:10.1038/nrn1219. PMID   14523379. S2CID   10863124.
  14. Universal protein resource accession number Q9H2A3 at UniProt.
  15. Harris WA, Sanes DH, Reh TA (2011). Development of the Nervous System (Third ed.). Boston: Academic Press. p.  15. ISBN   978-0-12-374539-2.
  16. Marquardt T, Pfaff SL (September 2001). "Cracking the transcriptional code for cell specification in the neural tube". Cell. 106 (6): 651–654. doi: 10.1016/S0092-8674(01)00499-8 . PMID   11572771. S2CID   2624758.
  17. 1 2 3 4 5 Lai HC, Johnson JE (April 2008). "Neurogenesis or neuronal specification: phosphorylation strikes again!". Neuron. 58 (1): 3–5. doi: 10.1016/j.neuron.2008.03.023 . PMID   18400155. S2CID   13530075.
  18. Ma YC, Song MR, Park JP, Henry Ho HY, Hu L, Kurtev MV, et al. (April 2008). "Regulation of motor neuron specification by phosphorylation of neurogenin 2". Neuron. 58 (1): 65–77. doi:10.1016/j.neuron.2008.01.037. PMC   2587148 . PMID   18400164.
  19. 1 2 3 4 5 6 7 Rukstalis JM, Habener JF (2009). "Neurogenin3: a master regulator of pancreatic islet differentiation and regeneration". Islets. 1 (3): 177–184. doi: 10.4161/isl.1.3.9877 . PMID   21099270.
  20. 1 2 Li HJ, Ray SK, Singh NK, Johnston B, Leiter AB (October 2011). "Basic helix-loop-helix transcription factors and enteroendocrine cell differentiation". Diabetes, Obesity & Metabolism. 13 Suppl 1 (1): 5–12. doi:10.1111/j.1463-1326.2011.01438.x. PMC   3467197 . PMID   21824251.
  21. 1 2 3 Watada H (June 2004). "Neurogenin 3 is a key transcription factor for differentiation of the endocrine pancreas". Endocrine Journal. 51 (3): 255–264. doi: 10.1507/endocrj.51.255 . PMID   15256770.
  22. Bramswig NC, Kaestner KH (October 2011). "Transcriptional regulation of α-cell differentiation". Diabetes, Obesity & Metabolism. 13 (Suppl 1): 13–20. doi: 10.1111/j.1463-1326.2011.01440.x . PMID   21824252. S2CID   691004.
  23. Gomez DL, O'Driscoll M, Sheets TP, Hruban RH, Oberholzer J, McGarrigle JJ, et al. (2015). "Neurogenin 3 Expressing Cells in the Human Exocrine Pancreas Have the Capacity for Endocrine Cell Fate". PLOS ONE. 10 (8): e0133862. Bibcode:2015PLoSO..1033862G. doi: 10.1371/journal.pone.0133862 . PMC   4545947 . PMID   26288179.
  24. Swales N, Martens GA, Bonné S, Heremans Y, Borup R, Van de Casteele M, et al. (2012). "Plasticity of adult human pancreatic duct cells by neurogenin3-mediated reprogramming". PLOS ONE. 7 (5): e37055. Bibcode:2012PLoSO...737055S. doi: 10.1371/journal.pone.0037055 . PMC   3351393 . PMID   22606327.
  25. Xu X, D'Hoker J, Stangé G, Bonné S, De Leu N, Xiao X, et al. (January 2008). "Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas". Cell. 132 (2): 197–207. doi: 10.1016/j.cell.2007.12.015 . PMID   18243096. S2CID   8058714.
  26. Van de Casteele M, Leuckx G, Baeyens L, Cai Y, Yuchi Y, Coppens V, et al. (March 2013). "Neurogenin 3+ cells contribute to β-cell neogenesis and proliferation in injured adult mouse pancreas". Cell Death & Disease. 4 (3): e523. doi:10.1038/cddis.2013.52. PMC   3613830 . PMID   23470530.
  27. Figeac F, Ilias A, Bailbe D, Portha B, Movassat J (October 2012). "Local in vivo GSK3β knockdown promotes pancreatic β cell and acinar cell regeneration in 90% pancreatectomized rat". Molecular Therapy. 20 (10): 1944–1952. doi:10.1038/mt.2012.112. PMC   3464647 . PMID   22828498.
  28. Li WC, Rukstalis JM, Nishimura W, Tchipashvili V, Habener JF, Sharma A, et al. (August 2010). "Activation of pancreatic-duct-derived progenitor cells during pancreas regeneration in adult rats". Journal of Cell Science. 123 (Pt 16): 2792–2802. doi:10.1242/jcs.065268. PMC   2915881 . PMID   20663919.
  29. Baeyens L, Lemper M, Leuckx G, De Groef S, Bonfanti P, Stangé G, et al. (January 2014). "Transient cytokine treatment induces acinar cell reprogramming and regenerates functional beta cell mass in diabetic mice". Nature Biotechnology. 32 (1): 76–83. doi:10.1038/nbt.2747. PMC   4096987 . PMID   24240391.
  30. Baeyens L, Bonné S, German MS, Ravassard P, Heimberg H, Bouwens L (November 2006). "Ngn3 expression during postnatal in vitro beta cell neogenesis induced by the JAK/STAT pathway". Cell Death and Differentiation. 13 (11): 1892–1899. doi: 10.1038/sj.cdd.4401883 . PMID   16514419.
  31. Lemper M, Leuckx G, Heremans Y, German MS, Heimberg H, Bouwens L, et al. (July 2015). "Reprogramming of human pancreatic exocrine cells to β-like cells". Cell Death and Differentiation. 22 (7): 1117–1130. doi:10.1038/cdd.2014.193. PMC   4572860 . PMID   25476775.
  32. Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA (October 2008). "In vivo reprogramming of adult pancreatic exocrine cells to beta-cells". Nature. 455 (7213): 627–632. Bibcode:2008Natur.455..627Z. doi:10.1038/nature07314. PMC   9011918 . PMID   18754011. S2CID   205214877.
  33. Sancho R, Gruber R, Gu G, Behrens A (August 2014). "Loss of Fbw7 reprograms adult pancreatic ductal cells into α, δ, and β cells". Cell Stem Cell. 15 (2): 139–153. doi:10.1016/j.stem.2014.06.019. PMC   4136739 . PMID   25105579.