scispace - formally typeset
Search or ask a question
Posted ContentDOI

NFIC regulates ribosomal biology and ER stress in pancreatic acinar cells and suppresses PDAC initiation

TL;DR: In this paper, NFIC binding sites are found at very short distances from NR5A2-bound genomic regions and both proteins co-occur in the same complex and NFIC dampens the ER stress program through its binding to ER stress gene promoters and is required for complete resolution of Tunicamycin-mediated ER stress.
Abstract: Tissue-specific differentiation is driven by specialized transcriptional networks. Pancreatic acinar cells crucially rely on the PTF1 complex, and on additional transcription factors, to deploy their transcriptional program. Here, we identify NFIC as a novel regulator of acinar differentiation using a variety of methodological strategies. NFIC binding sites are found at very short distances from NR5A2-bound genomic regions and both proteins co-occur in the same complex. Nfic knockout mice show reduced expression of acinar genes and, in ChIP-seq experiments, NFIC binds the promoters of acinar genes. In addition, NFIC binds to the promoter of, and regulates, genes involved in RNA and protein metabolism; in Nfic knockout mice, p-RS6K1 and p-IEF4E are down-regulated indicating reduced activity of the mTOR pathway. In 266-6 acinar cells, NFIC dampens the ER stress program through its binding to ER stress gene promoters and is required for complete resolution of Tunicamycin-mediated ER stress. Normal human pancreata from subjects with low NFIC mRNA levels display reduced epxression of genes down-regulated in Nfic knockout mice. Consistently, NFIC displays reduced expression upon induced acute pancreatitis and is required for proper recovery after damage. Finally, expression of NFIC is lower in samples of mouse and human pancreatic ductal adenocarcinoma and Nfic knockout mice develop an increased number of mutant Kras-driven pre-neoplastic lesions.

Summary (3 min read)

INTRODUCTION

  • Pancreatic acinar cells are highly specialized protein synthesis factories that have a well-developed rough endoplasmic reticulum (ER), a prominent Golgi complex, and abundant secretory granules 1 .
  • Monoallelic or homozygous inactivation of several acinar transcriptional regulators in the germline, the embryonic pancreas, or the adult pancreas can result in compromised acinar function that favors loss of cellular identity and poises acinar cells for transformation upon activation of mutant KRas 10, 11, 12 .
  • Additional roles have been proposed through the regulation of Trp53 15, 16, 17 .
  • (which was not certified by peer review) is the author/funder.
  • Using a combination of omics analyses and studies in knockout mice and cultured cells, the authors now uncover novel roles of NFIC as a regulator of acinar function whose major impact is at the level of the ER stress response in murine and human pancreas.

Identification of novel transcription factors involved in the regulation of pancreatic

  • To discover novel transcription factors that might cooperate with known acinar regulators (e.g. PTF1A, GATA6, NR5A2, and MIST1), the authors reanalyzed publicly available ChIP-sequencing data and used HOMER to search for motifs enriched in the sequencing reads.
  • Motifs corresponding to RBPJ/RBPJL and HNF1, known regulators of acinar differentiation, were also enriched, thus validating the strategy applied.
  • Of all NFI family members, NFIC is expressed at highest levels in both mouse and human pancreas ; therefore, the authors focused on NFIC for further study.
  • Analysis of the published ChIP-Seq data revealed several PTF1A and NR5A2 peaks in the proximal Nfic promoter and the binding was confirmed by ChIP-qPCR , strongly suggesting that Nfic is a PTF1A and NR5A2 target.
  • The copyright holder for this preprint this version posted August 9, 2021.

DISCUSSION

  • Increasing evidence supports an "analog" differentiation model whereby additional TFs are required for "completion" of this process.
  • The conservation of spacing between NR5A2 and NFIC motifs among NR5A2 target genes supports transcriptional cooperation.
  • Comparison of NR5A2 ChIP-Seq data from embryonic and adult pancreas indicates that the role of NFIC is mainly in the latter, supporting its requirement for completion of acinar maturation and highlighting a functional role distinct from that of NR5A2.
  • NFI proteins were first proposed to be involved in the regulation of ubiquitous genes but they can also regulate tissue-specific genes 13 , including CEL in the mammary gland and DSPP in odontoblasts 14, 19 .
  • These findings suggest that the activation of pro-inflammatory phenotypes in mice in which acinar cells fail to acquire normal maturation can result from both direct (NR5A2 and GATA6) 4, 7, 10, 12 and indirect (PTF1A and NFIC) mechanisms, in agreement with the ChIP-Seq data available.

NFIC regulates expression of ribosomal genes and mitigates ER stress in the

  • Accordingly, a coordinated down-regulation of gene sets related to the digestive process and to protein metabolism and oxidative phosphorylation occurs in adult Nfic -/pancreata.
  • The mTOR pathway is a central actor in protein biosynthesis and autophagy and, therefore, a candidate mediator of this phenotype (reviewed in [33]).
  • The authors found reduced levels of P-RS6K1, its substrate P-RS6, and P-EIF4E, together with an up-regulation of P-ERK, in acinar cells of Nfic -/mice.
  • The finding that NFIC -but not NR5A2 -binds the promoter of ER stress genes suggests a distinct role of the former in this process.
  • A re-analysis of published data shows that Nfic is down-regulated in Nr5a2 -/hepatocytes in basal conditions (fold change of -0.54) 35 , suggesting that the deficient response to TM in Nr5a2 -/hepatocytes might partially occur through Nfic down-regulation.

NFIC is dynamically regulated during pancreatitis and cancer.

  • A failure to achieve complete acinar maturation is associated with more severe damage and delayed recovery during caerulein-mediated pancreatitis, as shown upon inactivation of Gata6, Mist1, and Ptf1a in the pancreas 4, 44 .
  • The role of NFIC in acinar cell differentiation and mitigation of ER stress suggested a contribution during tumorigenesis.
  • The copyright holder for this preprint this version posted August 9, 2021.
  • TFs previously described, the role of NFIC is restricted to the adult pancreas and distinctly affects RNA and protein metabolism and the UPR-mediated ER stress.

MATERIAL AND METHODS

  • All crosses were maintained in a predominant C57BL/6 background.
  • In brief, animals were weighed before the procedure and caerulein was administered intraperitoneally.
  • For the glucose tolerance test, male mice were fasted for 16 h and basal glycaemia was measured in tail blood.
  • For most experiments, >5 mice per group were used.

Histology, immunofluorescence (IF) and immunohistochemical (IHC) analyses.

  • Histological processing was performed using standard procedures.
  • After washing with PBS, sections were mounted with Prolong Gold Antifade Reagent (Life Technology).
  • Sections were incubated with 2% BSA-PBS for 1h at room temperature, and then with the primary antibody overnight at 4 ºC.
  • Briefly, areas of interest (AOI) were selected for quantification and then exported as individual TIFF images.
  • For quantification of KI67 + positive cells, at least 10 random images from each pancreas were selected and only positive acinar cells were quantified.

Quantitative RT-PCR (RT-qPCR).

  • Total RNA was treated with DNase I for 30 min at 37 °C and cDNAs were prepared according to the manufacturer's specifications, using the TaqMan reverse transcription reagents (Applied (which was not certified by peer review) is the author/funder.
  • For immunoprecipitation of proteins from fresh total pancreas lysates, a piece of mouse pancreas was isolated and minced in 50 mM Tris-HCl pH 8, 150 mM NaCl, 5 mM EDTA, 0.5% NP-40 containing 3× phosphatase inhibitor cocktail (Sigma-Aldrich) and 3× EDTA-free complete protease inhibitor cocktail .
  • Afterwards, beads were washed three times with coupling buffer and once with 1 M Tris pH 9.
  • Protein lysates (10-15 mg, tissues) were then incubated overnight at 4 °C with antibody-coated dynabeads (Thermo Fisher Scientific).

Chromatin immunoprecipitation (ChIP).

  • Pancreas tissue was minced, washed with cold PBS supplemented with 3× protease and phosphatase cocktail inhibitors, and then fixed with 1% formaldehyde for 20 min at room temperature.
  • The supernatant was collected after centrifugation and chromatin was sonicated with a Covaris instrument for 40 min (20% duty cycle; 10% intensity; 200 cycle), yielding DNA fragments with a bulk size of 300-500 bp.
  • Data were analysed using the nextpresso pipeline http://bioinfo.cnio.es/nextpresso/).

Gene Set Enrichment Analysis (GSEA).

  • A ranking metric [-log10(p value)/sign(log2FoldChange)] was used to generate a ranked gene list from the DEseq output.
  • The resulting libraries were sequenced on Illumina HiSeq 2500, v4 Chemistry.
  • The copyright holder for this preprint this version posted August 9, 2021.
  • Among the two replicates, the one with highest number of identified target genes was taken: replicate 1 of NFIC ChIP-Seq in GM12878, NFIC ChIP-Seq in HepG2 and NFIC ChIP-Seq in SK-N-SH and replicate 2 of NFIC ChIP-Seq in ECC1 cell line.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

NFIC regulates ribosomal biology and ER stress in pancreatic acinar cells
1
and suppresses PDAC initiation
2
3
4
Isidoro Cobo,
1,2
Sumit Paliwal,
1
Júlia Melià-Alomà,
1,3
Ariadna Torres,
1,3
5
Jaime Martínez-Villarreal,
1
Fernando García,
4
Irene Millán,
1,2
Natalia del Pozo,
1,2
Joo-
6
Cheol Park,
5
Ray J. MacDonald,
6
Javier Muñoz,
4
and Francisco X. Real
1-3
7
8
9
Short title: NFIC in pancreatic homeostasis and cancer
10
11
1
Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO,
12
Madrid, Spain
13
2
CIBERONC, Madrid, Spain
14
3
Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra,
15
Barcelona, Spain
16
4
Proteomics Unit, Spanish National Cancer Research Centre-CNIO, Madrid, Spain.
17
ProteoRed - ISCIII
18
5
Department of Oral Histology-Developmental Biology, School of Dentistry, Seoul
19
National University, Seoul, Korea
20
6
Department of Molecular Biology, University of Texas Southwestern Medical Center,
21
Dallas, TX, USA
22
23
24
25
Correspondence: Francisco X. Real
26
Centro Nacional de Investigaciones Oncológicas-CNIO
27
Melchor Fernández Almagro, 3
28
28029-Madrid, Spain
29
E-mail: preal@cnio.es
30
31
32
Conflicts of interest: none to declare
33
34
35
Funding: This work was supported, in part, by grants SAF2011-29530, SAF2015-70553-
36
R, and RTI2018-101071-B-I00 from Ministerio de Ciencia, Innovación y Universidades
37
(Madrid, Spain) (co-funded by the ERDF-EU) and RTICC from Instituto de Salud Carlos
38
III (RD12/0036/0034) to FXR. IC was recipient of a Beca de Formación del Personal
39
Investigador from Ministerio de Economía y Competitividad (Madrid, Spain). The
40
research leading to these results has received funding from People Programme (Marie
41
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 9, 2021. ; https://doi.org/10.1101/2021.08.09.455477doi: bioRxiv preprint

2
Curie Actions) of the European Union’s Seventh Framework Programme (FP7/2007-
42
2013) (REA grant agreement 608765”). SP was supported by a Juan de la Cierva
43
Programme fellowship from Ministerio de Ciencia, Innovación y Universidades. IM was
44
supported by a Fellowship from Fundació Bancaria La Caixa (ID 100010434) (grant
45
number LCF/BQ/ES18/11670009). CNIO is supported by Ministerio de Ciencia,
46
Innovación y Universidades as a Centro de Excelencia Severo Ochoa SEV-2015-0510.
47
48
Statement of author contributions
49
IC: study concept and design; acquisition of data; analysis and interpretation of data;
50
statistical analysis; drafting of the manuscript;
51
SP: acquisition of data; analysis and interpretation of data; drafting of the manuscript
52
JMA: acquisition of data; analysis and interpretation of data; drafting of the manuscript
53
AT: acquisition of data; analysis and interpretation of data;
54
JMV: analysis and interpretation of data;
55
FG: acquisition of data; analysis and interpretation of data;
56
IM: analysis and interpretation of data;
57
NdP: technical support and acquisition of data;
58
JCP: material support;
59
RJM: critical revision of the data and important intellectual content;
60
JM: acquisition of data; analysis and interpretation of data;
61
FXR: study concept and design; analysis and interpretation of data; drafting of the
62
manuscript; overall study supervision; obtained funding.
63
All authors provided input about manuscript content.
64
Accession numbers: RNA sequencing data have been deposited in GEO with
65
accession number GSE126907 and NFIC ChIP sequencing data have been deposited
66
in GEO with accession number GSE181098
67
68
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 9, 2021. ; https://doi.org/10.1101/2021.08.09.455477doi: bioRxiv preprint

3
ABSTRACT
69
70
Tissue-specific differentiation is driven by specialized transcriptional networks.
71
Pancreatic acinar cells crucially rely on the PTF1 complex, and on additional
72
transcription factors, to deploy their transcriptional program. Here, we identify NFIC as a
73
novel regulator of acinar differentiation using a variety of methodological strategies. NFIC
74
binding sites are found at very short distances from NR5A2-bound genomic regions and
75
both proteins co-occur in the same complex. Nfic knockout mice show reduced
76
expression of acinar genes and, in ChIP-seq experiments, NFIC binds the promoters of
77
acinar genes. In addition, NFIC binds to the promoter of, and regulates, genes involved
78
in RNA and protein metabolism; in Nfic knockout mice, p-RS6K1 and p-IEF4E are down-
79
regulated indicating reduced activity of the mTOR pathway. In 266-6 acinar cells, NFIC
80
dampens the ER stress program through its binding to ER stress gene promoters and is
81
required for complete resolution of Tunicamycin-mediated ER stress. Normal human
82
pancreata from subjects with low NFIC mRNA levels display reduced epxression of
83
genes down-regulated in Nfic knockout mice. Consistently, NFIC displays reduced
84
expression upon induced acute pancreatitis and is required for proper recovery after
85
damage. Finally, expression of NFIC is lower in samples of mouse and human pancreatic
86
ductal adenocarcinoma and Nfic knockout mice develop an increased number of mutant
87
Kras-driven pre-neoplastic lesions.
88
89
Word count: 211
90
91
Keywords: NFIC, pancreas, acinar differentiation, ribosome, endoplasmic reticulum
92
stress, unfolded protein response, transcriptional networks, pancreatitis, pancreatic
93
cancer
94
95
Abbreviations: ChIP, chromatin immunoprecipitation; DEG, differentially expressed
96
genes; EMT, epithelial-mesenchymal transition; ER, endoplasmic reticulum; GSEA,
97
Gene set enrichment analysis; IF, immunofluorescence; IHC; immunohistochemistry;
98
PDAC, pancreatic ductal adenocarcinoma; TF, transcription factor; TM, tunicamycin;
99
UPR, unfolded protein response.
100
101
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 9, 2021. ; https://doi.org/10.1101/2021.08.09.455477doi: bioRxiv preprint

4
INTRODUCTION
102
103
Pancreatic acinar cells are highly specialized protein synthesis factories that
104
have a well-developed rough endoplasmic reticulum (ER), a prominent Golgi complex,
105
and abundant secretory granules
1
. Acinar differentiation is contingent on the activity of a
106
master regulator, the adult PTF1 complex, composed of the pancreas-specific
107
transcription factors (TFs) PTF1A and RPBJL and the ubiquitous protein E47
2,3
. PTF1
108
binds the proximal promoter of genes coding for digestive enzymes, secretory proteins
109
and other TFs, and activates their expression. The PTF1 complex is the main driver of
110
acinar differentiation but additional TF with tissue-restricted expression patterns are
111
implicated in the fine-tuning of this process, including GATA6
4
, MIST1
5
, and
112
NR5A2/LRH-1
6,7
. Acinar cells play a crucial role in acute and chronic pancreatitis, two
113
common and disabling conditions. Recent work using genetic mouse models has shown
114
that, upon expression of mutant KRas, acinar cells can be the precursors of Pancreatic
115
Intraepithelial Neoplasia (PanIN) and pancreatic ductal adenocarcinoma (PDAC)
8,9
.
116
Our laboratory and others have shown that the acinar differentiation program acts
117
as a tumor suppressor in the pancreas. Monoallelic or homozygous inactivation of
118
several acinar transcriptional regulators in the germline, the embryonic pancreas, or the
119
adult pancreas can result in compromised acinar function that favors loss of cellular
120
identity and poises acinar cells for transformation upon activation of mutant KRas
10,11,12
.
121
The tumor suppressive function of these TF is not obvious because the exocrine
122
pancreas has a large functional reserve, i.e. massive alterations in cellular function need
123
to occur in order to be reflected in histological or clinical changes.
124
Here, we use bioinformatics tools to identify NFIC as a novel acinar regulator.
125
NFIC is a member of the nuclear factor I family of TFs that regulate both ubiquitous and
126
tissue-restricted genes
13
. In the mammary gland, NFIC activates the expression of milk
127
genes involved in lactation
14
. Furthermore, it acts as a breast cancer tumor suppressor,
128
as it directly represses the expression of Ccnd1 and Foxf1, a potent inducer of epithelial-
129
mesenchymal transition (EMT), invasiveness, and tumorigenicity. Additional roles have
130
been proposed through the regulation of Trp53
15,16,17
. The physiological role of NFIC has
131
been best studied in dentinogenesis, since Nfic
-/-
mice develop short molar roots and
132
display aberrant odontoblast differentiation and dentin formation
18
. NFIC regulates
133
odontoblast-related genes, including Dssp
19
, Wnt
20
, and hedgehog signaling
21
.
134
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 9, 2021. ; https://doi.org/10.1101/2021.08.09.455477doi: bioRxiv preprint

5
Using a combination of omics analyses and studies in knockout mice and cultured
135
cells, we now uncover novel roles of NFIC as a regulator of acinar function whose major
136
impact is at the level of the ER stress response in murine and human pancreas. Unlike
137
most other TFs previously identified as required for full acinar function, NFIC belongs to
138
a novel family of acinar regulators with tissue-wide expression. NFIC dysregulation
139
sensitizes the pancreas to damage and neoplastic transformation.
140
141
142
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 9, 2021. ; https://doi.org/10.1101/2021.08.09.455477doi: bioRxiv preprint

Citations
More filters
Journal ArticleDOI
TL;DR: In this article , gene expression analysis from 2,000 individually micro-dissected ductal lesions representing 145 patients was performed to identify early processes and potential biomarkers, including CAMK2N1 , MNX1 , ADCY5 , HOXC11 and ANKRD22 , whose reduced expression is associated with the progression of DCIS to invasive breast cancer.
Abstract: Abstract Ductal carcinoma in situ (DCIS) is considered a non-invasive precursor to breast cancer, and although associated with an increased risk of developing invasive disease, many women with DCIS will never progress beyond their in situ diagnosis. The path from normal duct to invasive ductal carcinoma (IDC) is not well understood, and efforts to do so are hampered by the substantial heterogeneity that exists between patients, and even within patients. Here we show gene expression analysis from > 2,000 individually micro-dissected ductal lesions representing 145 patients. Combining all samples into one continuous trajectory we show there is a progressive loss in basal layer integrity heading towards IDC, coupled with two epithelial to mesenchymal transitions, one early and a second coinciding with the convergence of DCIS and IDC expression profiles. We identify early processes and potential biomarkers, including CAMK2N1 , MNX1 , ADCY5 , HOXC11 and ANKRD22 , whose reduced expression is associated with the progression of DCIS to invasive breast cancer.

10 citations

Journal ArticleDOI
TL;DR: This work provides new insights into the dynamics of activation of NPSCs in vivo in response to T3 during a critical period of neurogenesis in the metamorphosis, suggesting that NSPCs activation induced by T3 is characterized by an early proteome remodeling through the synthesis of the translation machinery and the degradation of proteins by the proteasome.
Abstract: The thyroid hormones—thyroxine (T4) and 3,5,3′triiodothyronine (T3)—regulate the development of the central nervous system (CNS) in vertebrates by acting in different cell types. Although several T3 target genes have been identified in the brain, the changes in the transcriptome in response to T3 specifically in neural stem and progenitor cells (NSPCs) during the early steps of NSPCs activation and neurogenesis have not been studied in vivo. Here, we characterized the transcriptome of FACS‐sorted NSPCs in response to T3 during Xenopus laevis metamorphosis.

2 citations

Posted ContentDOI
04 Mar 2022-bioRxiv
TL;DR: A ‘Timeline’ of disease progression is generated, utilising the variability within patients and combining >2,000 individually micro-dissected ductal lesions from 145 patients into one continuous trajectory, showing there is a progressive loss in basal layer integrity, coupled with two epithelial to mesenchymal transitions (EMT) early in the timeline.
Abstract: Ductal carcinoma in situ (DCIS) is considered a non-invasive precursor to breast cancer, and although associated with an increased risk of developing invasive disease, many women with DCIS will never progress beyond their in situ diagnosis. The path from normal duct to invasive disease is not well understood, and efforts to do so are hampered by the substantial heterogeneity that exists between patients and even within patients. Using gene expression analysis, we have generated a ‘Timeline’ of disease progression, utilising the variability within patients and combining >2,000 individually micro-dissected ductal lesions from 145 patients into one continuous trajectory. Using this Timeline we show there is a progressive loss in basal layer integrity, coupled with two epithelial to mesenchymal transitions (EMT), one early in the timeline and a second just prior to cells leaving the duct. We identify early processes and potential biomarkers, including CAMK2N1, MNX1, ADCY5, HOXC11 and ANKRD22, whose reduced expression is associated with the progression of DCIS to invasive breast cancer.
References
More filters
Journal ArticleDOI
15 Apr 2014-eLife
TL;DR: It is concluded that LRH-1 initiates a novel pathway of ER stress resolution that is independent of the UPR, yet equivalently required, and may be beneficial in human disorders associated with chronic ER stress.
Abstract: Chronic endoplasmic reticulum (ER) stress results in toxicity that contributes to multiple human disorders. We report a stress resolution pathway initiated by the nuclear receptor LRH-1 that is independent of known unfolded protein response (UPR) pathways. Like mice lacking primary UPR components, hepatic Lrh-1-null mice cannot resolve ER stress, despite a functional UPR. In response to ER stress, LRH-1 induces expression of the kinase Plk3, which phosphorylates and activates the transcription factor ATF2. Plk3-null mice also cannot resolve ER stress, and restoring Plk3 expression in Lrh-1-null cells rescues ER stress resolution. Reduced or heightened ATF2 activity also sensitizes or desensitizes cells to ER stress, respectively. LRH-1 agonist treatment increases ER stress resistance and decreases cell death. We conclude that LRH-1 initiates a novel pathway of ER stress resolution that is independent of the UPR, yet equivalently required. Targeting LRH-1 may be beneficial in human disorders associated with chronic ER stress.

62 citations

Journal ArticleDOI
TL;DR: A novel pathway in breast cancer that regulates epithelial-to-mesenchymal transition (EMT), motility, and invasiveness is described and the first evidence for a role of FoxF1 in cancer and in the regulation of EMT in cells of epithelial origin is provided.
Abstract: Progression to metastasis is the proximal cause of most cancer-related mortality. Yet much remains to be understood about what determines the spread of tumor cells. This paper describes a novel pathway in breast cancer that regulates epithelial-to-mesenchymal transition (EMT), motility, and invasiveness. We identify two transcription factors, nuclear factor 1-C2 (NF1-C2) and Forkhead box F1 (FoxF1), downstream of prolactin/nuclear Janus-activated kinase 2, with opposite effects on these processes. We show that NF1-C2 is lost during mammary tumor progression and is almost invariably absent from lymph node metastases. NF1-C2 levels in primary tumors correlate with better patient survival. Manipulation of NF1-C2 levels by expression of a stabilized version or using small interfering RNA showed that NF1-C2 counteracts EMT, motility, invasiveness, and tumor growth. FoxF1 was found to be a direct repressed target of NF1-C2. We provide the first evidence for a role of FoxF1 in cancer and in the regulation of EMT in cells of epithelial origin. Overexpression of FoxF1 was associated with a mesenchymal phenotype, increased invasiveness in vitro, and enhanced growth of breast carcinoma xenografts in nude mice. The relevance of these findings is strengthened by the correlation between FoxF1 expression and a mesenchymal phenoype in breast cancer cell isolates, consistent with the interpretation that FoxF1 promotes invasion and metastasis.

60 citations

Journal ArticleDOI
01 Aug 2012-Gut
TL;DR: It was found that Hnf1α binds to, and regulates, the promoter of Nr5a2, coding an orphan nuclear receptor that regulates acinar gene expression, which uncovers roles for Hn f1α in the regulation of acinar cell determination and function.
Abstract: Objectives During pancreatitis, specific transcriptional programmes govern functional regeneration after injury. The objective of this study was to analyse the dynamic regulation of pancreatic genes and the role of transcriptional regulators during recovery from pancreatitis. Design Wild-type and genetically modified mice (Hnf1α −/− and Ptf1a +/− ) were used. After caerulein or l-arginine induced pancreatitis, blood or pancreata were processed for enzymatic assays, ELISA, histology, immunohistochemistry, western blotting and quantitative reverse transcriptase-PCR. Nr5a2 promoter reporter and chromatin immunoprecipitation assays for Hnf1α were also performed. Results After caerulein pancreatic injury, expression of acinar and endocrine genes rapidly decreased, but eventually recovered, depicting distinct cell-type-specific patterns. Pdx1 and Hnf1α mRNAs underwent marked downregulation, matching endocrine/exocrine gene expression profiles. Ptf1a, Pdx1 and Hnf1α protein levels were also reduced and recovered gradually. These changes were associated with transient impairment of exocrine and endocrine function, including abnormal glucose tolerance. On l-arginine pancreatitis, changes in Ptf1a, Pdx1 and Hnf1α gene and protein expression were recapitulated. Reduced Hnf1α and Ptf1a levels after pancreatitis coincided with increased acinar cell proliferation, both in Hnf1α −/− and Ptf1a +/− mice. Moreover, Hnf1α −/− mice had reduced Ptf1a protein as well as transcripts for Ptf1a and digestive enzymes. Dispersed acini from Hnf1α −/− mice showed suboptimal secretory responses to caerulein. Bioinformatics analysis did not support a role for Hnf1α as a direct regulator of digestive enzyme genes. Instead, it was found that Hnf1α binds to, and regulates, the promoter of Nr5a2 , coding an orphan nuclear receptor that regulates acinar gene expression. Conclusions Dynamic changes in gene expression occur on pancreatitis induction, determining altered exocrine and endocrine function. This analysis uncovers roles for Hnf1α in the regulation of acinar cell determination and function. This effect may be mediated, in part, through direct regulation of Nr5a2 .

47 citations

Journal ArticleDOI
TL;DR: Retinoids regulate exocrine lineage selection through epithelial-mesenchyme interactions, mediated through up-regulation of mesenchymal laminin-1.

41 citations

Journal ArticleDOI
TL;DR: A novel pathway in breast cancer that regulates epithelial-to-mesenchymal transition (EMT), motility, and invasiveness is described: the NFI-C-KLF4-E-cadherin pathway.
Abstract: Progression to metastasis is the leading cause of most cancer-related mortality; however, much remains to be understood about what facilitates the spread of tumor cells. In the present study, we describe a novel pathway in breast cancer that regulates epithelial-to-mesenchymal transition (EMT), motility, and invasiveness. We examined nuclear factor I-C (NFI-C) expression in MCF10A human breast epithelial cells, MCF7 non-invasive breast cancer cells, and MDA-MB231 invasive breast cancer cells by real-time PCR and western blotting. To investigate the loss- and gain-function of NFI-C, we determined whether NFI-C regulated KLF4 expression by real-time PCR, western blotting, and promoter assay. To understand the biological functions of NFI-C, we observed cell invasion, migration, adhesion in human tumor cells by transwell assay, wound healing assay, quantitative RT-PCR, cell adhesion assay, western blotting, and immunohistochemistry. We identified the downstream factors of NFI-C, such as KLF4 and E-cadherin, which play roles in EMT. NFI-C is expressed in normal mammary gland or noninvasive breast cancer cells with epithelial characteristics. NFI-C overexpression induced expression of KLF4 and E-cadherin, but not Slug, in breast cancer cells. NFI-C bound directly to the KLF4 promoter and stimulated KLF4 transcriptional activity, thereby regulating E-cadherin expression during tumorigenesis. Cells overexpressing NFI-C maintained their epithelial differentiation status, which could drive mesenchymal-epithelial transition (MET) via the NFI-C-KLF4-E-cadherin axis in breast cancer cells. Consequently, NFI-C suppressed EMT, migration, and invasion in breast cancer cells. Our study reveals a novel signaling pathway that is important during breast cancer tumorigenesis: the NFI-C-KLF4-E-cadherin pathway. The results indicate the important role of NFI-C in regulating KLF4 during tumorigenesis.

40 citations

Related Papers (5)