Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2393 2022-04-20 12:45:11 |
2 format change Meta information modification 2393 2022-04-21 03:15:03 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Bartolomé, A.; , .; Chirikjian, M. MafA Regulation in β-Cells. Encyclopedia. Available online: https://encyclopedia.pub/entry/22009 (accessed on 26 July 2024).
Bartolomé A,  , Chirikjian M. MafA Regulation in β-Cells. Encyclopedia. Available at: https://encyclopedia.pub/entry/22009. Accessed July 26, 2024.
Bartolomé, Alberto, , Margot Chirikjian. "MafA Regulation in β-Cells" Encyclopedia, https://encyclopedia.pub/entry/22009 (accessed July 26, 2024).
Bartolomé, A., , ., & Chirikjian, M. (2022, April 20). MafA Regulation in β-Cells. In Encyclopedia. https://encyclopedia.pub/entry/22009
Bartolomé, Alberto, et al. "MafA Regulation in β-Cells." Encyclopedia. Web. 20 April, 2022.
MafA Regulation in β-Cells
Edit

β-cells are insulin-producing cells in the pancreas that maintain euglycemic conditions. Pancreatic β-cell maturity and function are regulated by a variety of transcription factors that enable the adequate expression of the cellular machinery involved in nutrient sensing and commensurate insulin secretion. One of the key factors in this regulation is MAF bZIP transcription factor A (MafA). MafA expression is decreased in type 2 diabetes, contributing to β-cell dysfunction and disease progression. The molecular biology underlying MafA is complex, with numerous transcriptional and post-translational regulatory nodes. 

MafA pancreatic beta cells beta cell maturity transcription factors diabetes

1. Introduction

Pancreatic β-cells are critical in maintaining euglycemia by responding to blood glucose oscillations. β-cell dysfunction is a feature of diabetes mellitus which is characterized by abnormally high blood glucose levels. β-cells are responsible for the synthesis, storage and secretion of insulin, a hormone that is paramount for metabolic homeostasis. One hundred years after the successful isolation of insulin by Banting and collaborators, studies on insulin and β-cells have determined many of the factors needed for normal β-cell function, which is best defined by the appropriate expression of canonical β-cell factors involved in glucose-stimulated insulin secretion. One of these factors is the MAF bZIP transcription factor A (MafA). MafA, a large basic leucine zipper (bZIP) transcription factor (TF), derives its name from the viral oncogene v-maf, which was isolated from musculoaponeurotic fibrosarcoma in chicken [1]. The seven members of the MAF family were subsequently identified by homology of their bZIP domain with v-maf and are divided into two subgroups according to molecular size: large MAFs (Maf, MafA, MafB and Nlr) and small MAFs (MafF, MafG and MafK), which lack a transactivation domain. These factors have roles in development and terminal differentiation across multiple tissues [2][3][4], but have also been identified in disease pathogenesis [5]. For instance, MafA was first identified as the trans-acting factor binding the RIPE3b/C1 element of the insulin promoter [6][7][8] and has since been characterized as an important TF for β-cell function that is dysregulated in diabetes progression [9][10]. These studies and others emphasized the critical role of MafA for β-cell function and suggested that elucidating mechanisms of MafA regulation would deepen the understanding of β-cell pathophysiology and potentially uncover therapeutic targets for the amelioration of β-cell dysfunction and diabetes progression.

2. MafA Regulates β-Cell Function

The importance of MafA for glucose homeostasis was shown in MafA-deficient mice, which displayed glucose intolerance after weaning due to impaired glucose-stimulated insulin secretion (GSIS) and age-dependent diabetes progression [11][12]. MafA-deficient mice had decreased mRNA levels of Ins1, Ins2, Pdx1, Neurod1 and Slc2a2, suggesting that MafA is essential for the transcriptional identity of β-cells. MafA works synergistically with Neurod1 and Pdx1 to transactivate the insulin promoter, which is an effect that could not be fully replicated with MafB nor Maf [13]. Along these lines, MafA promotes insulin transcription when overexpressed in rat islets [14] and when ectopically expressed in chick embryonic endoderm [15] or in other non-β-cell lines of endodermal origin [16][17].
Several studies suggest that MafA is dispensable for β-cell development but plays an essential role in β-cell maturation and glucose responsiveness of adult β-cells. In rodents, Mafa expression is low at birth, increasing during the postnatal period [18][19]. Interestingly, premature expression of MafA in pancreatic endocrine progenitors prevents differentiation into hormone-expressing cells [20][21], suggesting that the induction of MafA expression in β-cells is time sensitive. Consistently, in mice with whole pancreas MafA knockout (Mafa∆panc), the effects of MafA loss are not apparent until three weeks of age, when the KO mice showed lower β-cell mass, decreased insulin expression, and impaired glucose tolerance compared to controls [22]. Similarly, at twelve weeks of age in whole-body MafA knockout mice, there is a reduced β-cell:α-cell ratio, decreased islet insulin content and β-cell dedifferentiation into MafB-expressing, progenitor-like cells [12]. These results may recapitulate some aspects of normal islet development. MafB is actually the predominant large MAF protein expressed during pancreatic development in both α- and β-cells [4]. MafB can upregulate Ins1 and Ins2 transcription [8], but in adult mice, MafB is mainly expressed in α-cells and upregulates Gcg gene expression [4][23]. The decrease in β-cell MafB expression after birth is at least partly due to methylation, specifically by DNA methyltransferase 3a (Dnmt3a) which binds and represses at a region −1032 to −838 upstream of the Mafb transcriptional start site [24]. Mafb∆panc mice have higher blood glucose levels at P1, but two weeks after birth, blood glucose levels normalize. Likewise, at E15.5, Mafb∆panc mice have a lower number of insulin+ and glucagon+ cells compared to controls, but two weeks after birth, cell numbers became roughly the same [25]. MafB does seem to have a role in enhancing postnatal β-cell function under metabolic stress (pregnancy, high-fat diet) [24][26], but MafA remains critical for proper β-cell function and glucose homeostasis in mature β-cells, with forced MafB expression unable to compensate for MafA loss in adult mice [24].
Reduced MafA expression is associated with diabetes progression in mice and human patients. In islets isolated from diabetic db/db mice, MafA is decreased due to hyperglycemia-associated oxidative stress and c-Jun activity [9]. Similarly, islets from subjects with type 2 diabetes (T2D) display a marked decrease in MAFA mRNA levels and protein expression [10][27][28]. Several studies have shown possible mechanisms for this phenomenon. In β-cells from subjects with T2D [29] and in in vitro studies mimicking oxidative stress [10], MafA was found primarily in the cytoplasm rather than properly localized in the nucleus. However, when MafA levels were “rescued” in db/db mice, GSIS and β-cell mass improved, suggesting that preserving MafA expression in β-cells can still mitigate diabetes progression [30]. scRNA-seq analysis of human β-cells supports this concept as well, wherein metabolically inflexible β-cells show lower MAFA activity as compared to healthy β-cells [31]. This finding is thought to align with previous patterns of T2D-induced oxidative stress, in which case MafA activity is high under healthy islet conditions but can also increase as a protective mechanism against acute oxidative stress [32]. In a different scRNA-seq analysis of human islets, β-cells co-expressing MAFA/MAFB had increased expression of genes related to β-cell identity, glucose metabolism and exocytosis compared to β-cells that only expressed one or neither TF [33]. Since MAFA and MAFB are capable of forming heterodimers [8][34], it is possible that the co-expression of both can enhance the β-cell maturity transcriptional program. These and other studies have documented the functional and transcriptional heterogeneity of β-cells [31][33][35].

3. MafA Target Genes

MafA transcriptional activity is necessary for proper postnatal β-cell function. MafA was first identified as a TF of insulin that binds to the C1 element of the insulin gene promoter. MAF proteins, including MafA, bind to the MAF-recognition elements (MAREs), through a DNA consensus sequence TGCTGAC(G)TCAGCA [36]. However, in β-cells, the MARE sequence within the C1 element deviates slightly from the consensus sequence. The MARE sequence in C1 element of the rat and mouse Ins2 promoter is TGCAGCTTCAGCC whereas the equivalent in the human INS promoter is TGCAGCCTCAGCC (the underlined nucleotides indicate deviations from the MARE consensus sequence) [7][16][37]. Interestingly, in addition to the C1 element, three other MARE sites (MARE1, MARE2 and MARE3) were also identified in the rat Ins2 and human INS promoter. MafA can bind to MARE2 and MARE3 in the rat Ins2 promoter while MafA can bind to MARE1 in the human INS promoter [16].
Although MafA can activate the insulin promoter alone, the addition of Neurod1 and Pdx1, which bind the E box and A box of the insulin promoter, respectively, synergistically promote insulin transcription [13]. Consistently, mice with homozygous CRISPR-Cas9-introduced mutations in the C1 box of the Ins1 and Ins2 gene promoters are glucose intolerant. These C1 box mutations also prevented MafA from activating the promoter in luciferase assays, further supporting the importance of MafA in upregulating the promoter of its target genes [38]. Human mutations characterized by a deletion of the C1 and E1 elements of the INS promoter have been shown to cause permanent neonatal diabetes, which may be attributed to the disruption of MAFA and NEUROD1 binding sites [39][40]. Other factors are also involved in this regulation. For example, Gli-similar 3 (Glis3), a Krüppel-like zinc finger transcription factor, activates insulin transcription by recruiting histone acetyltransferase Creb-binding protein (CBP) and by acting as a scaffold for the MafA, NeuroD1 and Pdx1 complex [41].
Beyond insulin, MafA increases the expression of a wide range of genes involved in β-cell function, including genes implicated in glucose sensing, insulin processing, Ca2+ influx and oxidative phosphorylation. Many studies have identified a plethora of MafA target genes, all having important roles in proper β-cell function, further supporting MafA’s critical role in β-cells. While some studies have found direct MafA target genes (i.e., MafA directly binds to the promoter of the target genes), other studies describe genes that are differentially expressed when MafA protein is overexpressed or silenced, without showing direct MafA binding to the target gene promoters (i.e., MafA knockout or overexpression causes a decrease or increase in the target gene expression, respectively). Some of these MafA-target genes are summarized in Table 1. The lack of promoter binding evidence does not necessarily mean MafA cannot bind to the promoter of these genes, but rather that MafA may regulate these genes through intermediaries rather than direct promoter binding.
Table 1. MafA target genes. (*) denotes evidence that MafA directly binds target gene promoter.

Gene Symbol

Gene Name

Gene Function

Reference

Cacng4 *

Voltage-dependent calcium channel gamma-4 subunit

Enhances L-type Ca2+-mediated Ca2+ entry into β-cell

[42]

Chrnb4 *

Cholinergic receptor nicotinic beta 4

Subunit of the nicotinic acetylcholine receptor

[43]

Cox6a2 *

Cytochrome C oxidase subunit 6A2

One out of 13 subunits of cytochrome C oxidase complex (Complex IV), the last enzyme in the electron transport chain

[44]

G6pc2 *

Glucose-6-phosphate catalytic subunit related protein

Islet-specific enzyme that hydrolyzes glucose-6-phosphate, limits basal insulin secretion

[45]

Gck

Glucokinase

Phosphorylates glucose to glucose-6-phosphate in pancreatic islets and hepatocytes. Considered the β-cell glucose sensor

[46]

Glp1r

Glucagon-like peptide 1 receptor

Receptor for Glucagon-like peptide 1 (Glp1), a stimulator of insulin secretion

[46]

Ins1 *

Insulin I

One of two insulin genes in mouse, on chromosome 19

[11]

Ins2 *

Insulin II

One of two insulin genes in mouse, on chromosome 7

[11]

Maob *

Monoamine oxidase B

Metabolizes monoamine neurotransmitters, specifically benzylamine, dopamine and phenylethylamine

[47]

Neurod1

Neurogenic differentiation 1

Transactivator of genes important for β-cell maturation and function, including insulin

[46]

Nkx6.1

NK6 homeobox1

TF involved in β-cell development and regulation of genes involved in mature β-cell function

[46][48]

Pcsk1

Proprotein convertase subtilisin/kexin type 1

Proprotein convertase, which processes proinsulin in β-cells

[46]

Pcx

Pyruvate carboxylase

Catalyzes the conversion of pyruvate to oxaloacetate

[46]

Pdx1 *

Pancreatic and duodenal homeobox 1

TF important pancreas development and for mature β-cell function

[46][49]

PPP1R1A

Protein phosphatase 1, regulatory inhibitor subunit 1A

Regulates cAMP/PKA signaling pathway

Promotes Glp1-induced GSIS

[50]

Prlr

Prolactin Receptor

Involved in increasing β-cell mass during pregnancy

[51]

Slc2a2 *

Solute Carrier Family 2 Member 2

Glucose transporter 2, transmembrane glucose transporter with a high Km for glucose

[46][52]

Slc80a8

Solute carrier family 30 member 8

Zinc transporter on insulin granules in β-cells

[19][43]

4. Regulation of MafA Transcription

The first level of MafA regulation is through Mafa gene expression. The Mafa promoter consists of six regions (R1-R6), which contain highly conserved sequences 25 kb upstream the 5′ MafA untranslated region (UTR). R1 spans from −9389 to −9194, R2 spans from −8420 to −8293, R3 spans from −8118 to −7750, R4 spans from −6622 to −6441, R5 spans from −6217 to −6031 and R6 spans from −250 to +56. Of these, R3 has been shown to be most necessary, but not sufficient, for transcriptional initiation mediated by direct TF binding [53][54]. Many transcription factors bind R3 on the Mafa promoter, including Pax6, Nkx6.1, Nkx2.2, Pdx1, Hnf1a, Foxa2 and Isl1. Changes in these transcription factors directly correlate with Mafa expression. However, there are some exceptions. Neurod1, another important insulin transcription factor, can bind to R3 of the Mafa promoter, but Neurod1−/− mice do not have detectable changes in MafA staining [55]. However, in a more recent study, Neurod1∆endo mice had lower Mafa mRNA levels, suggesting that Neurod1 may regulate Mafa transcription [56]. Additionally, Pax4 is a negative regulator of Mafa. Pax4 overexpression limits R3-mediated promoter activity, but there is limited evidence of direct Pax4 binding to R3 via ChIP, possibly due to Pax4 regulating Mafa in an indirect manner [57].
Other TFs bind elsewhere in the MafA promoter or regulate Mafa transcription through other mechanisms, as summarized in Table 2. For instance, Onecut1, a TF involved in pancreatic endocrine development that is upregulated in adult db/db mice, acts as a negative regulator of Mafa transcription by preventing Foxa2 from interacting with the Mafa promoter, highlighting the importance of the transcriptional regulation of Mafa in metabolic disease states [58].
Table 2. Known transcriptional regulators of Mafa gene expression.

Protein Symbol

Protein Name

Mafa Transcription Mechanism

Reference

Pax6

Paired box protein Pax-6

Binds R1, R3 and R6 of the Mafa promoter

[55]

Nkx6.1

NK6 homeobox 1

Binds R3 of the Mafa promoter

[55]

Nkx2.2

NK2 homeobox 2

Binds R3 of the Mafa promoter

[54]

Pdx1

Pancreatic and duodenal homeobox 1

Binds R3 and R6 of the Mafa promoter

[54][55]

Hnf1a

Hepatocyte nuclear factor 1-alpha

Binds R3 of the Mafa promoter

[53]

Foxa2

Forkhead box A2

Binds R3 of the Mafa promoter

[54]

Isl1

Insulin gene enhancer protein ISL-1

Binds R3 of the Mafa promoter

[59]

Neurod1

Neurogenic differentiation 1

Binds R3 of the Mafa promoter

[54]

Pax4

Paired box protein Pax-4

Negative regulator of Mafa, potentially by interfering other factors from binding R3 of the Mafa promoter

[57]

Mafb

Transcription factor MafB

Binds R3 of the Mafa promoter

[55][60]

Onecut1

One cut domain, family member 1

Prevents Foxa2 from binding to the Mafa promoter

[58]

Foxo1

Forkhead box O1

Binds to the forkhead element of the Mafa promoter

[32]

Thra

Thyroid hormone receptor alpha

Binds to Thyroid hormone response element (TRE), which are located from −1927 to −1946 and from +647 to +659 (named Site 2 and Site 3, respectively)

[61]

CREB

cAMP responsive element binding protein

Constitutively binds to the cAMP response element (CRE), spanning from −1342 to −1346, of the Mafa promoter

[62]

5. MafA Post-Translational Modifications

The previously discussed studies indicate the importance and potential mechanisms of Mafa transcriptional regulation. Post-translational modifications (PTMs) add a second layer of regulation, allowing for dynamic changes in MafA activity or stability in response to changes in β-cell environment. While most documented PTMs are found in the N-terminal transactivation domain of MafA [63], other regions of MafA, such as the extended homology region, the basic domain and the leucine-zipper, affect DNA recognition and binding and are also likely affected by the addition of PTMs [5] (Figure 1).
Figure 1. Linear schematic of MAFA depicting MAFA domains and known post-translationally modified residues. Transactivation: transactivation domain. His: histamine-rich region. Basic: basic domain. bZIP: Leucine zipper.

References

  1. Nishizawa, M.; Kataoka, K.; Goto, N.; Fujiwara, K.T.; Kawai, S. V-maf, a viral oncogene that encodes a “leucine zipper” motif. Proc. Natl. Acad. Sci. USA 1989, 86, 7711–7715.
  2. Reza, H.M.; Yasuda, K. Roles of Maf family proteins in lens development. Dev. Dyn. 2004, 229, 440–448.
  3. Zhang, C.; Guo, Z.M. Multiple functions of Maf in the regulation of cellular development and differentiation. Diabetes Metab. Res. Rev. 2015, 31, 773–778.
  4. Artner, I.; Le Lay, J.; Hang, Y.; Elghazi, L.; Schisler, J.C.; Henderson, E.; Sosa-Pineda, B.; Stein, R. MafB: An activator of the glucagon gene expressed in developing islet alpha- and beta-cells. Diabetes 2006, 55, 297–304.
  5. Eychene, A.; Rocques, N.; Pouponnot, C. A new MAFia in cancer. Nat. Rev. Cancer 2008, 8, 683–693.
  6. Olbrot, M.; Rud, J.; Moss, L.G.; Sharma, A. Identification of beta-cell-specific insulin gene transcription factor RIPE3b1 as mammalian MafA. Proc. Natl. Acad. Sci. USA 2002, 99, 6737–6742.
  7. Kataoka, K.; Han, S.I.; Shioda, S.; Hirai, M.; Nishizawa, M.; Handa, H. MafA is a glucose-regulated and pancreatic beta-cell-specific transcriptional activator for the insulin gene. J. Biol. Chem. 2002, 277, 49903–49910.
  8. Matsuoka, T.A.; Zhao, L.; Artner, I.; Jarrett, H.W.; Friedman, D.; Means, A.; Stein, R. Members of the large Maf transcription family regulate insulin gene transcription in islet beta cells. Mol. Cell. Biol. 2003, 23, 6049–6062.
  9. Matsuoka, T.A.; Kaneto, H.; Miyatsuka, T.; Yamamoto, T.; Yamamoto, K.; Kato, K.; Shimomura, I.; Stein, R.; Matsuhisa, M. Regulation of MafA expression in pancreatic beta-cells in db/db mice with diabetes. Diabetes 2010, 59, 1709–1720.
  10. Guo, S.; Dai, C.; Guo, M.; Taylor, B.; Harmon, J.S.; Sander, M.; Robertson, R.P.; Powers, A.C.; Stein, R. Inactivation of specific beta cell transcription factors in type 2 diabetes. J. Clin. Investig. 2013, 123, 3305–3316.
  11. Zhang, C.; Moriguchi, T.; Kajihara, M.; Esaki, R.; Harada, A.; Shimohata, H.; Oishi, H.; Hamada, M.; Morito, N.; Hasegawa, K.; et al. MafA is a key regulator of glucose-stimulated insulin secretion. Mol. Cell. Biol. 2005, 25, 4969–4976.
  12. Nishimura, W.; Takahashi, S.; Yasuda, K. MafA is critical for maintenance of the mature beta cell phenotype in mice. Diabetologia 2015, 58, 566–574.
  13. Aramata, S.; Han, S.I.; Yasuda, K.; Kataoka, K. Synergistic activation of the insulin gene promoter by the beta-cell enriched transcription factors MafA, Beta2, and Pdx1. Biochim. Biophys. Acta 2005, 1730, 41–46.
  14. Zhao, L.; Guo, M.; Matsuoka, T.A.; Hagman, D.K.; Parazzoli, S.D.; Poitout, V.; Stein, R. The islet beta cell-enriched MafA activator is a key regulator of insulin gene transcription. J. Biol. Chem. 2005, 280, 11887–11894.
  15. Artner, I.; Hang, Y.; Guo, M.; Gu, G.; Stein, R. MafA is a dedicated activator of the insulin gene in vivo. J. Endocrinol. 2008, 198, 271–279.
  16. Matsuoka, T.A.; Artner, I.; Henderson, E.; Means, A.; Sander, M.; Stein, R. The MafA transcription factor appears to be responsible for tissue-specific expression of insulin. Proc. Natl. Acad. Sci. USA 2004, 101, 2930–2933.
  17. Matsuoka, T.A.; Kaneto, H.; Stein, R.; Miyatsuka, T.; Kawamori, D.; Henderson, E.; Kojima, I.; Matsuhisa, M.; Hori, M.; Yamasaki, Y. MafA regulates expression of genes important to islet beta-cell function. Mol. Endocrinol. 2007, 21, 2764–2774.
  18. Aguayo-Mazzucato, C.; Koh, A.; El Khattabi, I.; Li, W.C.; Toschi, E.; Jermendy, A.; Juhl, K.; Mao, K.; Weir, G.C.; Sharma, A.; et al. Mafa expression enhances glucose-responsive insulin secretion in neonatal rat beta cells. Diabetologia 2011, 54, 583–593.
  19. Artner, I.; Hang, Y.; Mazur, M.; Yamamoto, T.; Guo, M.; Lindner, J.; Magnuson, M.A.; Stein, R. MafA and MafB regulate genes critical to beta-cells in a unique temporal manner. Diabetes 2010, 59, 2530–2539.
  20. He, K.H.; Juhl, K.; Karadimos, M.; El Khattabi, I.; Fitzpatrick, C.; Bonner-Weir, S.; Sharma, A. Differentiation of pancreatic endocrine progenitors reversibly blocked by premature induction of MafA. Dev. Biol. 2014, 385, 2–12.
  21. Nishimura, W.; Bonner-Weir, S.; Sharma, A. Expression of MafA in pancreatic progenitors is detrimental for pancreatic development. Dev. Biol. 2009, 333, 108–120.
  22. Hang, Y.; Yamamoto, T.; Benninger, R.K.; Brissova, M.; Guo, M.; Bush, W.; Piston, D.W.; Powers, A.C.; Magnuson, M.; Thurmond, D.C.; et al. The MafA transcription factor becomes essential to islet beta-cells soon after birth. Diabetes 2014, 63, 1994–2005.
  23. Katoh, M.C.; Jung, Y.; Ugboma, C.M.; Shimbo, M.; Kuno, A.; Basha, W.A.; Kudo, T.; Oishi, H.; Takahashi, S. MafB Is Critical for Glucagon Production and Secretion in Mouse Pancreatic alpha Cells In Vivo. Mol. Cell. Biol. 2018, 38, e00504-17.
  24. Cyphert, H.A.; Walker, E.M.; Hang, Y.; Dhawan, S.; Haliyur, R.; Bonatakis, L.; Avrahami, D.; Brissova, M.; Kaestner, K.H.; Bhushan, A.; et al. Examining How the MAFB Transcription Factor Affects Islet beta-Cell Function Postnatally. Diabetes 2019, 68, 337–348.
  25. Conrad, E.; Dai, C.; Spaeth, J.; Guo, M.; Cyphert, H.A.; Scoville, D.; Carroll, J.; Yu, W.M.; Goodrich, L.V.; Harlan, D.M.; et al. The MAFB transcription factor impacts islet alpha-cell function in rodents and represents a unique signature of primate islet beta-cells. Am. J. Physiol. Endocrinol. Metab. 2016, 310, E91–E102.
  26. Xiafukaiti, G.; Maimaiti, S.; Ogata, K.; Kuno, A.; Kudo, T.; Shawki, H.H.; Oishi, H.; Takahashi, S. MafB Is Important for Pancreatic beta-Cell Maintenance under a MafA-Deficient Condition. Mol. Cell. Biol. 2019, 39, e00080-19.
  27. Bonnavion, R.; Jaafar, R.; Kerr-Conte, J.; Assade, F.; van Stralen, E.; Leteurtre, E.; Pouponnot, C.; Gargani, S.; Pattou, F.; Bertolino, P.; et al. Both PAX4 and MAFA are expressed in a substantial proportion of normal human pancreatic alpha cells and deregulated in patients with type 2 diabetes. PLoS ONE 2013, 8, e72194.
  28. Butler, A.E.; Robertson, R.P.; Hernandez, R.; Matveyenko, A.V.; Gurlo, T.; Butler, P.C. Beta cell nuclear musculoaponeurotic fibrosarcoma oncogene family A (MafA) is deficient in type 2 diabetes. Diabetologia 2012, 55, 2985–2988.
  29. Cinti, F.; Bouchi, R.; Kim-Muller, J.Y.; Ohmura, Y.; Sandoval, P.R.; Masini, M.; Marselli, L.; Suleiman, M.; Ratner, L.E.; Marchetti, P.; et al. Evidence of beta-Cell Dedifferentiation in Human Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2016, 101, 1044–1054.
  30. Matsuoka, T.A.; Kaneto, H.; Kawashima, S.; Miyatsuka, T.; Tochino, Y.; Yoshikawa, A.; Imagawa, A.; Miyazaki, J.; Gannon, M.; Stein, R.; et al. Preserving Mafa expression in diabetic islet beta-cells improves glycemic control in vivo. J. Biol. Chem. 2015, 290, 7647–7657.
  31. Son, J.; Ding, H.; Farb, T.B.; Efanov, A.M.; Sun, J.; Gore, J.L.; Syed, S.K.; Lei, Z.; Wang, Q.; Accili, D.; et al. BACH2 inhibition reverses beta cell failure in type 2 diabetes models. J. Clin. Investig. 2021, 131, e153876.
  32. Kitamura, Y.I.; Kitamura, T.; Kruse, J.P.; Raum, J.C.; Stein, R.; Gu, W.; Accili, D. FoxO1 protects against pancreatic beta cell failure through NeuroD and MafA induction. Cell Metab. 2005, 2, 153–163.
  33. Shrestha, S.; Saunders, D.C.; Walker, J.T.; Camunas-Soler, J.; Dai, X.Q.; Haliyur, R.; Aramandla, R.; Poffenberger, G.; Prasad, N.; Bottino, R.; et al. Combinatorial transcription factor profiles predict mature and functional human islet alpha and beta cells. JCI Insight 2021, 6, e151621.
  34. Benkhelifa, S.; Provot, S.; Lecoq, O.; Pouponnot, C.; Calothy, G.; Felder-Schmittbuhl, M.P. mafA, a novel member of the maf proto-oncogene family, displays developmental regulation and mitogenic capacity in avian neuroretina cells. Oncogene 1998, 17, 247–254.
  35. Camunas-Soler, J.; Dai, X.Q.; Hang, Y.; Bautista, A.; Lyon, J.; Suzuki, K.; Kim, S.K.; Quake, S.R.; MacDonald, P.E. Patch-Seq Links Single-Cell Transcriptomes to Human Islet Dysfunction in Diabetes. Cell Metab. 2020, 31, 1017–1031.e1014.
  36. Lu, X.; Guanga, G.P.; Wan, C.; Rose, R.B. A novel DNA binding mechanism for maf basic region-leucine zipper factors inferred from a MafA-DNA complex structure and binding specificities. Biochemistry 2012, 51, 9706–9717.
  37. Kataoka, K.; Noda, M.; Nishizawa, M. Maf nuclear oncoprotein recognizes sequences related to an AP-1 site and forms heterodimers with both Fos and Jun. Mol. Cell. Biol. 1994, 14, 700–712.
  38. Noguchi, H.; Miyagi-Shiohira, C.; Nakashima, Y.; Kinjo, T.; Saitoh, I.; Watanabe, M. Mutations in the C1 element of the insulin promoter lead to diabetic phenotypes in homozygous mice. Commun. Biol. 2020, 3, 309.
  39. Garin, I.; Edghill, E.L.; Akerman, I.; Rubio-Cabezas, O.; Rica, I.; Locke, J.M.; Maestro, M.A.; Alshaikh, A.; Bundak, R.; del Castillo, G.; et al. Recessive mutations in the INS gene result in neonatal diabetes through reduced insulin biosynthesis. Proc. Natl. Acad. Sci. USA 2010, 107, 3105–3110.
  40. Akerman, I.; Maestro, M.A.; De Franco, E.; Grau, V.; Flanagan, S.; Garcia-Hurtado, J.; Mittler, G.; Ravassard, P.; Piemonti, L.; Ellard, S.; et al. Neonatal diabetes mutations disrupt a chromatin pioneering function that activates the human insulin gene. Cell Rep. 2021, 35, 108981.
  41. ZeRuth, G.T.; Takeda, Y.; Jetten, A.M. The Kruppel-like protein Gli-similar 3 (Glis3) functions as a key regulator of insulin transcription. Mol. Endocrinol. 2013, 27, 1692–1705.
  42. Luan, C.; Ye, Y.; Singh, T.; Barghouth, M.; Eliasson, L.; Artner, I.; Zhang, E.; Renstrom, E. The calcium channel subunit gamma-4 is regulated by MafA and necessary for pancreatic beta-cell specification. Commun. Biol. 2019, 2, 106.
  43. Ganic, E.; Singh, T.; Luan, C.; Fadista, J.; Johansson, J.K.; Cyphert, H.A.; Bennet, H.; Storm, P.; Prost, G.; Ahlenius, H.; et al. MafA-Controlled Nicotinic Receptor Expression Is Essential for Insulin Secretion and Is Impaired in Patients with Type 2 Diabetes. Cell Rep. 2016, 14, 1991–2002.
  44. Nagai, Y.; Matsuoka, T.A.; Shimo, N.; Miyatsuka, T.; Miyazaki, S.; Tashiro, F.; Miyazaki, J.I.; Katakami, N.; Shimomura, I. Glucotoxicity-induced suppression of Cox6a2 expression provokes beta-cell dysfunction via augmented ROS production. Biochem. Biophys. Res. Commun. 2021, 556, 134–141.
  45. Martin, C.C.; Flemming, B.P.; Wang, Y.; Oeser, J.K.; O’Brien, R.M. Foxa2 and MafA regulate islet-specific glucose-6-phosphatase catalytic subunit-related protein gene expression. J. Mol. Endocrinol. 2008, 41, 315–328.
  46. Wang, H.; Brun, T.; Kataoka, K.; Sharma, A.J.; Wollheim, C.B. MAFA controls genes implicated in insulin biosynthesis and secretion. Diabetologia 2007, 50, 348–358.
  47. Ganic, E.; Johansson, J.K.; Bennet, H.; Fex, M.; Artner, I. Islet-specific monoamine oxidase A and B expression depends on MafA transcriptional activity and is compromised in type 2 diabetes. Biochem. Biophys. Res. Commun. 2015, 468, 629–635.
  48. Aigha, I.I.; Abdelalim, E.M. NKX6.1 transcription factor: A crucial regulator of pancreatic beta cell development, identity, and proliferation. Stem Cell Res. Ther. 2020, 11, 459.
  49. Vanhoose, A.M.; Samaras, S.; Artner, I.; Henderson, E.; Hang, Y.; Stein, R. MafA and MafB regulate Pdx1 transcription through the Area II control region in pancreatic beta cells. J. Biol. Chem. 2008, 283, 22612–22619.
  50. Cataldo, L.R.; Vishnu, N.; Singh, T.; Bertonnier-Brouty, L.; Bsharat, S.; Luan, C.; Renstrom, E.; Prasad, R.B.; Fex, M.; Mulder, H.; et al. The MafA-target gene PPP1R1A regulates GLP1R-mediated amplification of glucose-stimulated insulin secretion in beta-cells. Metabolism 2021, 118, 154734.
  51. Eto, K.; Nishimura, W.; Oishi, H.; Udagawa, H.; Kawaguchi, M.; Hiramoto, M.; Fujiwara, T.; Takahashi, S.; Yasuda, K. MafA is required for postnatal proliferation of pancreatic beta-cells. PLoS ONE 2014, 9, e104184.
  52. Ono, Y.; Kataoka, K. MafA, NeuroD1, and HNF1beta synergistically activate the Slc2a2 (Glut2) gene in beta-cells. J. Mol. Endocrinol. 2021, 67, 71–82.
  53. Hunter, C.S.; Maestro, M.A.; Raum, J.C.; Guo, M.; Thompson, F.H., 3rd; Ferrer, J.; Stein, R. Hnf1alpha (MODY3) regulates beta-cell-enriched MafA transcription factor expression. Mol. Endocrinol. 2011, 25, 339–347.
  54. Raum, J.C.; Gerrish, K.; Artner, I.; Henderson, E.; Guo, M.; Sussel, L.; Schisler, J.C.; Newgard, C.B.; Stein, R. FoxA2, Nkx2.2, and PDX-1 regulate islet beta-cell-specific mafA expression through conserved sequences located between base pairs -8118 and -7750 upstream from the transcription start site. Mol. Cell. Biol. 2006, 26, 5735–5743.
  55. Raum, J.C.; Hunter, C.S.; Artner, I.; Henderson, E.; Guo, M.; Elghazi, L.; Sosa-Pineda, B.; Ogihara, T.; Mirmira, R.G.; Sussel, L.; et al. Islet beta-cell-specific MafA transcription requires the 5′-flanking conserved region 3 control domain. Mol. Cell. Biol. 2010, 30, 4234–4244.
  56. Romer, A.I.; Singer, R.A.; Sui, L.; Egli, D.; Sussel, L. Murine Perinatal beta-Cell Proliferation and the Differentiation of Human Stem Cell-Derived Insulin-Expressing Cells Require NEUROD1. Diabetes 2019, 68, 2259–2271.
  57. Hu He, K.H.; Lorenzo, P.I.; Brun, T.; Jimenez Moreno, C.M.; Aeberhard, D.; Vallejo Ortega, J.; Cornu, M.; Thorel, F.; Gjinovci, A.; Thorens, B.; et al. In vivo conditional Pax4 overexpression in mature islet beta-cells prevents stress-induced hyperglycemia in mice. Diabetes 2011, 60, 1705–1715.
  58. Yamamoto, K.; Matsuoka, T.A.; Kawashima, S.; Takebe, S.; Kubo, F.; Miyatsuka, T.; Kaneto, H.; Shimomura, I. A novel function of Onecut1 protein as a negative regulator of MafA gene expression. J. Biol. Chem. 2013, 288, 21648–21658.
  59. Du, A.; Hunter, C.S.; Murray, J.; Noble, D.; Cai, C.L.; Evans, S.M.; Stein, R.; May, C.L. Islet-1 is required for the maturation, proliferation, and survival of the endocrine pancreas. Diabetes 2009, 58, 2059–2069.
  60. Artner, I.; Blanchi, B.; Raum, J.C.; Guo, M.; Kaneko, T.; Cordes, S.; Sieweke, M.; Stein, R. MafB is required for islet beta cell maturation. Proc. Natl. Acad. Sci. USA 2007, 104, 3853–3858.
  61. Aguayo-Mazzucato, C.; Zavacki, A.M.; Marinelarena, A.; Hollister-Lock, J.; El Khattabi, I.; Marsili, A.; Weir, G.C.; Sharma, A.; Larsen, P.R.; Bonner-Weir, S. Thyroid hormone promotes postnatal rat pancreatic beta-cell development and glucose-responsive insulin secretion through MAFA. Diabetes 2013, 62, 1569–1580.
  62. Blanchet, E.; Van de Velde, S.; Matsumura, S.; Hao, E.; LeLay, J.; Kaestner, K.; Montminy, M. Feedback inhibition of CREB signaling promotes beta cell dysfunction in insulin resistance. Cell Rep. 2015, 10, 1149–1157.
  63. Guo, S.; Vanderford, N.L.; Stein, R. Phosphorylation within the MafA N terminus regulates C-terminal dimerization and DNA binding. J. Biol. Chem. 2010, 285, 12655–12661.
More
Information
Subjects: Cell Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 559
Revisions: 2 times (View History)
Update Date: 21 Apr 2022
1000/1000
Video Production Service