1
|
Chen YH, Zhang X, Chou CH, Hsueh MF, Attarian D, Li YJ, Kraus VB. Association of Dipeptidylpeptidase 4 (CD26) With Chondrocyte Senescence and Radiographic Progression in Knee Osteoarthritis. Arthritis Rheumatol 2023; 75:1120-1131. [PMID: 36704903 PMCID: PMC10313751 DOI: 10.1002/art.42455] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 12/01/2022] [Accepted: 03/07/2023] [Indexed: 01/28/2023]
Abstract
OBJECTIVE To evaluate the association of dipeptidylpeptidase 4 (DPP-4; also known as CD26) with cellular senescence of human cartilage and progression of knee osteoarthritis (OA). METHODS Articular cartilage sections and chondrocytes were acquired from 35 individuals undergoing total knee replacement for OA to evaluate the following: 1) the association between OA severity and established senescence markers (senescence-associated β-galactosidase activity and p16), which was quantified using immunohistochemistry and flow cytometry (n = 19 samples); 2) the coexpression of DPP-4 with established senescence markers, which was assessed using flow cytometry; and 3) expression levels of anabolic and catabolic genes, senescence-related genes, and senescence-associated secretory phenotypes in DPP-4+ and DPP-4- cells, which were isolated using fluorescence-activated cell sorting or magnetic-activated cell sorting (n = 16 samples). The concentration of soluble DPP-4 was measured in samples of synovial fluid and samples of plasma from the Prediction of Osteoarthritis Progression cohort and then evaluated for association with the severity of radiographic knee OA at baseline (n = 65 samples) and the progression of structural radiographic OA (n = 57 samples) over a 3-year period. RESULTS DPP-4 expression was associated with higher senescence-associated β-galactosidase activity, p16 expression, senescence-related gene and catabolic gene (ADAMTS5, MMP13, IL6, and IL8) expression, higher senescence-associated secretory phenotype secretion, and lower anabolic gene (COL2A1 and ACAN) expression in primary chondrocytes. Synovial fluid DPP-4 concentration was associated with radiographic OA progression (odds ratio 105.32; P = 0.015), proteases (synovial fluid matrix metalloproteinase 1 and matrix metalloproteinase 3), aggrecan degradation (synovial fluid sulfated glycosaminoglycan), indicators of activated macrophages (synovial fluid CD14 and CD163), and inflammation (synovial fluid interleukin-6). CONCLUSION Our study identifies DPP-4 as a key surface marker in senescent chondrocytes and a predictor of radiographic OA progression.
Collapse
Affiliation(s)
- Yu-Hsiu Chen
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
- Division of Rheumatology/Immunology/Allergy, Department of Internal Medicine Tri-Service General Hospital, National Defense Medical Center, Taiwan
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Xin Zhang
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Ching-Heng Chou
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Ming-Feng Hsueh
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - David Attarian
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Yi-Ju Li
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Virginia Byers Kraus
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
2
|
Moffitt LR, Bilandzic M, Wilson AL, Chen Y, Gorrell MD, Oehler MK, Plebanski M, Stephens AN. Hypoxia Regulates DPP4 Expression, Proteolytic Inactivation, and Shedding from Ovarian Cancer Cells. Int J Mol Sci 2020; 21:8110. [PMID: 33143089 PMCID: PMC7672561 DOI: 10.3390/ijms21218110] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 10/28/2020] [Indexed: 01/03/2023] Open
Abstract
The treatment of ovarian cancer has not significantly changed in decades and it remains one of the most lethal malignancies in women. The serine protease dipeptidyl peptidase 4 (DPP4) plays key roles in metabolism and immunity, and its expression has been associated with either pro- or anti-tumour effects in multiple tumour types. In this study, we provide the first evidence that DPP4 expression and enzyme activity are uncoupled under hypoxic conditions in ovarian cancer cells. Whilst we identified strong up-regulation of DPP4 mRNA expression under hypoxic growth, the specific activity of secreted DPP4 was paradoxically decreased. Further investigation revealed matrix metalloproteinases (MMP)-dependent inactivation and proteolytic shedding of DPP4 from the cell surface, mediated by at least MMP10 and MMP13. This is the first report of uncoupled DPP4 expression and activity in ovarian cancer cells, and suggests a previously unrecognized, cell- and tissue-type-dependent mechanism for the regulation of DPP4 in solid tumours. Further studies are necessary to identify the functional consequences of DPP4 processing and its potential prognostic or therapeutic value.
Collapse
Affiliation(s)
- Laura R. Moffitt
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Maree Bilandzic
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Amy L. Wilson
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Yiqian Chen
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Mark D. Gorrell
- Centenary Institute, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Martin K. Oehler
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia;
- Robinson Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia;
| | - Andrew N. Stephens
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| |
Collapse
|
3
|
Fallah S, Beaulieu JF. The Hippo Pathway Effector YAP1 Regulates Intestinal Epithelial Cell Differentiation. Cells 2020; 9:1895. [PMID: 32823612 PMCID: PMC7463744 DOI: 10.3390/cells9081895] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 07/31/2020] [Accepted: 08/11/2020] [Indexed: 12/15/2022] Open
Abstract
The human intestine is covered by epithelium, which is continuously replaced by new cells provided by stem cells located at the bottom of the glands. The maintenance of intestinal stem cells is supported by a niche which is composed of several signaling proteins including the Hippo pathway effectors YAP1/TAZ. The role of YAP1/TAZ in cell proliferation and regeneration is well documented but their involvement on the differentiation of intestinal epithelial cells is unclear. In the present study, the role of YAP1/TAZ on the differentiation of intestinal epithelial cells was investigated using the HT29 cell line, the only multipotent intestinal cell line available, with a combination of knockdown approaches. The expression of intestinal differentiation cell markers was tested by qPCR, Western blot, indirect immunofluorescence and electron microscopy analyses. The results show that TAZ is not expressed while the abolition of YAP1 expression led to a sharp increase in goblet and absorptive cell differentiation and reduction of some stem cell markers. Further studies using double knockdown experiments revealed that most of these effects resulting from YAP1 abolition are mediated by CDX2, a key intestinal cell transcription factor. In conclusion, our results indicate that YAP1/TAZ negatively regulate the differentiation of intestinal epithelial cells through the inhibition of CDX2 expression.
Collapse
Affiliation(s)
- Sepideh Fallah
- Laboratory of Intestinal Physiopathology, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
4
|
Kim KM, Noh JH, Bodogai M, Martindale JL, Yang X, Indig FE, Basu SK, Ohnuma K, Morimoto C, Johnson PF, Biragyn A, Abdelmohsen K, Gorospe M. Identification of senescent cell surface targetable protein DPP4. Genes Dev 2017; 31:1529-1534. [PMID: 28877934 PMCID: PMC5630018 DOI: 10.1101/gad.302570.117] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/02/2017] [Indexed: 12/13/2022]
Abstract
Kim et al. show that DPP4 (dipeptidyl peptidase 4) was selectively expressed on the surface of senescent human diploid fibroblasts and that this enabled their preferential elimination. Senescent cell accumulation in aging tissues is linked to age-associated diseases and declining function, prompting efforts to eliminate them. Mass spectrometry analysis revealed that DPP4 (dipeptidyl peptidase 4) was selectively expressed on the surface of senescent, but not proliferating, human diploid fibroblasts. Importantly, the differential presence of DPP4 allowed flow cytometry-mediated isolation of senescent cells using anti-DPP4 antibodies. Moreover, antibody-dependent cell-mediated cytotoxicity (ADCC) assays revealed that the cell surface DPP4 preferentially sensitized senescent, but not dividing, fibroblasts to cytotoxicity by natural killer cells. In sum, the selective expression of DPP4 on the surface of senescent cells enables their preferential elimination.
Collapse
Affiliation(s)
- Kyoung Mi Kim
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Ji Heon Noh
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Monica Bodogai
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Jennifer L Martindale
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Xiaoling Yang
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Fred E Indig
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Sandip K Basu
- National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, USA
| | - Kei Ohnuma
- Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Chikao Morimoto
- Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Peter F Johnson
- National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, USA
| | - Arya Biragyn
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Kotb Abdelmohsen
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Myriam Gorospe
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| |
Collapse
|
5
|
Gong Q, Rajagopalan S, Zhong J. Dpp4 inhibition as a therapeutic strategy in cardiometabolic disease: Incretin-dependent and -independent function. Int J Cardiol 2015; 197:170-179. [PMID: 26142202 PMCID: PMC7114201 DOI: 10.1016/j.ijcard.2015.06.076] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/03/2015] [Accepted: 06/20/2015] [Indexed: 12/25/2022]
Abstract
Cardiometabolic disorders including obesity, diabetes and cardiovascular disease are among the most severe health problems worldwide. DPP4 enzymatic inhibitors were first developed as anti-diabetic reagents which preserve incretin hormones and promote post-prandial insulin secretion. It's been shown in animal studies that incretin-based therapy has a beneficial effect on cardiovascular disease. Recent studies demonstrated novel non-catalytic functions of DPP4 that may play a role in cardiometabolic disease. Although the role of DPP4 inhibition-mediated incretin effects has been well-reviewed, little information of its incretin-independent actions was introduced in cardiometabolic disease. In the current review, we will summarize the catalytic dependent and independent effects of DPP4 inhibition on cardiometabolic disease.
Collapse
Affiliation(s)
- Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei 434023, PR China
| | - Sanjay Rajagopalan
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Jixin Zhong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei 434023, PR China; Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
6
|
Brial F, Lussier CR, Belleville K, Sarret P, Boudreau F. Ghrelin Inhibition Restores Glucose Homeostasis in Hepatocyte Nuclear Factor-1α (MODY3)-Deficient Mice. Diabetes 2015; 64:3314-20. [PMID: 25979074 DOI: 10.2337/db15-0124] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 05/11/2015] [Indexed: 11/13/2022]
Abstract
Hepatocyte nuclear factor-1α (HNF1α) is a transcription factor expressed in tissues of endoderm origin. Mutations in HNF1A are associated with maturity-onset diabetes of the young 3 (MODY3). Mice deficient for Hnf1α are hyperglycemic, with their pancreatic β-cells being defective in glucose-sensing insulin secretion. The specific mechanisms involved in this defect are unclear. Gut hormones control glucose homeostasis. Our objective was to explore whether changes in these hormones play a role in glucose homeostasis in the absence of Hnf1α. An increase in ghrelin gene transcript and a decrease in glucose-dependent insulinotropic polypeptide (GIP) gene transcripts were observed in the gut of Hnf1α-null mice. These changes correlated with an increase of ghrelin and a decrease of GIP-labeled cells. Ghrelin serological levels were significantly induced in Hnf1α-null mice. Paradoxically, GIP levels were also induced in these mice. Treatment of Hnf1α-null mice with a ghrelin antagonist led to a recovery of the diabetic symptoms. We conclude that upregulation of ghrelin in the absence of Hnf1α impairs insulin secretion and can be reversed by pharmacological inhibition of ghrelin/GHS-R interaction. These observations open up on future strategies to counteract ghrelin action in a program that could become beneficial in controlling non-insulin-dependent diabetes.
Collapse
Affiliation(s)
- François Brial
- Department of Anatomy and Cell Biology, Université de Sherbrooke, Quebec, Canada
| | - Carine R Lussier
- Department of Anatomy and Cell Biology, Université de Sherbrooke, Quebec, Canada
| | - Karine Belleville
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Quebec, Canada
| | - Philippe Sarret
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Quebec, Canada
| | - François Boudreau
- Department of Anatomy and Cell Biology, Université de Sherbrooke, Quebec, Canada
| |
Collapse
|
7
|
Zhong J, Gong Q, Goud A, Srinivasamaharaj S, Rajagopalan S. Recent Advances in Dipeptidyl-Peptidase-4 Inhibition Therapy: Lessons from the Bench and Clinical Trials. J Diabetes Res 2015; 2015:606031. [PMID: 26075284 PMCID: PMC4446505 DOI: 10.1155/2015/606031] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/23/2015] [Accepted: 04/27/2015] [Indexed: 12/23/2022] Open
Abstract
DPP4 inhibitors (DPP4i) are a class of newly developed antidiabetic drugs which preserve incretin hormones and promote postprandial insulin secretion. Although the cardiovascular effect of DPP4 inhibition has been substantially studied, the exact role of DPP4 in cardiovascular disease especially in humans remains elusive. Previous small studies and meta-analyses have suggested a benefit in both surrogate outcomes and cardiovascular events for these agents. However, there was growing evidence in recent years questioning the cardioprotective effect of DPP4i. Further, a signal of heart failure hospitalization in a recent large scale clinical trial SAVOR-TIMI 53 has called into question the safety of these agents and their utility in the treatment of cardiovascular disease. In this review, we will revisit the physiologic function of DPP4 and discuss its role in cardiometabolic disease based on recent experimental and clinical studies.
Collapse
Affiliation(s)
- Jixin Zhong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei 434023, China
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei 434023, China
| | - Aditya Goud
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Srividya Srinivasamaharaj
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sanjay Rajagopalan
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
8
|
Varamini P, Hussein WM, Mansfeld FM, Toth I. Synthesis, biological activity and structure–activity relationship of endomorphin-1/substance P derivatives. Bioorg Med Chem 2012; 20:6335-43. [DOI: 10.1016/j.bmc.2012.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 08/24/2012] [Accepted: 09/01/2012] [Indexed: 10/27/2022]
|
9
|
Abe M, Havre PA, Urasaki Y, Ohnuma K, Morimoto C, Dang LH, Dang NH. Mechanisms of confluence-dependent expression of CD26 in colon cancer cell lines. BMC Cancer 2011; 11:51. [PMID: 21284881 PMCID: PMC3038146 DOI: 10.1186/1471-2407-11-51] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 02/01/2011] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND CD26 (dipeptidyl peptidase IV, DPPIV) is a 110 kDa surface glycoprotein expressed in most normal tissues, and is a potential novel therapeutic target for selected cancers. Our work evaluates the mechanism involved in confluence-dependent CD26 expression in colon cancer. METHODS Colon adenocarcinoma cells were grown to confluence, and expression of CD26 and transcription factors implicated in its regulation was confirmed by immunofluorescence and Western blotting. Real-time PCR was also performed to evaluate CD26 upregulation at the transcriptional level. The influence of c-Myc on CD26 expression during different growth conditions was further evaluated following transient transfection of a c-Myc-expressing plasmid and a c-Myc specific siRNA. RESULTS We found that the colon cancer cell lines HCT-116 and HCT-15 exhibited a confluence-dependent increase in CD26 mRNA and protein, associated with decreased expression of c-Myc, increased USF-1 and Cdx 2 levels, and unchanged HNF-1α expression. Meanwhile, ectopic expression of c-Myc in both cell lines led to decreased CD26 expression. In contrast, transfection of a siRNA targeted to Cdx2 resulted in decreased CD26 level. Importantly, culturing of cells in serum-depleted media, but not acidic conditions, upregulated CD26. While HIF-1α level also increased when cells were cultured in serum-depleted media, its expression was required but not sufficient for CD26 upregulation. CONCLUSIONS CD26 mRNA and protein levels increase in a confluence-dependent manner in colon carcinoma cell lines, with c-Myc acting as a repressor and Cdx2 acting as an enhancer of CD26 expression. The enhanced expression of CD26 in serum-depleted media and a requirement for HIF-1α suggest a role for nutrients or growth factors in the regulation of CD26 protein expression.
Collapse
Affiliation(s)
- Masako Abe
- Department of Hematologic Malignancies, Nevada Cancer Institute, Las Vegas, Nevada, 89135, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
Liu H, Ren H, Spear BT. The mouse alpha-albumin (afamin) promoter is differentially regulated by hepatocyte nuclear factor 1α and hepatocyte nuclear factor 1β. DNA Cell Biol 2010; 30:137-47. [PMID: 20979532 DOI: 10.1089/dna.2010.1097] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Alpha-albumin (AFM), a member of the albumin gene family that also includes albumin, alpha-fetoprotein, and vitamin D-binding protein, is expressed predominantly in the liver and activated at birth. Here, we identify two hepatocyte nuclear factor 1 (HNF1)-binding sites in the AFM promoter that are highly conserved in different mammals. These two sites bind HNF1α and HNF1β. The distal site (centered at -132, relative to AFM exon 1) is more important than proximal site (centered at -58), based on HNF1 binding and mutational analysis in transfected cells. Our data indicate that HNF1α is a more potent activator of AFM promoter than is HNF1β. However, HNF1β can act in a dominant manner to inhibit HNF1α-dependent transactivation of the AFM promoter when both proteins are expressed together. This suggests that the differential timing with which the albumin family genes are activated in the liver may be influenced by their responsiveness to HNF1α and HNF1β. Our comparison of HNF1-binding sites in the promoters in the albumin family of genes indicates that the primordial albumin-like gene contained two HNF1 sites; one of these sites was lost from the albumin promoter, but both sites still are present in other members of this gene family.
Collapse
Affiliation(s)
- Hua Liu
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | |
Collapse
|
11
|
D'Angelo A, Bluteau O, Garcia-Gonzalez MA, Gresh L, Doyen A, Garbay S, Robine S, Pontoglio M. Hepatocyte nuclear factor 1alpha and beta control terminal differentiation and cell fate commitment in the gut epithelium. Development 2010; 137:1573-82. [PMID: 20388655 DOI: 10.1242/dev.044420] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The intestinal epithelium is a complex system characterized by massive and continuous cell renewal and differentiation. In this context, cell-type-specific transcription factors are thought to play a crucial role by modulating specific transcription networks and signalling pathways. Hnf1alpha and beta are closely related atypical homeoprotein transcription factors expressed in several epithelia, including the gut. With the use of a conditional inactivation system, we generated mice in which Hnf1b is specifically inactivated in the intestinal epithelium on a wild-type or Hnf1a(-/-) genetic background. Whereas the inactivation of Hnf1a or Hnf1b alone did not lead to any major intestinal dysfunction, the concomitant inactivation of both genes resulted in a lethal phenotype. Double-mutant animals had defective differentiation and cell fate commitment. The expression levels of markers of all the differentiated cell types, both enterocytes and secretory cells, were affected. In addition, the number of goblet cells was increased, whereas mature Paneth cells were missing. At the molecular level, we show that Hnf1alpha and beta act upstream of the Notch pathway controlling directly the expression of two crucial components: Jag1 and Atoh1. We demonstrate that the double-mutant mice present with a defect in intestinal water absorption and that Hnf1alpha and beta directly control the expression of Slc26a3, a gene whose mutations are associated with chloride diarrhoea in human patients. Our study identifies new direct target genes of the Hnf1 transcription factors and shows that they play crucial roles in both defining cell fate and controlling terminal functions in the gut epithelium.
Collapse
Affiliation(s)
- Anna D'Angelo
- Expression Génique, Développement et Maladies Equipe 26/ INSERM U567/ CNRS UMR 8104 / Université Paris-Descartes Institut Cochin Dpt. Génétique et Développement, 75014 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Borlak J, Zwadlo C. Expression of drug-metabolizing enzymes, nuclear transcription factors and ABC transporters in Caco-2 cells. Xenobiotica 2008; 33:927-43. [PMID: 14514442 DOI: 10.1080/00498250310001614286] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
1. Caco-2 cells are frequently used in intestinal drug absorption and metabolism studies, but little is known about the effects of drugs on the simultaneous expression of genes coding for drug-metabolizing enzymes (DMEs), nuclear transcription factors and ABC transporters. 2. The gene expression and enzyme activities of control and Aroclor 1254-treated cultures were therefore explored, the latter being a powerful inducer of DMEs. Fourteen- and 80-fold induction of CYP1A1 and CYP1A2 mRNA were shown, whereas expression of other DMEs was either increased (CYP2C8-2C19, 10-fold; CYP3A5, twofold; FMO1, 2 and 5, twofold; epoxide hydrolase, threefold) or repressed (CYP2D6 and CYP2E1 to 75% of control values). 3. Notably, gene copies of CYP3A4 and CYP2B6/7 were below the limit of detection, but a three- and 10-fold induction of HNF 1alpha + beta, HNF-4alpha4 and a similar 10-fold increase in STAT 3 and 4 was observed. 4. Similarly, c/EBP transcripts were only detected in treated cell cultures, but MRP1, its isoforms 3-5 as well as MDR-1 were increased threefold after dosing with Aroclor 1254. 5. Overall, CYP gene expression correlated well with the cognate enzyme activity using testosterone as a marker substrate.
Collapse
Affiliation(s)
- J Borlak
- Fraunhofer Institute of Toxicology and Experimental Medicine, Center for Drug Research and Medical Biotechnology, Nicolai-Fuchs-Str. 1 D-30659, Hannover, Germany.
| | | |
Collapse
|
13
|
Gu N, Tsuda M, Matsunaga T, Adachi T, Yasuda K, Ishihara A, Tsuda K. Glucose regulation of dipeptidyl peptidase IV gene expression is mediated by hepatocyte nuclear factor-1alpha in epithelial intestinal cells. Clin Exp Pharmacol Physiol 2008; 35:1433-9. [PMID: 18671716 DOI: 10.1111/j.1440-1681.2008.05015.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
1. Dipeptidyl peptidase IV (DPP-IV) is a new drug target in the treatment of Type 2 diabetes. Dipeptidyl peptidase IV enzyme activity is significantly altered in Type 2 diabetic patients with hyperglycaemia, but the underlying molecular mechanisms remain unclear. 2. The first aim of the present study was to clarify whether glucose regulates DPP-IV enzyme activity. To address this, DPP-IV gene expression and enzyme activity were measured in Caco2 cells cultured in the presence of low (2.5 mmol/L) or high (16.7 mmol/L) concentrations of glucose. We observed that high glucose inhibited DPP-IV gene expression and enzyme activity. 3. The second aim of the present study was to investigate whether hepatocyte nuclear factor (HNF)-1alpha contributes to glucose regulation of DPP-IV gene expression. To explore this question, associations between the gene expression of DPP-IV and HNF-1alpha were examined in Caco-2 cells cultured in the presence of low (2.5 mmol/L) or high (16.7 mmol/L) glucose. We found that the pattern of glucose-regulated DPP-IV gene expression is similar to that of HNF-1alpha. Moreover, to elucidate whether glucose regulation of DPP-IV gene expression is affected when HNF-1alpha is inhibited, we produced two stable cell lines in which a dominant-negative mutant HNF-1alphaR271G or basic vectors were stably expressed. We found that glucose regulation of DPP-IV gene expression was compromised in HNF-1alphaR271G cells, but was well maintained in basic vector cells. 4. These results suggest that glucose regulation of DPP-IV gene expression is mediated by HNF-1alpha.
Collapse
Affiliation(s)
- Ning Gu
- Laboratory of Metabolism, Graduate School of Human and Environmental Studies, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
14
|
Koda Y, Shiotani K, Toth I, Tsuda Y, Okada Y, Blanchfield JT. Comparison of the in vitro apparent permeability and stability of opioid mimetic compounds with that of the native peptide. Bioorg Med Chem Lett 2007; 17:2043-6. [PMID: 17300932 DOI: 10.1016/j.bmcl.2007.01.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2006] [Revised: 12/30/2006] [Accepted: 01/05/2007] [Indexed: 11/28/2022]
Abstract
Three dimethyl-L-tyrosine (Dmt) based peptide analogues were identified in a previous study as excellent agonists for the mu-opioid receptor showing very low K(i) values and good in vivo antinociceptive activity upon intracerebroventricular administration to mice. This activity decreased markedly when the compounds were delivered subcutaneously or orally. To establish the cause of this decrease of activity the apparent permeability across Caco-2 cell monolayers of each compound and their relative stability to the digestive enzymes present in the cell line has been determined and compared to that of the native peptide endomorphin 2. The compounds' permeabilities clearly correlate with their increasing lipophilicity suggesting that the analogues cross the monolayer via passive diffusion and the results show that the compound with high K(i) value for the mu-receptor (K(i)mu=0.114 nM) exhibited the highest permeability suggesting that this may be the better lead compound despite the lower binding affinity than that of compound 2 or 3.
Collapse
Affiliation(s)
- Yasuko Koda
- School of Pharmacy, University of Queensland, St. Lucia, Qld 4072, Australia
| | | | | | | | | | | |
Collapse
|
15
|
Ng L, Nichols K, O'Rourke K, Maslen A, Kirby GM. Repression of human GSTA1 by interleukin-1beta is mediated by variant hepatic nuclear factor-1C. Mol Pharmacol 2007; 71:201-8. [PMID: 17021248 DOI: 10.1124/mol.106.028563] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Down-regulation of glutathione transferase A1 (GSTA1) expression has profound implications in cytoprotection against toxic by-products of lipid peroxidation produced during inflammation. We investigated the role of hepatic nuclear factor 1 (HNF-1) in repression of human GSTA1 expression by interleukin (IL)-1beta in Caco-2 cells. In luciferase reporter assays, overexpression of HNF-1alpha increased GSTA1 transcriptional activity via an HNF-1 response element (HRE) in the proximal promoter. In addition, constitutive mRNA levels of GSTA1 and HNF-1alpha rose concurrently in Caco-2 cells with increasing stage of confluence. IL-1beta reduced GSTA1 mRNA levels at all stages of confluence; however, HNF-1alpha mRNA levels were not altered. IL-1beta repressed GSTA1 transcriptional activity, an effect that was abolished by mutating the HRE. Similar results were observed in HT-29 and HepG2 cells. Overexpression of HNF-1alpha did not counteract IL-1beta-mediated repression of GSTA1 transcription either in reporter assays or at the mRNA level. Involvement of the transdominant repressor C isoform of variant HNF-1 (vHNF-1C) in GSTA1 repression was demonstrated, because vHNF-1C overexpression significantly reduced GSTA1 transcriptional activity. Finally, IL-1beta caused concentration-related up-regulation of vHNF-1C mRNA levels and increased binding of vHNF-1C protein to the HRE, whereas HNF-1alpha-HRE complex formation was reduced. These findings indicate that IL-1beta represses GSTA1 transcription via a mechanism involving overexpression of vHNF-1C.
Collapse
Affiliation(s)
- Lorraine Ng
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada N1G 2W1
| | | | | | | | | |
Collapse
|
16
|
Gautier-Stein A, Zitoun C, Lalli E, Mithieux G, Rajas F. Transcriptional regulation of the glucose-6-phosphatase gene by cAMP/vasoactive intestinal peptide in the intestine. Role of HNF4alpha, CREM, HNF1alpha, and C/EBPalpha. J Biol Chem 2006; 281:31268-31278. [PMID: 16893891 DOI: 10.1074/jbc.m603258200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Gluconeogenesis is induced in both the liver and intestine by increased cAMP levels. However, hepatic and intestinal glucose production can have opposite effects on glucose homeostasis. Glucose release into the portal vein by the intestine increases glucose uptake and reduces food intake. In contrast, glucose production by the liver contributes to hyperglycemia in type II diabetes. Glucose-6-phosphatase (Glc6Pase) is the key enzyme of gluconeogenesis in both the liver and intestine. Here we specify the cAMP/protein kinase A regulation of the Glc6Pase gene in the intestine compared with the liver. Similarly to the liver, the molecular mechanism of cAMP/protein kinase A regulation involves cAMP-response element-binding protein, HNF4alpha, CAAT/enhancer-binding protein, and HNF1. In contrast to the situation in the liver, we find that different isoforms of CAAT/enhancer-binding protein and HNF1 contribute to the specific regulation of the Glc6Pase gene in the intestine. Moreover, we show that cAMP-response element binding modulator specifically contributes to the regulation of the Glc6Pase gene in the intestine but not in the liver. These results allow us to identify intestine-specific regulators of the Glc6Pase gene and to improve the understanding of the differences in the regulation of gluconeogenesis in the intestine compared with the liver.
Collapse
|
17
|
Gautier-Stein A, Zitoun C, Lalli E, Mithieux G, Rajas F. Transcriptional Regulation of the Glucose-6-phosphatase Gene by cAMP/Vasoactive Intestinal Peptide in the Intestine. J Biol Chem 2006. [DOI: 10.1016/s0021-9258(19)84039-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
18
|
Gu N, Adachi T, Matsunaga T, Takeda J, Tsujimoto G, Ishihara A, Yasuda K, Tsuda K. Mutant HNF-1α and mutant HNF-1β identified in MODY3 and MODY5 downregulate DPP-IV gene expression in Caco-2 cells. Biochem Biophys Res Commun 2006; 346:1016-23. [PMID: 16781669 DOI: 10.1016/j.bbrc.2006.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2006] [Accepted: 06/02/2006] [Indexed: 11/26/2022]
Abstract
Dipeptidylpeptidase IV (DPP-IV) is a well-documented drug target for the treatment of type 2 diabetes. Hepatocyte nuclear factors (HNF)-1alpha and HNF-1beta, known as the causal genes of MODY3 and MODY5, respectively, have been reported to be involved in regulation of DPP-IV gene expression. But, it is not completely clear (i) that they play roles in regulation of DPP-IV gene expression, and (ii) whether DPP-IV gene activity is changed by mutant HNF-1alpha and mutant HNF-1beta in MODY3 and MODY5. To explore these questions, we investigated transactivation effects of wild HNF-1alpha and 13 mutant HNF-1alpha, as well as wild HNF-1beta and 2 mutant HNF-1beta, on DPP-IV promoter luciferase gene in Caco-2 cells by means of a transient experiment. Both wild HNF-1alpha and wild HNF-1beta significantly transactivated DPP-IV promoter, but mutant HNF-1alpha and mutant HNF-1beta exhibited low transactivation activity. Moreover, to study whether mutant HNF-1alpha and mutant HNF-1beta change endogenous DPP-IV enzyme activity, we produced four stable cell lines from Caco-2 cells, in which wild HNF-1alpha or wild HNF-1beta, or else respective dominant-negative mutant HNF-1alphaT539fsdelC or dominant-negative mutant HNF-1betaR177X, was stably expressed. We found that DPP-IV gene expression and enzyme activity were significantly increased in wild HNF-1alpha cells and wild HNF-1beta cells, whereas they decreased in HNF-1alphaT539fsdelC cells and HNF-1betaR177X cells, compared with DPP-IV gene expression and enzyme activity in Caco-2 cells. These results suggest that both wild HNF-1alpha and wild HNF-1beta have a stimulatory effect on DPP-IV gene expression, but that mutant HNF-1alpha and mutant HNF-1beta attenuate the stimulatory effect.
Collapse
Affiliation(s)
- Ning Gu
- Laboratory of Metabolism, Graduate School of Human and Environmental Studies, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Senkel S, Lucas B, Klein-Hitpass L, Ryffel GU. Identification of target genes of the transcription factor HNF1beta and HNF1alpha in a human embryonic kidney cell line. ACTA ACUST UNITED AC 2005; 1731:179-90. [PMID: 16297991 DOI: 10.1016/j.bbaexp.2005.10.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2005] [Revised: 09/14/2005] [Accepted: 10/10/2005] [Indexed: 01/01/2023]
Abstract
Hepatocyte nuclear factor 1beta (HNF1beta, TCF2) is a tissue-specific transcription factor whose mutation in humans leads to renal cysts, genital malformations, pancreas atrophy and maturity onset diabetes of the young (MODY5). Furthermore, HNF1beta overexpression has been observed in clear cell cancer of the ovary. To identify potential HNF1beta target genes whose activity may be deregulated in human patients, we established a human embryonic kidney cell line (HEK293) expressing HNF1beta conditionally. Using Flp recombinase, we introduced wild type or mutated HNF1beta at a defined chromosomal position allowing a most reproducible induction of the HNF1beta derivatives upon tetracycline addition. By oligonucleotide microarrays we identified 25 HNF1beta-regulated genes. By an identical approach, we identified that the related transcription factor HNF1alpha (TCF1) affects only nine genes in HEK293 cells and thus is a less efficient factor in these kidney cells. The HNF1beta target genes dipeptidyl peptidase 4 (DPP4), angiotensin converting enzyme 2 (ACE2) and osteopontin (SPP1) are most likely direct target genes, as they contain functional HNF1 binding sites in their promoter region. Since nine of the potential HNF1beta target genes are deregulated in clear cell carcinoma of the ovary, we propose that HNF1beta overexpression in the ovarian cancer participates in the altered expression pattern.
Collapse
Affiliation(s)
- Sabine Senkel
- Institut für Zellbiologie (Tumorforschung), Universitätsklinikum Essen, D-45122 Essen, Germany
| | | | | | | |
Collapse
|
20
|
Isshiki S, Kudo T, Nishihara S, Ikehara Y, Togayachi A, Furuya A, Shitara K, Kubota T, Watanabe M, Kitajima M, Narimatsu H. Lewis type 1 antigen synthase (beta3Gal-T5) is transcriptionally regulated by homeoproteins. J Biol Chem 2003; 278:36611-20. [PMID: 12855703 DOI: 10.1074/jbc.m302681200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The type 1 carbohydrate chain, Galbeta1-3GlcNAc, is synthesized by UDP-galactose:beta-N-acetylglucosamine beta1,3-galactosyltransferase (beta3Gal-T). Among six beta3Gal-Ts cloned to date, beta3Gal-T5 is an essential enzyme for the synthesis of type 1 chain in epithelium of digestive tracts or pancreatic tissue. It forms the type 1 structure on glycoproteins produced from such tissues. In the present study, we found that the transcriptional regulation of the beta3Gal-T5 gene is controlled by homeoproteins, i.e. members of caudal-related homeobox protein (Cdx) and hepatocyte nuclear factor (HNF) families. We found an important region (-151 to -121 from the transcription initiation site), named the beta3Gal-T5 control element (GCE), for the promoter activity. GCE contained the consensus sequences for members of the Cdx and HNF families. Mutations introduced into this sequence abolished the transcriptional activity. Four factors, Cdx1, Cdx2, HNF1alpha, and HNF1beta, could bind to GCE and transcriptionally activate the beta3Gal-T5 gene. Transcriptional regulation of the beta3Gal-T5 gene was consistent with that of members of the Cdx and HNF1 families in two in vivo systems. 1) During in vitro differentiation of Caco-2 cells, transcriptional up-regulation of beta3Gal-T5 was observed in correlation with the increase in transcripts for Cdx2 and HNF1alpha. 2) Both transcript and protein levels of beta3Gal-T5 were determined to be significantly reduced in colon cancer. This down-regulation was correlated with the decrease of Cdx1 and HNF1beta expression in cancer tissue. This is the first finding that a glycosyltransferase gene is transcriptionally regulated under the control of homeoproteins in a tissue-specific manner. beta3Gal-T5, controlled by the intestinal homeoproteins, may play an important role in the specific function of intestinal cells by modifying the carbohydrate structure of glycoproteins.
Collapse
Affiliation(s)
- Soichiro Isshiki
- Division of Cell Biology, Institute of Life Science, Soka University, Tangi-cho, Hachioji, Tokyo 192-8577, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lambeir AM, Durinx C, Scharpé S, De Meester I. Dipeptidyl-peptidase IV from bench to bedside: an update on structural properties, functions, and clinical aspects of the enzyme DPP IV. Crit Rev Clin Lab Sci 2003; 40:209-94. [PMID: 12892317 DOI: 10.1080/713609354] [Citation(s) in RCA: 732] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dipeptidyl-peptidase IV/CD26 (DPP IV) is a cell-surface protease belonging to the prolyloligopeptidase family. It selectively removes the N-terminal dipeptide from peptides with proline or alanine in the second position. Apart from its catalytic activity, it interacts with several proteins, for instance, adenosine deaminase, the HIV gp120 protein, fibronectin, collagen, the chemokine receptor CXCR4, and the tyrosine phosphatase CD45. DPP IV is expressed on a specific set of T lymphocytes, where it is up-regulated after activation. It is also expressed in a variety of tissues, primarily on endothelial and epithelial cells. A soluble form is present in plasma and other body fluids. DPP IV has been proposed as a diagnostic or prognostic marker for various tumors, hematological malignancies, immunological, inflammatory, psychoneuroendocrine disorders, and viral infections. DPP IV truncates many bioactive peptides of medical importance. It plays a role in glucose homeostasis through proteolytic inactivation of the incretins. DPP IV inhibitors improve glucose tolerance and pancreatic islet cell function in animal models of type 2 diabetes and in diabetic patients. The role of DPP IV/ CD26 within the immune system is a combination of its exopeptidase activity and its interactions with different molecules. This enables DPP IV/CD26 to serve as a co-stimulatory molecule to influence T cell activity and to modulate chemotaxis. DPP IV is also implicated in HIV-1 entry, malignant transformation, and tumor invasion.
Collapse
Affiliation(s)
- Anne-Marie Lambeir
- Department of Pharmaceutical Sciences, Laboratory of Medical Biochemistry, University of Antwerp, Universiteitsplein 1, Wilrijk, Belgium.
| | | | | | | |
Collapse
|
22
|
Lambeir AM, Durinx C, Scharpé S, De Meester I. Dipeptidyl-Peptidase IV from Bench to Bedside: An Update on Structural Properties, Functions, and Clinical Aspects of the Enzyme DPP IV. Crit Rev Clin Lab Sci 2003. [DOI: 10.1080/713609354/?{alert(1)}] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2022]
|
23
|
Bai Y, Pontoglio M, Hiesberger T, Sinclair AM, Igarashi P. Regulation of kidney-specific Ksp-cadherin gene promoter by hepatocyte nuclear factor-1beta. Am J Physiol Renal Physiol 2002; 283:F839-51. [PMID: 12217876 DOI: 10.1152/ajprenal.00128.2002] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Kidney-specific cadherin (Ksp-cadherin) is a tissue-specific member of the cadherin family that is expressed exclusively in the kidney and developing genitourinary tract. Recent studies have shown that the proximal 250 bp of the Ksp-cadherin gene promoter are sufficient to direct tissue-specific gene expression in vivo and in vitro. The proximal 120 bp of the promoter are evolutionarily conserved between mouse and human and contain a DNase I hypersensitive site that is kidney cell specific. At position -55, the promoter contains a consensus recognition site for hepatocyte nuclear factor-1 (HNF-1). Mutations of the consensus HNF-1 site and downstream GC-boxes inhibit promoter activity in transfected cells. HNF-1alpha and HNF-1beta bind specifically to the -55 site, and both proteins transactivate the promoter directly. Expression of Ksp-cadherin is not altered in the kidneys of HNF-1alpha-deficient mice. However, expression of a gain-of-function HNF-1beta mutant stimulates Ksp-cadherin promoter activity in transfected cells, whereas expression of a dominant-negative mutant inhibits activity. These studies identify Ksp-cadherin as the first kidney-specific promoter that has been shown to be regulated by HNF-1beta. Mutations of HNF-1beta, as occur in humans with inherited renal cysts and diabetes, may cause dysregulated Ksp-cadherin promoter activity.
Collapse
Affiliation(s)
- Yun Bai
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, 75390, USA
| | | | | | | | | |
Collapse
|
24
|
Lim S, Jin K, Friedman E. Mirk protein kinase is activated by MKK3 and functions as a transcriptional activator of HNF1alpha. J Biol Chem 2002; 277:25040-6. [PMID: 11980910 DOI: 10.1074/jbc.m203257200] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mirk/Dyrk1B is an arginine-directed serine/threonine protein kinase that is expressed at low levels in most normal tissues but at elevated levels in many tumor cell lines and in normal skeletal muscle. Colon carcinoma cell lines stably overexpressing Mirk proliferated in serum-free medium, but the mechanism of Mirk action is unknown. DCoHm (dimerization cofactor of hepatocyte nuclear factor 1alpha ( HNF1alpha) from muscle), a novel gene of the DCoH family with 78% amino acid identity to DCoH, was identified as a Mirk-binding protein by yeast two-hybrid analysis and cloned. Mirk co-immunoprecipitated with DCoHm and bound to DCoHm in glutathione S-transferase pull-down assays. DCoH stabilizes HNF1alpha as a dimer and enhances its transcriptional activity on the beta-fibrinogen promoter reporter, and DCoHm had similar activity. Mirk enhanced HNF1alpha transcriptional activity in a dose-dependent manner, whereas two kinase-inactive Mirk mutants and a Mirk N-terminal deletion mutant did not. Mirk, DCoHm, and HNF1alpha formed a complex. Mirk bound to a specific region within the CREB-binding protein-binding region of HNF1alpha and phosphorylated HNF1alpha at a site adjacent to the Mirk-binding region. Conversely, the HNF1alpha binding domain was located within the first five conserved kinase subdomains of Mirk. Mirk co-immunoprecipitated with the MAPK kinase MKK3, an upstream activator of p38. MKK3 enhanced Mirk kinase activity and the transcriptional activation of HNF1alpha by Mirk, suggesting that Mirk, like p38, is activated by certain environmental stress agents. The Mirk-binding protein DCoH has been shown to be selectively expressed in colon carcinomas but not in normal tissue. Mirk may function as an HNF1alpha transcriptional activator in response to an MKK3-mediated stress signal, and the selective expression of DCoH could restrict the Mirk response to carcinoma cells.
Collapse
Affiliation(s)
- Seunghwan Lim
- Pathology Department, Upstate Medical University, Syracuse, New York 13210, USA
| | | | | |
Collapse
|
25
|
Hwang ST, Urizar NL, Moore DD, Henning SJ. Bile acids regulate the ontogenic expression of ileal bile acid binding protein in the rat via the farnesoid X receptor. Gastroenterology 2002; 122:1483-92. [PMID: 11984532 DOI: 10.1053/gast.2002.32982] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS In the rat, an increase in ileal bile acid binding protein (IBABP) expression occurs during the third postnatal week. In vitro studies suggest that bile acids (BAs) increase IBABP transcription by activating the BA receptor, farnesoid X receptor (FXR). Thus, we investigated the role of BAs on the ontogenic expression of IBABP and whether FXR may mediate these effects. METHODS Suckling rats were gavage-fed taurocholate for 3 days or were allowed to develop normally. Ileums were collected for Northern and Western blot analyses. Electrophoretic mobility shift assays for functional FXR were performed using nuclear extracts from ileums of both adult and developing rats. RESULTS Taurocholate gavage significantly elevated IBABP messenger RNA and protein levels in suckling animals. Gelshift assays using adult ileal nuclear extracts incubated with a radiolabeled consensus inverted repeat-1 oligonucleotide (response element for FXR) revealed a high-molecular weight DNA/protein complex. Cold competition and supershift assays showed that this complex is sequence specific and confirmed that FXR is a component of the complex. Gelshift assays with nuclear extracts from rat ileum at different ages revealed absence of the DNA/protein complex in the second postnatal week when there is lack of IBABP expression and presence of these complexes at later ages when there is normally high expression. Western blot analyses showed FXR and its heterodimer partner, retinoid X receptor alpha, protein levels are low in the ileum during the suckling period and increase during the third postnatal week. CONCLUSIONS BAs play a role in the normal developmental expression of IBABP through FXR activation, and decreased functional FXR in ileal nuclei during the suckling period may account, in part, for the lack of IBABP expression at this time.
Collapse
Affiliation(s)
- Sandy T Hwang
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA.
| | | | | | | |
Collapse
|
26
|
Yamada S, Kojima H, Fujimiya M, Nakamura T, Kashiwagi A, Kikkawa R. Differentiation of immature enterocytes into enteroendocrine cells by Pdx1 overexpression. Am J Physiol Gastrointest Liver Physiol 2001; 281:G229-36. [PMID: 11408276 DOI: 10.1152/ajpgi.2001.281.1.g229] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The development of a variety of enteroendocrine cells of the gut is poorly understood. We tested whether immature intestinal stem cells were switched to multiple enteroendocrine hormone-producing cells by in vitro transfer of a homeobox gene. We transfected the pancreatic-duodenal homeobox 1 gene (Pdx1) into IEC-6 cells, an embryonic intestinal epithelial cell line derived from a normal rat, and selected the cells that overexpressed Pdx1 by 150-fold compared with control. The cells were examined for differentiation into enteroendocrine cells by immunocytochemical and electron microscopic analyses. Transfected cells cultured on micropore filters formed a trabecular network piled up on monolayer cells. These trabecular cells showed nuclear localization of Pdx1 protein and contained well-developed rough endoplasmic reticulum as well as many secretory granules of pleomorphic shape in the cytoplasm. Antibodies against chromogranin A, serotonin, cholecystokinin, gastrin, and somatostatin stained these secretory granules in the cytoplasm. Furthermore, immunofluorescence double staining analysis showed that different hormones were produced within a cell. These results provide the evidence that immature intestinal epithelial cells can differentiate into multiple hormone-producing enteroendocrine cells in response to overexpression of Pdx1.
Collapse
Affiliation(s)
- S Yamada
- Third Department of Medicine, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Study of the molecular and cellular biology of the small-intestinal mucosa is providing insights into the remarkable properties of this unique tissue. With its structured pattern of cell proliferation, differentiation, and apoptosis, and its ability to adapt following exposure to luminal nutrients or injury from surgery or pathogens, it functions in a regulated but responsive manner. We review recent publications on factors affecting development, gene expression, cell turnover, and adaptation.
Collapse
Affiliation(s)
- Paul A. Kitchen
- Gastroenterology Section, Department of Medicine, Imperial College School of Medicine, Hammersmith Hospital, London, UK
| | | |
Collapse
|
28
|
Gum JR, Erickson RH, Hicks JW, Rius JL, Kim YS. Analysis of dipeptidyl peptidase IV gene regulation in transgenic mice: DNA elements sufficient for promoter activity in the kidney, but not the intestine, reside on the proximal portion of the gene 5'-flanking region. FEBS Lett 2000; 482:49-53. [PMID: 11018521 DOI: 10.1016/s0014-5793(00)02013-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The dipeptidyl peptidase IV (DPPIV) gene encodes a brush border membrane exopeptidase that is expressed in a tissue-restricted fashion. To examine the regulation of DPPIV transcription in various tissues in vivo, we examined the expression of DPPIV 5'-flanking region (promoter)-human growth hormone reporter constructs in transgenic mice. These mice exhibited cell-type specific reporter expression in kidney. Surprisingly, however, only very low to non-detectable levels of reporter were found in small intestine. These results indicate that DNA elements sufficient for DPPIV expression in kidney, but not intestine, reside in the 5'-flanking region of the gene.
Collapse
Affiliation(s)
- J R Gum
- Gastrointestinal Research Laboratory, Department of Veterans' Affairs Medical Center, San Francisco, CA 94121, USA.
| | | | | | | | | |
Collapse
|