1
|
Nasser S, Abdallah DM, Ahmed KA, Abdel-Mottaleb Y, El-Abhar HS. The novel anti-colitic effect of β-adrenergic receptors via modulation of PS1/BACE-1/Aβ axis and NOTCH signaling in an ulcerative colitis model. Front Pharmacol 2022; 13:1008085. [PMID: 36386153 PMCID: PMC9641009 DOI: 10.3389/fphar.2022.1008085] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2023] Open
Abstract
Although dysautonomia was documented in inflammatory bowel disease, with activation of the stress-related sympathetic system, the role of agonists/antagonists of the adrenergic receptors is not conclusive. Moreover, ulcerative colitis was recently linked to dementia, but the potential role of the presenilin 1(PS1)/BACE-1/beta-amyloid (Aβ) axis has not been evaluated. Hence, we investigated the impact of mirabegron (β3-agonist) and/or carvedilol (β1/β2 antagonist) on iodoacetamide-induced ulcerative colitis with emphasis on the novel pathomechanism of the PS1/BACE-1/Aβ axis in ulcerative colitis, and its relation to the inflammatory cascade, fibrotic processes, and the gut barrier dysfunction. Ulcerated rats were either left untreated or treated for 8 days with mirabegron and/or carvedilol. Besides minimizing colon edema and weight loss, and improving colon structure, mirabegron and/or carvedilol abated colonic PS1/BACE-1/Aβ axis and the NOTCH1/NICD/HES1 hub besides the inflammatory cascade GSK3-β/NF-κΒ/TNF-α, and the oxidative stress marker malondialdehyde. The anti-fibrotic effect was verified by boosting SMAD-7 and inhibiting TGF-β1, α-SMA immunoexpression, and MTC staining. Moreover, the drugs improved the gut barrier function, attested by the increased goblet cells and expression of E-cadherin, and the inhibited expression of p (Y654)-β-catenin to preserve the E-cadherin/β-catenin adherens junction (AJ). These signaling pathways may be orchestrated by the replenished PPAR-γ, a transcription factor known for its anti-colitic effect. Conclusion: Besides maintaining the gut barrier, mirabegron and/or carvedilol mediated their anti-colitic effect by their anti-oxidant, anti-inflammatory, and anti-fibrotic capacities. The therapeutic effect of these drugs depends partly on suppressing the harmful signaling pathways PS1/BACE-1/Aβ, NOTCH1/NICD/HES1, GSK3-β/NF-κΒ/TNF-α, and TGF-1β/α-SMA while enhancing PPAR-γ, SMAD-7, mucus, and AJ.
Collapse
Affiliation(s)
- Salma Nasser
- Pharmacology, Toxicology and Biochemistry Department, Faculty of Pharmacy, Future University in Egypt (FUE), New Cairo, Egypt
| | - Dalaal M. Abdallah
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Kawkab A. Ahmed
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Yousra Abdel-Mottaleb
- Pharmacology, Toxicology and Biochemistry Department, Faculty of Pharmacy, Future University in Egypt (FUE), New Cairo, Egypt
| | - Hanan S. El-Abhar
- Pharmacology, Toxicology and Biochemistry Department, Faculty of Pharmacy, Future University in Egypt (FUE), New Cairo, Egypt
| |
Collapse
|
2
|
MSC secreted extracellular vesicles carrying TGF-beta upregulate Smad 6 expression and promote the regrowth of neurons in spinal cord injured rats. Stem Cell Rev Rep 2021; 18:1078-1096. [PMID: 34449013 PMCID: PMC8942898 DOI: 10.1007/s12015-021-10219-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2021] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) constitute a promising therapy for spinal cord injury (SCI) because they can provide a favorable environment for the regrowth of neurons by inhibiting receptor-regulated Smads (R-Smads) expression in endogenous neural stem cells (NSCs). However, their mechanism of action and effect on the expression of inhibitory Smads (I-Smads) remain unclear. Herein, we demonstrated that extracellular vesicles (EVs) from MSCs were able to upregulate the Smad 6 expression by carrying TGF-β, and the Smad 6 knockdown in NSCs partially weakened the bone marrow MSC (BMSC)-EV-induced effect on neural differentiation. We found that the expression of Smad 6 did not reduced owing to the TGF-β type I receptor kinase inhibitor, SB 431,542, treatment in the acute phase of injury in rats with SCI, thereby indicating that the Smad 6 expression was not only mediated by TGF-β, but also by the inflammatory factors and bone morphogenetic proteins (BMPs) as well. However, in the later phase of SCI, the Smad 6 expression decreased by the addition of SB 431,542, suggesting that TGF-β plays a key role in the mediation of Smad 6 expression in this phase. In addition, immunohistochemistry staining; hematoxylin-eosin staining; and the Basso, Beattie, and Bresnahan (BBB) scores revealed that the early inhibition of TGF-β did not increase neuron regrowth. However, this inhibition increased the cavity and the caspase-3 expression at 24 h post-injury, leading to a worse functional outcome. Conversely, the later treatment with the TGF-β inhibitor promoted the regrowth of neurons around the cavity, resulting in a better neurological outcome. Together, these results indicate that Smad 6 acts as a feedback regulator to prevent the over-differentiation of NSCs to astrocytes and that BMSC-EVs can upregulate Smad 6 expression by carrying TGF-β.
Collapse
|
3
|
Abu Rmilah AA, Zhou W, Nyberg SL. Hormonal Contribution to Liver Regeneration. Mayo Clin Proc Innov Qual Outcomes 2020; 4:315-338. [PMID: 32542223 PMCID: PMC7283948 DOI: 10.1016/j.mayocpiqo.2020.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/01/2020] [Accepted: 02/07/2020] [Indexed: 02/07/2023] Open
Abstract
An understanding of the molecular basis of liver regeneration will open new horizons for the development of novel therapies for chronic liver failure. Such therapies would solve the drawbacks associated with liver transplant, including the shortage of donor organs, long waitlist time, high medical costs, and lifelong use of immunosuppressive agents. Regeneration after partial hepatectomy has been studied in animal models, particularly fumarylacetoacetate hydrolase-deficient (FAH -/-) mice and pigs. The process of regeneration is distinctive, complex, and well coordinated, and it depends on the interplay among several signaling pathways (eg, nuclear factor κβ, Notch, Hippo), cytokines (eg, tumor necrosis factor α, interleukin 6), and growth factors (eg, hepatocyte growth factor, epidermal growth factor, vascular endothelial growth factor), and other components. Furthermore, endocrinal hormones (eg, norepinephrine, growth hormone, insulin, thyroid hormones) also can influence the aforementioned pathways and factors. We believe that these endocrinal hormones are important hepatic mitogens that strongly induce and accelerate hepatocyte proliferation (regeneration) by directly and indirectly triggering the activity of the involved signaling pathways, cytokines, growth factors, and transcription factors. The subsequent induction of cyclins and associated cyclin-dependent kinase complexes allow hepatocytes to enter the cell cycle. In this review article, we comprehensively summarize the current knowledge regarding the roles and mechanisms of these hormones in liver regeneration. Articles used for this review were identified by searching MEDLINE and EMBASE databases from inception through June 1, 2019.
Collapse
Key Words
- CDK, cyclin-dependent kinase
- EGF, epidermal growth factor
- EGFR, EGF receptor
- ERK, extracellular signal-regulated kinase
- FAH, fumarylacetoacetate hydrolase
- GH, growth hormone
- Ghr-/-, growth hormone receptor gene knockout
- HGF, hepatocyte growth factor
- HNF, hepatocyte nuclear factor
- HPC, hepatic progenitor cell
- IGF, insulinlike growth factor
- IL, interleukin
- IR, insulin receptor
- InsP3, inositol 1,4,5-trisphosphate
- JNK, JUN N-terminal kinase
- LDLT, living donor liver transplant
- LRP, low-density lipoprotein-related protein
- MAPK, mitogen-activated protein kinase
- NF-κβ, nuclear factor κβ
- NOS, nitric oxide synthase
- NTBC, 2-nitro-4-trifluoro-methyl-benzoyl-1,3-cyclohexanedione
- PCNA, proliferating cell nuclear antigen
- PCR, polymerase chain reaction
- PH, partial hepatectomy
- PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase
- PKB, protein kinase B
- PTU, 6-n-propyl-2-thiouracil
- ROS, reactive oxygen species
- STAT, signal transducer and activator of transcription
- T3, triiodothyronine
- TGF, transforming growth factor
- TNF, tumor necrosis factor
- TR, thyroid receptor
- hESC, human embryonic stem cell
- hiPSC, human induced pluripotent stem cells
- mRNA, messenger RNA
- mTOR, mammalian target of rapamycin
Collapse
Affiliation(s)
| | - Wei Zhou
- Division of Transplantation Surgery, Mayo Clinic, Rochester, MN.,First Affiliated Hospital of China, Medical University, Department of Hepatobiliary Surgery, Shenyang, China
| | - Scott L Nyberg
- Division of Transplantation Surgery, Mayo Clinic, Rochester, MN
| |
Collapse
|
4
|
Tao Y, Wang M, Chen E, Tang H. Liver Regeneration: Analysis of the Main Relevant Signaling Molecules. Mediators Inflamm 2017; 2017:4256352. [PMID: 28947857 PMCID: PMC5602614 DOI: 10.1155/2017/4256352] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/19/2017] [Accepted: 08/10/2017] [Indexed: 02/06/2023] Open
Abstract
Liver regeneration is a highly organized tissue regrowth process and is the most important reaction of the liver to injury. The overall process of liver regeneration includes three phases: priming stage, proliferative phase, and termination phase. The initial step aims to induce hepatocytes to be sensitive to growth factors with the aid of some cytokines, including TNF-α and IL-6. The proliferation phase promotes hepatocytes to re-enter G1 with the stimulation of growth factors. While during the termination stage, hepatocytes will discontinue to proliferate to maintain normal liver mass and function. Except for cytokine- and growth factor-mediated pathways involved in regulating liver regeneration, new substances and technologies emerge to influence the regenerative process. Here, we reviewed novel and important signaling molecules involved in the process of liver regeneration to provide a cue for further research.
Collapse
Affiliation(s)
- Yachao Tao
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Menglan Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Enqiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Marlo TL, Giuliano EA, Tripathi R, Sharma A, Mohan RR. Altering equine corneal fibroblast differentiation through Smad gene transfer. Vet Ophthalmol 2017; 21:132-139. [PMID: 28685927 DOI: 10.1111/vop.12485] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE To explore the impact of equine corneal fibroblast (ECF) to myofibroblast (ECM) differentiation by altering the expression of the Smad genes either individually or in combination. Specifically, we sought to examine the ECF differentiation after (a) silencing of Smad2, 3, and 4 profibrotic genes individually and (b) overexpression of antifibrotic Smad7 gene and in a combination with pro- and antifibrotic Smad genes. METHODS Equine corneal fibroblast primary cultures were generated as previously described. ECFs were transfected with individual plasmids which silenced gene expression of either Smad2, 3, or 4 or in combination with a plasmid overexpressing Smad7 using Lipofectamine 2000™ or Lipofectamine BLOCK-iT™. Smad-transfected clones were then exposed to TGF-β1 to induce differentiation to myofibroblasts. Immunofluorescence and qRT-PCR techniques quantified levels of ECF differentiation to ECM by measuring alpha smooth muscle actin, a known marker of ECM transdifferentiation. RESULTS Silencing of individual Smad2, 3, or 4 genes or overexpression of Smad7 showed significant inhibition of ECF transdifferentiation (73-83% reduction). Silencing of Smad2 showed the greatest inhibition of ECF transdifferentiation in (a) and was therefore utilized for the combination gene transfer testing. The combination gene transfer consisting of Smad7 overexpression and Smad2 silencing attenuated ECF differentiation significantly; however, the level was not significant compared to the overexpression of Smad7 individually. CONCLUSIONS Using gene transfer technology involving profibrotic Smad silencing, antifibrotic Smad overexpression or its combination is a novel strategy to control TGF-β1-mediated fibrosis in equine fibroblasts. Combination gene therapy was not better than single gene therapy in this study.
Collapse
Affiliation(s)
- Todd L Marlo
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Elizabeth A Giuliano
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Ratnakar Tripathi
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.,Harry S. Truman Veterans Memorial Hospital, Columbia, MO, USA
| | - Ajay Sharma
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.,Harry S. Truman Veterans Memorial Hospital, Columbia, MO, USA
| | - Rajiv R Mohan
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.,Harry S. Truman Veterans Memorial Hospital, Columbia, MO, USA.,Mason Eye Institute, University of Missouri, 1 Hospital Drive, Columbia, MO, USA
| |
Collapse
|
6
|
Abstract
Inhibitory Smads (I-Smads) have conserved carboxy-terminal MH2 domains but highly divergent amino-terminal regions when compared with receptor-regulated Smads (R-Smads) and common-partner Smads (co-Smads). Smad6 preferentially inhibits Smad signaling initiated by the bone morphogenetic protein (BMP) type I receptors ALK-3 and ALK-6, whereas Smad7 inhibits both transforming growth factor β (TGF-β)- and BMP-induced Smad signaling. I-Smads also regulate some non-Smad signaling pathways. Here, we discuss the vertebrate I-Smads, their roles as inhibitors of Smad activation and regulators of receptor stability, as scaffolds for non-Smad signaling, and their possible roles in the nucleus. We also discuss the posttranslational modification of I-Smads, including phosphorylation, ubiquitylation, acetylation, and methylation.
Collapse
Affiliation(s)
- Keiji Miyazawa
- Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi 409-3898, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
7
|
Shi Y, Zhang H, Han Z, Mi X, Zhang W, Lv M. HBx interacted with Smad4 to deprive activin a growth inhibition function in hepatocyte HL7702 on CRM1 manner. Tumour Biol 2016; 37:3405-15. [PMID: 26449823 DOI: 10.1007/s13277-015-4076-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/13/2015] [Indexed: 01/16/2023] Open
Abstract
Hepatitis B virus (HBV) is implicated in the pathogenesis of hepatocellular carcinoma, which has been found to be associated with TGF-beta signaling. Activin A is a TGF-β family cytokine that exhibits cell proliferation inhibition on normal hepatocyte. How HBV-encoded X oncoprotein play in activin's activity on hepatocyte has not been developed. In this study, a nontumor hepatic cell line HL7702 with HBX ectogenic expression has been established. MTT and BrdU assays showed that HBx promoted growth of HL7702 cells in vitro and downregulated activin signaling. Deregulated activin signaling pathway by HBX failed to activate target gene p21/waf1 and p15 transcription. In addition, mammalian two-hybrid and coimmunoprecipitation assays revealed that HBX could directly interact with activin signaling transduction protein Smad4, making activated Smad2/3/4 nucleus translocation suppressed. Furthermore, we detected that leptomycin B, the inhibitor of CRM1 protein, could recover nuclear translocation of endogenous Smads complex in HL7702 with HBX expression, indicating that HBX antagonized Smads nucleus translocation, at least partially, on CRM1-dependent manner. Leptomycin B was found to have antigrowth activity on HBX-expressed HL7702, according to its antitumor function in previous study. Above all, HBX antagonized activin signaling in normal human liver cells by interacting with Smad4 might one of the considerable causes of HBX-induced hepatocyte transformation, which deprived activin's cell growth inhibition function at an early stage of tumorigenesis.
Collapse
Affiliation(s)
- Ying Shi
- Institute of Translational Medicine, First Hospital of Jilin University, Changchun, People's Republic of China
| | - Haipeng Zhang
- The First Clinical Medical College of Jilin University, Changchun, People's Republic of China
| | - Zhu Han
- Institute of Translational Medicine, First Hospital of Jilin University, Changchun, People's Republic of China
| | - Xuguang Mi
- Tumor Biological Treatment Center of Jilin Province People's Hospital, Changchun, People's Republic of China
| | - Wenyan Zhang
- Institute of Translational Medicine, First Hospital of Jilin University, Changchun, People's Republic of China.
| | - Mingyu Lv
- Institute of Translational Medicine, First Hospital of Jilin University, Changchun, People's Republic of China.
| |
Collapse
|
8
|
Abstract
Liver regeneration is perhaps the most studied example of compensatory growth aimed to replace loss of tissue in an organ. Hepatocytes, the main functional cells of the liver, manage to proliferate to restore mass and to simultaneously deliver all functions hepatic functions necessary to maintain body homeostasis. They are the first cells to respond to regenerative stimuli triggered by mitogenic growth factor receptors MET (the hepatocyte growth factor receptor] and epidermal growth factor receptor and complemented by auxiliary mitogenic signals induced by other cytokines. Termination of liver regeneration is a complex process affected by integrin mediated signaling and it restores the organ to its original mass as determined by the needs of the body (hepatostat function). When hepatocytes cannot proliferate, progenitor cells derived from the biliary epithelium transdifferentiate to restore the hepatocyte compartment. In a reverse situation, hepatocytes can also transdifferentiate to restore the biliary compartment. Several hormones and xenobiotics alter the hepatostat directly and induce an increase in liver to body weight ratio (augmentative hepatomegaly). The complex challenges of the liver toward body homeostasis are thus always preserved by complex but unfailing responses involving orchestrated signaling and affecting growth and differentiation of all hepatic cell types.
Collapse
Affiliation(s)
- George K Michalopoulos
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
9
|
Kang LI, Mars WM, Michalopoulos GK. Signals and cells involved in regulating liver regeneration. Cells 2012; 1:1261-1292. [PMID: 24710554 PMCID: PMC3901148 DOI: 10.3390/cells1041261] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 11/27/2012] [Accepted: 12/07/2012] [Indexed: 12/11/2022] Open
Abstract
Liver regeneration is a complex phenomenon aimed at maintaining a constant liver mass in the event of injury resulting in loss of hepatic parenchyma. Partial hepatectomy is followed by a series of events involving multiple signaling pathways controlled by mitogenic growth factors (HGF, EGF) and their receptors (MET and EGFR). In addition multiple cytokines and other signaling molecules contribute to the orchestration of a signal which drives hepatocytes into DNA synthesis. The other cell types of the liver receive and transmit to hepatocytes complex signals so that, in the end of the regenerative process, complete hepatic tissue is assembled and regeneration is terminated at the proper time and at the right liver size. If hepatocytes fail to participate in this process, the biliary compartment is mobilized to generate populations of progenitor cells which transdifferentiate into hepatocytes and restore liver size.
Collapse
Affiliation(s)
- Liang-I Kang
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Wendy M Mars
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | |
Collapse
|
10
|
Voumvouraki A, Notas G, Koulentaki M, Georgiadou M, Klironomos S, Kouroumalis E. Increased serum activin-A differentiates alcoholic from cirrhosis of other aetiologies. Eur J Clin Invest 2012; 42:815-22. [PMID: 22304651 DOI: 10.1111/j.1365-2362.2012.02647.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Activin-A is a molecule of the TGF superfamily, implicated in liver fibrosis, regeneration and stem cell differentiation. However, data on activins in liver diseases are few. We therefore studied serum levels of activin-A in chronic liver diseases. To identify the origin of activin-A, levels in the hepatic vein were also estimated. MATERIALS AND METHODS Nineteen controls and 162 patients participated in the study: 39 with hepatocellular carcinoma (HCC: 19 viral associated and 20 alcohol associated), 18 with chronic hepatitis C (CHC), 47 with primary biliary cirrhosis (26 PBC stage I-II and 21 stage IV), 22 with alcoholic cirrhosis (AC, hepatic vein blood available in 16), 20 with HCV cirrhosis (hepatic vein blood available in 18) and 16 patients with alcoholic fatty liver with mild to moderate fibrosis but no cirrhosis. RESULTS Activin-A levels were significantly increased (P < 0·001) in serum of patients with AC (median 673 pg/mL, range 449-3279), compared with either controls (149 pg/mL, 91-193) or patients with viral cirrhosis (189 pg/mL, 81-480), CHC (142 pg/mL, 65-559) PBC stage I-II (100 pg/mL, 59-597) and PBC stage IV (104 pg/mL, 81-579). Only patients with AC-associated HCC had significantly increased levels of activin-A (2403 pg/mL, 1561-7220 pg/mL). Activin-A serum levels could accurately discriminate AC from cirrhosis of other aetiologies and noncirrhotic alcoholic fatty liver with fibrosis. CONCLUSIONS Increased serum levels of activin-A only in patients with alcohol-related cirrhosis or HCC suggest a possible role of this molecule in the pathophysiology of AC. Further research is warranted to elucidate its role during the profibrotic process and its possible clinical applications.
Collapse
Affiliation(s)
- Argyro Voumvouraki
- University Hospital Department of Gastroenterology, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | | | | | | | | | | |
Collapse
|
11
|
|
12
|
Muenster U, Korupolu R, Rastogi R, Read J, Fischer WH. Antagonism of activin by activin chimeras. VITAMINS AND HORMONES 2011; 85:105-28. [PMID: 21353878 DOI: 10.1016/b978-0-12-385961-7.00006-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Activins are pluripotent hormones/growth factors that belong to the TGF-β superfamily of growth and differentiation factors (GDFs). They play a role in cell growth, differentiation and apoptosis, endocrine function, metabolism, wound repair, immune responses, homeostasis, mesoderm induction, bone growth, and many other biological processes. Activins and the related bone morphogenic proteins (BMPs) transduce their signal through two classes of single transmembrane receptors. The receptors possess intracellular serine/threonine kinase domains. Signaling occurs when the constitutively active type II kinase domain phosphorylates the type I receptor, which upon activation, phosphorylates intracellular signaling molecules. To generate antagonistic ligands, we generated chimeric molecules that disrupt the receptor interactions and thereby the phosphorylation events. The chimeras were designed based on available structural data to maintain high-affinity binding to type II receptors. The predicted type I receptor interaction region was replaced by residues present in inactive homologs or in related ligands with different type I receptor affinities.
Collapse
Affiliation(s)
- Uwe Muenster
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, California, USA
| | | | | | | | | |
Collapse
|
13
|
Zhang DF, Li XG, Su LJ, Meng QL. Expression of activin A and follistatin in glioblastoma and their effects on U87 in vitro. J Int Med Res 2010; 38:1343-53. [PMID: 20926007 DOI: 10.1177/147323001003800416] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In some cancer cell lines, the gene encoding activin A (inhibin βA [INHBA]) is over-expressed and enhances cancer proliferation. Protein levels of activin A and follistatin were assessed in glioblastoma and normal brain tissues in this study, and the effect of activin A and follistatin treatment on the proliferation of U87 human glioblastoma cells in vitro was also studied. High levels of activin A were observed in glioblastomas compared with normal brain tissue. In contrast, follistatin levels were similar between the two tissues. [(3)H]Thymidine assay showed that activin A (3 - 30 ng/ml) produced a dose-dependent increase in DNA synthesis of U87 cells compared with controls. Flow cytometric analyses showed that activin A increased the proliferative index of U87 cells compared with controls. Activin A also induced up-regulation of p-SMAD2/3 in a dose-dependent manner. Treatment of U87 cells with follistatin blocked these activin A-induced effects. The disequilibrium between activin A and follistatin may play a role in the development of glioblastoma.
Collapse
Affiliation(s)
- D F Zhang
- Department of Neurosurgery, Qilu Hospital, Medical College of Shandong University, Jinan, China
| | | | | | | |
Collapse
|
14
|
IL-13 promotes the proliferation of rat pancreatic stellate cells through the suppression of NF-kappaB/TGF-beta1 pathway. Biochem Biophys Res Commun 2010; 393:61-5. [PMID: 20100461 DOI: 10.1016/j.bbrc.2010.01.078] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Accepted: 01/17/2010] [Indexed: 11/22/2022]
Abstract
In chronic pancreatitis, pancreatic stellate cells (PSCs) play a central role in tissue fibrogenesis. Transforming growth factor beta(1) (TGF-beta(1)) and the Th2 lymphokines such as interleukin (IL)-13 are major profibrogenic cytokines in many organs. Activated PSCs produce various inflammatory cytokines including TGF-beta(1). In this study, we investigated whether IL-13 affects pancreatic fibrogenesis by modulating the functions of PSCs. IL-13 promoted PSCs proliferation without activation through the suppression of autocrine TGF-beta(1). IL-13 enhanced Stat6 phosphorylation in PSCs but Stat6 was not involved in the suppression of TGF-beta(1). IL-13 inhibited the transcriptional activity of NF-kappaB, and the expression of mutant I-kappaB reproduced the suppression of autocrine TGF-beta(1) and promoted PSCs proliferation. Taken together, we demonstrated that IL-13 promotes PSCs proliferation through the suppression of the transcriptional activity of NF-kappaB, resulting in the decrease of autocrine TGF-beta(1). This finding provides an unequivocal evidence of IL-13 participation in pancreatic fibrosis, illustrating a new strategy for chronic pancreatitis.
Collapse
|
15
|
Shinozaki S, Ohnishi H, Hama K, Kita H, Yamamoto H, Osawa H, Sato K, Tamada K, Mashima H, Sugano K. Indian hedgehog promotes the migration of rat activated pancreatic stellate cells by increasing membrane type-1 matrix metalloproteinase on the plasma membrane. J Cell Physiol 2008; 216:38-46. [PMID: 18286538 DOI: 10.1002/jcp.21372] [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: 01/11/2023]
Abstract
Indian hedgehog (Ihh) is a member of hedgehog peptides family that exerts diverse effects on multiple cellular functions. Since Ihh expression is elevated in the pancreas of chronic pancreatitis patients, Ihh has been assumed to participate in the chronic pancreatic injury, especially in pancreatic fibrosis. However, its function in pancreatic fibrosis is still unknown. We thus examined Ihh effects on rat activated pancreatic stellate cells (PSCs) that play a central role in pancreatic fibrosis. Activated PSCs express both patched-1 and smoothened that are essential components of hedgehog receptor system. Ihh did not alter the PSC expression of collagen-1 or alpha-smooth muscle actin, a parameter of PSC transformation, or did not change PSC proliferation. However, Ihh enhanced PSC migration in both chemotactic and chemokinetic manners. Furthermore, Ihh increased the amount of membrane-type 1 matrix metalloproteinase (MT1-MMP) and altered its localization on the plasma membrane, which plays a stimulatory role in cellular migration. In addition, tissue inhibitor of metalloproteinase-2 (TIMP-2) attenuated Ihh-stimulated PSC migration. Since most hedgehog intracellular signals are mediated by Gli-1 transcription factor, we investigated its contribution to Ihh-enhancement of PSC migration. Ihh induced Gli-1 nuclear accumulation in PSCs, indicating that Ihh stimulates Gli-1-dependent signaling pathway in PSCs. Unexpectedly, however, adenovirus-mediated Gli-1 overexpression blocked the Ihh enhancement of both MT1-MMP localization on the plasma membrane and PSC migration. Furthermore, reduction of Gli-1 expression with RNA interference augmented Ihh-stimulated PSC migration. These data indicate that Ihh promotes PSC migration by enhancing MT1-MMP localization on the plasma membrane but is negatively regulated by Gli-1.
Collapse
Affiliation(s)
- Satoshi Shinozaki
- Division of Gastroenterology, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Deli A, Kreidl E, Santifaller S, Trotter B, Seir K, Berger W, Schulte-Hermann R, Rodgarkia-Dara C, Grusch M. Activins and activin antagonists in hepatocellular carcinoma. World J Gastroenterol 2008; 14:1699-709. [PMID: 18350601 PMCID: PMC2695910 DOI: 10.3748/wjg.14.1699] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In many parts of the world hepatocellular carcinoma (HCC) is among the leading causes of cancer-related mortality but the underlying molecular pathology is still insufficiently understood. There is increasing evidence that activins, which are members of the transforming growth factor β (TGFβ) superfamily of growth and differentiation factors, could play important roles in liver carcinogenesis. Activins are disulphide-linked homo- or heterodimers formed from four different β subunits termed βA, βB, βC, and βE, respectively. Activin A, the dimer of two βA subunits, is critically involved in the regulation of cell growth, apoptosis, and tissue architecture in the liver, while the hepatic function of other activins is largely unexplored so far. Negative regulators of activin signals include antagonists in the extracellular space like the binding proteins follistatin and FLRG, and at the cell membrane antagonistic co-receptors like Cripto or BAMBI. Additionally, in the intracellular space inhibitory Smads can modulate and control activin activity. Accumulating data suggest that deregulation of activin signals contributes to pathologic conditions such as chronic inflammation, fibrosis and development of cancer. The current article reviews the alterations in components of the activin signaling pathway that have been observed in HCC and discusses their potential significance for liver tumorigenesis.
Collapse
|
17
|
Breitkopf K, Weng H, Dooley S. TGF-β/Smad-signaling in liver cells: Target genes and inhibitors of two parallel pathways. ACTA ACUST UNITED AC 2006. [DOI: 10.1002/sita.200600097] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
18
|
Osawa H, Ohnishi H, Takano K, Noguti T, Mashima H, Hoshino H, Kita H, Sato K, Matsui H, Sugano K. Sonic hedgehog stimulates the proliferation of rat gastric mucosal cells through ERK activation by elevating intracellular calcium concentration. Biochem Biophys Res Commun 2006; 344:680-7. [PMID: 16630542 DOI: 10.1016/j.bbrc.2006.03.188] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2006] [Accepted: 03/28/2006] [Indexed: 12/20/2022]
Abstract
Sonic Hedgehog (Shh), a member of hedgehog peptides family, is expressed in gastric gland epithelium. To elucidate Shh function to gastric mucosal cells, we examined the effect of Shh on the proliferation of a rat normal gastric mucosal cell line, RGM-1. RGM-1 cells express essential components of Shh receptor system, patched-1, and smoothened. Shh enhanced DNA synthesis in RGM-1 cells and elevated intracellular calcium concentration ([Ca2+]i). In addition, Shh as well as calcium ionophore A32187 rapidly activated ERK. However, Shh failed to activate ERK under calcium-free culture condition. Pretreatment of cells with PD98059 attenuated the DNA synthesis promoted by Shh. Moreover, when cells were pretreated with cyclopamine, Shh could not elevate [Ca2+]i, activate ERK or promote DNA synthesis. On the other hand, although Shh induced Gli-1 nuclear accumulation in RGM-1 cells, Shh activated ERK even in cells pretreated with actinomycin D. These results indicate that Shh promotes the proliferation of RGM-1 cells through an intracellular calcium- and ERK-dependent but transcription-independent pathway via Patched/Smoothened receptor system.
Collapse
Affiliation(s)
- Hiroyuki Osawa
- Department of Gastroenterology, Jichi Medical School, Tochigi 329-0498, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hama K, Ohnishi H, Aoki H, Kita H, Yamamoto H, Osawa H, Sato K, Tamada K, Mashima H, Yasuda H, Sugano K. Angiotensin II promotes the proliferation of activated pancreatic stellate cells by Smad7 induction through a protein kinase C pathway. Biochem Biophys Res Commun 2005; 340:742-50. [PMID: 16380081 DOI: 10.1016/j.bbrc.2005.12.069] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Accepted: 12/12/2005] [Indexed: 12/14/2022]
Abstract
Activated pancreatic stellate cells (PSCs) play major roles in promoting pancreatic fibrosis. We previously reported that angiotensin II (Ang II) enhances activated PSC proliferation through EGF receptor transactivation. In the present study, we elucidated a novel intracellular mechanism by which Ang II stimulates cellular proliferation. TGF-beta1 inhibits activated PSC proliferation via a Smad3 and Smad4-dependent pathway in an autocrine manner. We demonstrated that Ang II inhibited TGF-beta1-induced nuclear accumulation of Smad3 and Smad4. Furthermore, Ang II rapidly induced inhibitory Smad7 mRNA expression. Adenovirus-mediated Smad7 overexpression inhibited TGF-beta1-induced nuclear accumulation of Smad3 and Smad4, and potentiated activated PSC proliferation. PKC inhibitor Go6983 blocked the induction of Smad7 mRNA expression by Ang II. In addition, 12-O-tetradecanoyl-phorbol 13-acetate, a PKC activator, increased Smad7 mRNA expression. These results suggest that Ang II enhances activated PSC proliferation by blocking autocrine TGF-beta1-mediated growth inhibition by inducing Smad7 expression via a PKC-dependent pathway.
Collapse
Affiliation(s)
- Kouji Hama
- Department of Gastroenterology, Jichi Medical School, Tochigi 329-0498, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hama K, Ohnishi H, Yasuda H, Ueda N, Mashima H, Satoh Y, Hanatsuka K, Kita H, Ohashi A, Tamada K, Sugano K. Angiotensin II stimulates DNA synthesis of rat pancreatic stellate cells by activating ERK through EGF receptor transactivation. Biochem Biophys Res Commun 2004; 315:905-11. [PMID: 14985098 DOI: 10.1016/j.bbrc.2004.01.155] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2004] [Indexed: 02/08/2023]
Abstract
Although angiotensin II (Ang II) is known to participate in pancreatic fibrosis, little is known as to the mechanism by which Ang II promotes pancreatic fibrosis. To elucidate the mechanism, we examined the action of Ang II on the proliferation of rat pancreatic stellate cells (PSCs) that play central roles in pancreatic fibrosis. Immunocytochemistry and Western blotting demonstrated that both Ang II type 1 and type 2 receptors were expressed in PSCs. [3H]Thymidine incorporation assay revealed that Ang II enhanced DNA synthesis in PSCs, which was blocked by Ang II type 1 receptor antagonist losartan. Western blotting using anti-phospho-epidermal growth factor (EGF) receptor and anti-phospho-extracellular signal regulated kinase (ERK) antibodies showed that Ang II-activated EGF receptor and ERK. Both EGF receptor kinase inhibitor AG1478 and MEK1 inhibitor PD98059 attenuated ERK activation and DNA synthesis enhanced by Ang II. These results indicate that Ang II stimulates PSC proliferation through EGF receptor transactivation-ERK activation pathway.
Collapse
Affiliation(s)
- Kouji Hama
- Department of Gastroenterology, Jichi Medical School, Tochigi 329-0498, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Ohnishi H, Miyata T, Yasuda H, Satoh Y, Hanatsuka K, Kita H, Ohashi A, Tamada K, Makita N, Iiri T, Ueda N, Mashima H, Sugano K. Distinct roles of Smad2-, Smad3-, and ERK-dependent pathways in transforming growth factor-beta1 regulation of pancreatic stellate cellular functions. J Biol Chem 2003; 279:8873-8. [PMID: 14688282 DOI: 10.1074/jbc.m309698200] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pancreatic stellate cells (PSCs) play a major role in promoting pancreatic fibrosis. Transforming growth factor-beta(1) (TGF-beta(1)) regulates PSC activation and proliferation in an autocrine manner. The intracellular signaling pathways of the regulation were examined in this study. Immunoprecipitation and immunocytochemistry revealed that Smad2, Smad3, and Smad4 were functionally expressed in PSCs. Adenovirus-mediated expression of Smad2, Smad3, or dominant-negative Smad2/3 did not alter TGF-beta(1) mRNA expression level or the amount of autocrine TGF-beta(1) peptide. However, expression of dominant-negative Smad2/3 inhibited PSC activation and enhanced their proliferation. Co-expression of Smad2 with dominant-negative Smad2/3 restored PSC activation inhibited by dominant-negative Smad2/3 expression without changing their proliferation. By contrast, co-expression of Smad3 with dominant-negative Smad2/3 attenuated PSC proliferation enhanced by dominant-negative Smad2/3 expression without altering their activation. Exogenous TGF-beta(1) increased TGFbeta(1) mRNA expression in PSCs. However, PD98059, a specific inhibitor of mitogen-activated protein kinase kinase (MEK1), inhibited ERK activation by TGF-beta(1), and consequently attenuated TGF-beta(1) enhancement of its own mRNA expression in PSCs. We propose that TGF-beta(1) differentially regulates PSC activation, proliferation, and TGF-beta(1) mRNA expression through Smad2-, Smad3-, and ERK-dependent pathways, respectively.
Collapse
Affiliation(s)
- Hirohide Ohnishi
- Department of Gastroenterology, Jichi Medical School, Tochigi 329-0498, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Welt C, Sidis Y, Keutmann H, Schneyer A. Activins, inhibins, and follistatins: from endocrinology to signaling. A paradigm for the new millennium. Exp Biol Med (Maywood) 2002; 227:724-52. [PMID: 12324653 DOI: 10.1177/153537020222700905] [Citation(s) in RCA: 225] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
It has been 70 years since the name inhibin was used to describe a gonadal factor that negatively regulated pituitary hormone secretion. The majority of this period was required to achieve purification and definitive characterization of inhibin, an event closely followed by identification and characterization of activin and follistatin (FS). In contrast, the last 15-20 years saw a virtual explosion of information regarding the biochemistry, physiology, and biosynthesis of these proteins, as well as identification of activin receptors, and a unique mechanism for FS action-the nearly irreversible binding and neutralization of activin. Many of these discoveries have been previously summarized; therefore, this review will cover the period from the mid 1990s to present, with particular emphasis on emerging themes and recent advances. As the field has matured, recent efforts have focused more on human studies, so the endocrinology of inhibin, activin, and FS in the human is summarized first. Another area receiving significant recent attention is local actions of activin and its regulation by both FS and inhibin. Because activin and FS are produced in many tissues, we chose to focus on a few particular examples with the most extensive experimental support, the pituitary and the developing follicle, although nonreproductive actions of activin and FS are also discussed. At the cellular level, it now seems that activin acts largely as an autocrine and/or paracrine growth factor, similar to other members of the transforming growh factor beta superfamily. As we discuss in the next section, its actions are regulated extracellularly by both inhibin and FS. In the final section, intracellular mediators and modulators of activin signaling are reviewed in detail. Many of these are shared with other transforming growh factor beta superfamily members as well as unrelated molecules, and in a number of cases, their physiological relevance to activin signal propagation remains to be elucidated. Nevertheless, taken together, recent findings suggest that it may be more appropriate to consider a new paradigm for inhibin, activin, and FS in which activin signaling is regulated extracellularly by both inhibin and FS whereas a number of intracellular proteins act to modulate cellular responses to these activin signals. It is therefore the balance between activin and all of its modulators, rather than the actions of any one component, that determines the final biological outcome. As technology and model systems become more sophisticated in the next few years, it should become possible to test this concept directly to more clearly define the role of activin, inhibin, and FS in reproductive physiology.
Collapse
Affiliation(s)
- Corrine Welt
- Reproductive Endocrine Unit and Endocrine Unit, Massachusetts General Hospital, Boston 02114, USA
| | | | | | | |
Collapse
|