1
|
Kaur S, Rajoria P, Chopra M. HDAC6: A unique HDAC family member as a cancer target. Cell Oncol (Dordr) 2022; 45:779-829. [PMID: 36036883 DOI: 10.1007/s13402-022-00704-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND HDAC6, a structurally and functionally distinct member of the HDAC family, is an integral part of multiple cellular functions such as cell proliferation, apoptosis, senescence, DNA damage and genomic stability, all of which when deregulated contribute to carcinogenesis. Among several HDAC family members known so far, HDAC6 holds a unique position. It differs from the other HDAC family members not only in terms of its subcellular localization, but also in terms of its substrate repertoire and hence cellular functions. Recent findings have considerably expanded the research related to the substrate pool, biological functions and regulation of HDAC6. Studies in HDAC6 knockout mice highlighted the importance of HDAC6 as a cell survival player in stressful situations, making it an important anticancer target. There is ample evidence stressing the importance of HDAC6 as an anti-cancer synergistic partner of many chemotherapeutic drugs. HDAC6 inhibitors have been found to enhance the effectiveness of conventional chemotherapeutic drugs such as DNA damaging agents, proteasome inhibitors and microtubule inhibitors, thereby highlighting the importance of combination therapies involving HDAC6 inhibitors and other anti-cancer agents. CONCLUSIONS Here, we present a review on HDAC6 with emphasis on its role as a critical regulator of specific physiological cellular pathways which when deregulated contribute to tumorigenesis, thereby highlighting the importance of HDAC6 inhibitors as important anticancer agents alone and in combination with other chemotherapeutic drugs. We also discuss the synergistic anticancer effect of combination therapies of HDAC6 inhibitors with conventional chemotherapeutic drugs.
Collapse
Affiliation(s)
- Sumeet Kaur
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Prerna Rajoria
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Madhu Chopra
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
2
|
Bochicchio MT, Di Battista V, Poggio P, Carrà G, Morotti A, Brancaccio M, Lucchesi A. Understanding Aberrant Signaling to Elude Therapy Escape Mechanisms in Myeloproliferative Neoplasms. Cancers (Basel) 2022; 14:cancers14040972. [PMID: 35205715 PMCID: PMC8870427 DOI: 10.3390/cancers14040972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/01/2023] Open
Abstract
Aberrant signaling in myeloproliferative neoplasms may arise from alterations in genes coding for signal transduction proteins or epigenetic regulators. Both mutated and normal cells cooperate, altering fragile balances in bone marrow niches and fueling persistent inflammation through paracrine or systemic signals. Despite the hopes placed in targeted therapies, myeloid proliferative neoplasms remain incurable diseases in patients not eligible for stem cell transplantation. Due to the emergence of drug resistance, patient management is often very difficult in the long term. Unexpected connections among signal transduction pathways highlighted in neoplastic cells suggest new strategies to overcome neoplastic cell adaptation.
Collapse
Affiliation(s)
- Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Valeria Di Battista
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Pietro Poggio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy;
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| | - Alessandro Lucchesi
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| |
Collapse
|
3
|
The p300/YY1/miR-500a-5p/HDAC2 signalling axis regulates cell proliferation in human colorectal cancer. Nat Commun 2019; 10:663. [PMID: 30737378 PMCID: PMC6368584 DOI: 10.1038/s41467-018-08225-3] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/21/2018] [Indexed: 12/12/2022] Open
Abstract
The biological role of miR-500a-5p has not yet been reported in the context of colorectal cancer (CRC). Here, we show that miR-500a-5p expression is decreased in CRC tissues compared with adjacent normal tissues. Low miR-500a-5p expression is associated with malignant progression. Moreover, transfection of CRC cells with miR-500a-5p induces G0/G1 cell cycle arrest and inhibits their growth and migration. Mechanistically, miR-500a-5p directly targets HDAC2 and inhibits HDAC2-mediated proliferation in CRC in nude mice. Furthermore, YY1 binds to the promoter of miR-500a-5p and negatively regulates its transcription. Restoration of miR-500a-5p expression is up-regulated via the p300/YY1/HDAC2 complex. Besides, therapeutic delivery of miR-500a-5p significantly suppresses tumour development in a xenograft tumour model and a HDAC2 inhibitor FK228-treated CRC model. Our studies demonstrate that miR-500a-5p functions as a tumour suppressor in CRC by targeting the p300/YY1/HDAC2 axis, which contributes to the development of and provides new potential candidates for CRC therapy.
Collapse
|
4
|
Chiba T, Maeda T, Kudo K. [Endogenous Serotonin and Milk Production Regulation in the Mammary Gland]. YAKUGAKU ZASSHI 2018; 138:829-836. [PMID: 29863055 DOI: 10.1248/yakushi.18-00003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intrinsic serotonin (5-hydroxytryptamine; 5-HT) synthesized within the mammary epithelium has an important physiological role in milk volume homeostasis in many species including mice, cows, and humans. During lactation, mammary epithelial cells activate 5-HT synthesis by tryptophan hydroxylase 1 (TPH1). TPH1 catalyzes the rate-limiting step in 5-HT biosynthesis within mammary glands. 5-HT synthesized in mammary glands is released into both the apical (milk) and basolateral spaces by a vesicular monoamine transporter. 5-HT released into milk is incorporated by the apical membrane-expressed serotonin reuptake transporter and degraded by the monoamine oxidase A enzyme. Suckling maintains 5-HT at low levels in milk. When the mammary gland becomes filled with milk, 5-HT provides a negative feedback signal that suppresses further milk synthesis in the mammary epithelium. Our research, using human mammary epithelial MCF-12A cells, shows that the expression of β-casein, a differentiation marker, is suppressed via 5-HT-mediated inhibition of signal transducer and activator of transcription 5. Additionally, our results show that reduced β-casein expression in MCF-12A cells is associated with 5-HT7 receptor expression. Furthermore, we show that 5-HT7 receptor-mediated suppression of β-casein expression is involved in the activation of protein kinase A and protein-tyrosine phosphatase 1B. Thus, this mechanism might be associated with the feedback signals by 5-HT within the mammary epithelium. Hence, further research that builds on our findings should include the elucidation of the physiological roles of 5-HT present in milk synthesized by mammary epithelial cells in vivo and its effects on nursing infants.
Collapse
Affiliation(s)
- Takeshi Chiba
- Department of Pharmaceutics and Clinical Practice, School of Pharmacy, Iwate Medical University.,Department of Pharmacy, Iwate Medical University Hospital
| | - Tomoji Maeda
- Department of Pharmacotherapy, Nihon Pharmaceutical University
| | - Kenzo Kudo
- Department of Pharmaceutics and Clinical Practice, School of Pharmacy, Iwate Medical University.,Department of Pharmacy, Iwate Medical University Hospital
| |
Collapse
|
5
|
The interplay between histone deacetylases and rho kinases is important for cancer and neurodegeneration. Cytokine Growth Factor Rev 2017; 37:29-45. [PMID: 28606734 DOI: 10.1016/j.cytogfr.2017.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/18/2017] [Accepted: 05/21/2017] [Indexed: 12/24/2022]
Abstract
Rho associated coiled-coil containing kinases (ROCKs) respond to defined extra- and intracellular stimuli to control cell migration, cell proliferation, and apoptosis. Histone deacetylases (HDACs) are epigenetic modifiers that regulate nuclear and cytoplasmic signaling through the deacetylation of histones and non-histone proteins. ROCK and HDAC functions are important compounds of basic and applied research interests. Recent evidence suggests a physiologically important interplay between HDACs and ROCKs in various cells and organisms. Here we summarize the crosstalk between these enzymatic families and its implications for cancer and neurodegeneration.
Collapse
|
6
|
Chiba T, Maeda T, Sanbe A, Kudo K. Serotonin suppresses β-casein expression via PTP1B activation in human mammary epithelial cells. Biochem Biophys Res Commun 2016; 473:323-328. [DOI: 10.1016/j.bbrc.2016.03.103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 03/21/2016] [Indexed: 12/19/2022]
|
7
|
Chiba T, Kimura S, Takahashi K, Morimoto Y, Maeda T, Sanbe A, Ueda H, Kudo K. Serotonin regulates β-casein expression via 5-HT7 receptors in human mammary epithelial MCF-12A cells. Biol Pharm Bull 2015; 38:448-53. [PMID: 25757927 DOI: 10.1248/bpb.b14-00723] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that serotonin (5-hydroxytryptamine; 5-HT) suppresses β-casein expression, a differentiation marker in mammary epithelial cells, via inhibition of the signal transducer and activator of transcription 5 (STAT5) phosphorylation in the human mammary epithelial cell line, MCF-12A. In this study, we investigated the expression pattern of the different 5-HT receptor subtypes in MCF-12A cells, and identified the receptors involved in 5-HT-mediated suppression of β-casein protein expression. β-Casein mRNA expression was inhibited by 30 µM 5-HT in a time-dependent manner. Treatment with 30 µM 5-HT for 72 h decreased β-casein protein levels and STAT5 phosphorylation (pSTAT5). The cells expressed four 5-HT receptors subtypes (5-HTR1D, 2B, 3A, and 7) at the mRNA and protein level, and their expression was elevated by prolactin (PRL) treatment. Additionally, the mRNA levels of 5-HTR1D and 5-HTR7 were significantly higher than the other 5-HT receptors in the cells. Tryptophan hydroxylase 1 mRNA was detectable in the cells in the absence of PRL, and PRL treatment significantly increased its expression. β-Casein and pSTAT5/STAT5 levels in the cells co-treated with 5-HT and a selective 5-HTR1D inhibitor, BRL15572, were equal to those observed in cells treated with 5-HT alone. However, in the cells co-treated with 5-HT and a selective 5-HTR7 inhibitor, SB269970, β-casein and pSTAT5/STAT5 levels increased in a SB269970 concentration-dependent manner. In conclusion, we showed that 5-HT regulates β-casein expression via 5-HTR7 in MCF-12A human mammary epithelial cells.
Collapse
Affiliation(s)
- Takeshi Chiba
- Department of Clinical Pharmaceutics and Pharmacy Practice, School of Pharmacy, Iwate Medical University
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Yin H, Hou X, Tao T, Lv X, Zhang L, Duan W. Neurite outgrowth resistance to rho kinase inhibitors in PC12 Adh cell. Cell Biol Int 2015; 39:563-76. [PMID: 25571866 DOI: 10.1002/cbin.10423] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 12/26/2014] [Indexed: 01/21/2023]
Abstract
Rho kinase (ROCK) inhibitor is a promising agent for neural injury disorders, which mechanism is associated with neurite outgrowth. However, neurite outgrowth resistance occurred when PC12 Adh cell was treated with ROCK inhibitors for a longer time. PC12 Adh cells were treated with ROCK inhibitor Y27632 or NGF for different durations. Neurite outgrowth resistance occurred when PC12 Adh cell exposed to Y27632 (33 µM) for 3 or more days, but not happen when exposed to nerve growth factor (NGF, 100 ng/mL). The gene expression in the PC12 Adh cells treated with Y27632 (33 µM) or NGF (100 ng/mL) for 2 or 4 days was assayed by gene microarray, and the reliability of the results were confirmed by real-time RT-PCR. Cluster analysis proved that the gene expression profile of PC12 Adh cell treated with Y27632 for 4 days was different from that treated with Y27632 for 2 days and those treated with NGF for 2 and 4 days, respectively. Pathway analysis hinted that the neurite outgrowth resistance could be associated with up-regulation of inflammatory pathways, especially rno04610 (complement and coagulation cascades), and down-regulation of cell cycle pathways, especially rno04110.
Collapse
Affiliation(s)
- Hua Yin
- Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Traditional Chinese Medicine, 1076, Yuhua Road, University City of Chenggong, Kunming, 650500, China
| | | | | | | | | | | |
Collapse
|
9
|
D'Amato L, Dell'Aversana C, Conte M, Ciotta A, Scisciola L, Carissimo A, Nebbioso A, Altucci L. ARHGEF3 controls HDACi-induced differentiation via RhoA-dependent pathways in acute myeloid leukemias. Epigenetics 2015; 10:6-18. [PMID: 25494542 DOI: 10.4161/15592294.2014.988035] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Altered expression and activity of histone deacetylases (HDACs) have been correlated with tumorigenesis. Inhibitors of HDACs (HDACi) induce acetylation of histone and non-histone proteins affecting gene expression, cell cycle progression, cell migration, terminal differentiation and cell death. Here, we analyzed the regulation of ARHGEF3, a RhoA-specific guanine nucleotide exchange factor, by the HDACi MS275 (entinostat). MS275 is a well-known benzamide-based HDACi, which induces differentiation of the monoblastic-like human histiocytic lymphoma cell line U937 to monocytes/macrophages. Incubation of U937 cells with MS275 resulted in an up regulation of ARHGEF3, followed by a significant enhancement of the marker of macrophage differentiation CD68. ARHGEF3 protein is primarily nuclear, but MS275 treatment rapidly induced its translocation into the cytoplasm. ARHGEF3 cytoplasmic localization is associated with activation of the RhoA/Rho-associated Kinase (ROCK) pathway. In addition to cytoskeletal rearrangements orchestrated by RhoA, we showed that ARHGEF3/RhoA-dependent signals involve activation of SAPK/JNK and then Elk1 transcription factor. Importantly, MS275-induced CD68 expression was blocked by exposure of U937 cells to exoenzyme C3 transferase and Y27632, inhibitors of Rho and ROCK respectively. Moreover, ARHGEF3 silencing prevented RhoA activation leading to a reduction in SAPK/JNK phosphorylation, Elk1 activation and CD68 expression, suggesting a crucial role for ARHGEF3 in myeloid differentiation. Taken together, our results demonstrate that ARHGEF3 modulates acute myeloid leukemia differentiation through activation of RhoA and pathways directly controlled by small GTPase family proteins. The finding that GEF protein modulation by HDAC inhibition impacts on cell differentiation may be important for understanding the antitumor mechanism(s) by which HDACi treatment stimulates differentiation in cancer.
Collapse
Affiliation(s)
- Loredana D'Amato
- a Dipartimento di Biochimica, Biofisica e Patologia Generale ; Seconda Università di Napoli ; Napoli , Italy
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Lin G, LaPensee CR, Qin ZS, Schwartz J. Reciprocal occupancy of BCL6 and STAT5 on Growth Hormone target genes: contrasting transcriptional outcomes and promoter-specific roles of p300 and HDAC3. Mol Cell Endocrinol 2014; 395:19-31. [PMID: 25088465 PMCID: PMC4176921 DOI: 10.1016/j.mce.2014.07.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 06/30/2014] [Accepted: 07/28/2014] [Indexed: 12/30/2022]
Abstract
Expression of the Growth Hormone (GH)-stimulated gene Socs2 (Suppressor of Cytokine Signaling 2) is mediated by the transcription activator STAT5 (Signal Transducer and Activator of Transcription 5) and the transcription repressor BCL6 (B-Cell Lymphoma 6). ChIP-Sequencing identified Cish (Cytokine-Inducible SH2-containing protein) and Bcl6 as having similar patterns of reciprocal occupancy by BCL6 and STAT5 in response to GH, though GH stimulates Cish and inhibits Bcl6 expression. The co-activator p300 occupied Socs2, Cish and Bcl6 promoters, and enhanced STAT5-mediated activation of Socs2 and Cish. In contrast, on Bcl6, p300 functioned as a repressor and inhibited in conjunction with STAT5 or BCL6. The co-repressor HDAC3 (Histone deacetylase 3) inhibited the Socs2, Cish and Bcl6 promoters in the presence of STAT5. Thus transcriptional outcomes on GH-regulated genes occupied by BCL6 and STAT5 are determined in a promoter-specific fashion by co-regulatory proteins which mediate the distinction between activating and repressive transcription factors.
Collapse
Affiliation(s)
- Grace Lin
- Cellular & Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Christopher R LaPensee
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Zhaohui S Qin
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Jessica Schwartz
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
11
|
Orgaz JL, Pandya P, Dalmeida R, Karagiannis P, Sanchez-Laorden B, Viros A, Albrengues J, Nestle FO, Ridley AJ, Gaggioli C, Marais R, Karagiannis SN, Sanz-Moreno V. Diverse matrix metalloproteinase functions regulate cancer amoeboid migration. Nat Commun 2014; 5:4255. [PMID: 24963846 PMCID: PMC4118761 DOI: 10.1038/ncomms5255] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 05/29/2014] [Indexed: 12/17/2022] Open
Abstract
Rounded-amoeboid cancer cells use actomyosin contractility driven by Rho-ROCK and JAK-STAT3 to migrate efficiently. It has been suggested that rounded-amoeboid cancer cells do not require matrix metalloproteinases (MMPs) to invade. Here we compare MMP levels in rounded-amoeboid and elongated-mesenchymal melanoma cells. Surprisingly, we find that rounded-amoeboid melanoma cells secrete higher levels of several MMPs, including collagenase MMP-13 and gelatinase MMP-9. As a result, rounded-amoeboid melanoma cells degrade collagen I more efficiently than elongated-mesenchymal cells. Furthermore, using a non-catalytic mechanism, MMP-9 promotes rounded-amoeboid 3D migration through regulation of actomyosin contractility via CD44 receptor. MMP-9 is upregulated in a panel of rounded-amoeboid compared with elongated-mesenchymal melanoma cell lines and its levels are controlled by ROCK-JAK-STAT3 signalling. MMP-9 expression increases during melanoma progression and it is particularly prominent in the invasive fronts of lesions, correlating with cell roundness. Therefore, rounded-amoeboid cells use both catalytic and non-catalytic activities of MMPs for invasion.
Collapse
Affiliation(s)
- Jose L Orgaz
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Pahini Pandya
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Rimple Dalmeida
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Panagiotis Karagiannis
- NIHR Biomedical Research Centre at Guy's and St Thomas' Hospitals, Cutaneous Medicine and Immunotherapy Unit, St John's Institute of Dermatology, Division of Genetics and Molecular Medicine at Guy's Hospital, King's College London, London SE1 9RT, UK
| | - Berta Sanchez-Laorden
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Amaya Viros
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Jean Albrengues
- INSERM, U1081, CNRS, UMR7284, Institute for Research on Cancer and Aging in Nice (IRCAN), Faculté de Médecine, University of Nice Sophia-Antipolis, 28 Avenue de Valombrose, F-06107 Nice, France
| | - Frank O Nestle
- NIHR Biomedical Research Centre at Guy's and St Thomas' Hospitals, Cutaneous Medicine and Immunotherapy Unit, St John's Institute of Dermatology, Division of Genetics and Molecular Medicine at Guy's Hospital, King's College London, London SE1 9RT, UK
| | - Anne J Ridley
- Cell Signalling in Invasion and Motility Team, Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Cedric Gaggioli
- INSERM, U1081, CNRS, UMR7284, Institute for Research on Cancer and Aging in Nice (IRCAN), Faculté de Médecine, University of Nice Sophia-Antipolis, 28 Avenue de Valombrose, F-06107 Nice, France
| | - Richard Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Sophia N Karagiannis
- NIHR Biomedical Research Centre at Guy's and St Thomas' Hospitals, Cutaneous Medicine and Immunotherapy Unit, St John's Institute of Dermatology, Division of Genetics and Molecular Medicine at Guy's Hospital, King's College London, London SE1 9RT, UK
| | - Victoria Sanz-Moreno
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
12
|
Yu L, Weng X, Liang P, Dai X, Wu X, Xu H, Fang M, Fang F, Xu Y. MRTF-A mediates LPS-induced pro-inflammatory transcription by interacting with the COMPASS complex. J Cell Sci 2014; 127:4645-57. [DOI: 10.1242/jcs.152314] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Chronic inflammation underscores the pathogenesis of a range of human diseases. Lipopolysaccharide (LPS) elicits strong pro-inflammatory response in macrophages via the transcription factor NF-κB. The epigenetic mechanism underlying LPS-induced pro-inflammatory transcription is not completely appreciated. Herein we describe a role for myocardin related transcription factor A, or MRTF-A, in this process. MRTF-A over-expression potentiated while MRTF-A silencing dampened NF-κB dependent pro-inflammatory transcription. MRTF-A deficiency also alleviated the synthesis of pro-inflammatory mediators in a mouse model of colitis. LPS promoted the recruitment of MRTF-A to the promoters of pro-inflammatory genes in a NF-κB dependent manner. Reciprocally, MRTF-A influenced the nuclear enrichment and target binding of NF-κB. Mechanistically, MRTF-A was necessary for the accumulation of active histone modifications on NF-κB target promoters by communicating with the histone H3K4 methyltransferase complex (COMPASS). Silencing of individual members of COMPASS, including ASH2, WDR5, and SET1, down-regulated the production of pro-inflammatory mediators and impaired the NF-κB kinetics. In summary, our work has uncovered a previously unknown function for MRTF-A and provided insights into the rationalized development of anti-inflammatory therapeutic strategies.
Collapse
|
13
|
Huang H, Lee DH, Zabolotny JM, Kim YB. Metabolic actions of Rho-kinase in periphery and brain. Trends Endocrinol Metab 2013; 24:506-14. [PMID: 23938132 PMCID: PMC3783562 DOI: 10.1016/j.tem.2013.06.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Revised: 06/11/2013] [Accepted: 06/13/2013] [Indexed: 01/01/2023]
Abstract
Obesity has increased at an alarming rate in recent years and is now a worldwide public health problem. Elucidating the mechanisms behind the metabolic dysfunctions associated with obesity is of high priority. The metabolic function of Rho-kinase (Rho-associated coiled-coil-containing kinase; ROCK) has been the subject of a great deal of investigation in metabolic-related diseases. It appears that inhibition of ROCK activity is beneficial for the treatment of a wide range of cardiovascular-related diseases. However, recent studies with genetic models of ROCK demonstrate that ROCK plays a positive role in insulin and leptin signaling. Here we discuss the newly identified functions of ROCK in regulating glucose and energy metabolism, with particular emphasis on metabolic actions of insulin and leptin.
Collapse
Affiliation(s)
- Hu Huang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, MA 02215, USA
| | - Dae-Ho Lee
- Department of Internal Medicine, School of Medicine, Wonkwang University, Iksan, Korea 570-749
| | - Janice M Zabolotny
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, MA 02215, USA
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, MA 02215, USA
- Lee Gil Ya Cancer & Diabetes Institute, Graduate Schools of Medicine, Gachon University of Medicine & Science, Incheon, Korea 406-799
- Corresponding author: Young-Bum Kim, Ph.D., Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, Phone: (617) 735-3216, Fax: (617) 735-3323,
| |
Collapse
|
14
|
Peelman F, Tavernier J. ROCKing the JAKs. JAKSTAT 2013; 2:e24074. [PMID: 24069551 PMCID: PMC3772103 DOI: 10.4161/jkst.24074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 02/21/2013] [Accepted: 02/21/2013] [Indexed: 11/19/2022] Open
Abstract
The endocrine cytokine leptin is mainly secreted by white adipose tissue and plasma leptin levels positively correlate with body fat mass. Via its action on neurons in the hypothalamic arcuate nucleus (ARC), leptin regulates body weight by stimulating energy expenditure and inhibiting food intake. The main signaling pathway of the leptin receptor is the JAK2-STAT3 pathway. A recent publication of Huang et al. in Nature Neuroscience shows that leptin’s hypothalamic signaling via JAK2 requires the kinase ROCK1 (Rho-associated coiled-coil-containing protein kinase 1). ROCK1 directly phosphorylates JAK2, and this phosphorylation is required for the JAK2-STAT3 pathway of the leptin receptor. Gene deletion of ROCK1 in ARC neurons targeted by leptin makes these neurons less sensitive to leptin. This is reflected by a pronounced weight gain with hyperphagia, reduced locomotor activity, and increased fat accumulation. In this article we comment on the article of Huang et al. While the mechanism of ROCK1 activation in the neurons remains uncharacterized for the moment, a literature survey suggests that the interplay between ROCK1 and a JAK kinase may be a common theme for receptors that function via a JAK2 and even for other members of the JAK kinase family.
Collapse
Affiliation(s)
- Frank Peelman
- Department of Medical Protein Research; VIB; Ghent, Belgium ; Department of Biochemistry; Faculty of Medicine and Health Sciences; Ghent University; Ghent, Belgium
| | | |
Collapse
|
15
|
Salvianolic acid B lowers portal pressure in cirrhotic rats and attenuates contraction of rat hepatic stellate cells by inhibiting RhoA signaling pathway. J Transl Med 2012; 92:1738-48. [PMID: 22986787 DOI: 10.1038/labinvest.2012.113] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The contraction of hepatic stellate cells (HSCs) has a critical role in the regulation of intrahepatic vascular resistance and portal hypertension. Previous studies have confirmed that salvianolic acid B (Sal B) is effective against liver fibrosis. In the present study, we evaluated the effect of Sal B on portal hypertension and on HSCs contractility. Liver cirrhosis was induced in rats by peritoneal injection of dimethylnitrosamine and the portal pressure was measured. HSCs contraction was evaluated by collagen gel contraction assay. Glycerol-urea gel electrophoresis was performed to determine the phosphorylation of myosin light chain 2 (MLC2). F-actin stress fiber polymerization was detected by fluorescein isothiocyanate-labeled phalloidin. Intracellular Ca(2+) and RhoA signaling activation were also measured. Sal B effectively reduced the portal pressure in DMN-induced cirrhotic rats. It decreased the contraction by endothelin-1 (ET-1)-activated HSCs by ∼66.5% and caused the disassembly of actin stress fibers and MLC2 dephosphorylation. Although Sal B reduced ET-1-induced intracellular Ca(2+) increase, blocking Ca(2+) increase completely by BAPTA-AM, a Ca(2+) chelator, barely affected the magnitude of contraction. Sal B decreased ET-1-induced RhoA and Rho-associated coiled coil-forming protein kinase (ROCK) II activation by 66.84% and by 76.79%, respectively, and inhibited Thr(696) phosphorylation of MYPT1 by 80.09%. In vivo, Sal B lowers the portal pressure in rats with DMN-induced cirrhosis. In vitro, Sal B attenuates ET-1-induced HSCs contraction by inhibiting the activation of RhoA and ROCK II and the downstream MYPT1 phosphorylation at Thr(696). We consider Sal B a potential candidate for the pharmacological treatment of portal hypertension.
Collapse
|
16
|
Fonseca AV, Corbeil D. The hematopoietic stem cell polarization and migration: A dynamic link between RhoA signaling pathway, microtubule network and ganglioside-based membrane microdomains. Commun Integr Biol 2011; 4:201-4. [PMID: 21655440 DOI: 10.4161/cib.4.2.14419] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 12/06/2010] [Indexed: 11/19/2022] Open
Abstract
The polarization and migration of eukaryotic cells are fundamental processes for the development and maintenance of a tissue. These aspects gain especial interest when it comes to stem and progenitor cells in the way that their manipulation might open new avenues in regenerative therapy. In recent years, novel biological facets of migrating hematopoietic stem cells were revealed by several groups, including ours. Among these features, the polarization of their membranous (proteins and lipids) and cytoplasmic constituents, which leads to the formation of a specialized sub-cellular structure located at the rear pole-the uropod-has gained increasing interest. In a new study we have demonstrated that such phenomena involve a coordinated mechanism between Rho GTPase signaling and the microtubule network. Specifically, our results based on the use of synthetic inhibitors and RNA interference suggest that the activity of RhoA and its effector ROCK I is indispensable for cell polarization and the active reorganization of microtubules that are required for migration.
Collapse
Affiliation(s)
- Ana-Violeta Fonseca
- Tissue Engineering Laboratories (BIOTEC) and DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden (CRTD); Technische Universität Dresden; Dresden, Germany
| | | |
Collapse
|
17
|
Takesono A, Heasman SJ, Wojciak-Stothard B, Garg R, Ridley AJ. Microtubules regulate migratory polarity through Rho/ROCK signaling in T cells. PLoS One 2010; 5:e8774. [PMID: 20098744 PMCID: PMC2808253 DOI: 10.1371/journal.pone.0008774] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 12/29/2009] [Indexed: 01/03/2023] Open
Abstract
Background Migrating leukocytes normally have a polarized morphology with an actin-rich lamellipodium at the front and a uropod at the rear. Microtubules (MTs) are required for persistent migration and chemotaxis, but how they affect cell polarity is not known. Methodology/Principal Findings Here we report that T cells treated with nocodazole to disrupt MTs are unable to form a stable uropod or lamellipodium, and instead often move by membrane blebbing with reduced migratory persistence. However, uropod-localized receptors and ezrin/radixin/moesin proteins still cluster in nocodazole-treated cells, indicating that MTs are required specifically for uropod stability. Nocodazole stimulates RhoA activity, and inhibition of the RhoA target ROCK allows nocodazole-treated cells to re-establish lamellipodia and uropods and persistent migratory polarity. ROCK inhibition decreases nocodazole-induced membrane blebbing and stabilizes MTs. The myosin inhibitor blebbistatin also stabilizes MTs, indicating that RhoA/ROCK act through myosin II to destabilize MTs. Conclusions/Significance Our results indicate that RhoA/ROCK signaling normally contributes to migration by affecting both actomyosin contractility and MT stability. We propose that regulation of MT stability and RhoA/ROCK activity is a mechanism to alter T-cell migratory behavior from lamellipodium-based persistent migration to bleb-based migration with frequent turning.
Collapse
Affiliation(s)
- Aya Takesono
- University College London, Department of Biochemistry and Molecular Biology and Ludwig Institute for Cancer Research, London, United Kingdom
- School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Sarah J. Heasman
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Beata Wojciak-Stothard
- Department of Experimental Medicine and Toxicology, Imperial College London, London, United Kingdom
| | - Ritu Garg
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Anne J. Ridley
- University College London, Department of Biochemistry and Molecular Biology and Ludwig Institute for Cancer Research, London, United Kingdom
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
18
|
Duan WG, Shang J, Jiang ZZ, Yao JC, Yun Y, Yan M, Shu B, Lin Q, Yu ZP, Zhang LY. Rho kinase inhibitor Y-27632 down-regulates norepinephrine synthesis and release in PC12 cells. Basic Clin Pharmacol Toxicol 2009; 104:434-40. [PMID: 19489787 DOI: 10.1111/j.1742-7843.2008.00314.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Rho kinase inhibition is beneficial for neurite outgrowth and nerve disorders, and the Rho kinase inhibitors have been regarded as promising agents to treat neural diseases. The main aim of the study was to elucidate how Rho kinase inhibitor Y-27632 regulates neurotransmitter norepinephrine synthesis and release in PC12 cells when neurite outgrowth was induced. PC12 cells were treated with Y-27632 for 6 days. The amount of norepinephrine synthesized in PC12 cells and the amount released evoked by acetylcholine or by KCl were determined by norepinephrine enzyme-linked immunosorbent assay kits. The results showed that the amount of norepinephrine both synthesized and released was down-regulated with a concentration-dependent relationship. Further results of Western blotting found that the protein expression of tyrosine hydroxylase and synapsin I (especially its active form, synapsin I phosphoSer603) was also down-regulated, which were directly related to synthesis and release of norepinephrine, respectively. All the results suggest that Y-27632 is able to down-regulate norepinephrine synthesis and release, the direct mechanism of which may be associated with down-regulation on expression of some proteins, including tyrosine hydroxylase and synapsin I.
Collapse
Affiliation(s)
- Wei-Gang Duan
- Jiangsu Center for Drug Screening, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Mohankumar KM, Perry JK, Kannan N, Kohno K, Gluckman PD, Emerald BS, Lobie PE. Transcriptional activation of signal transducer and activator of transcription (STAT) 3 and STAT5B partially mediate homeobox A1-stimulated oncogenic transformation of the immortalized human mammary epithelial cell. Endocrinology 2008; 149:2219-29. [PMID: 18276758 DOI: 10.1210/en.2007-1320] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We have previously demonstrated that the p44/42 MAPK pathway is one pathway involved in homeobox (HOX) A1-stimulated oncogenesis. However, inhibition of MAPK kinase 1 does not completely prevent HOXA1-stimulated oncogenic transformation, suggesting the involvement of additional signal transduction pathways. Here, we report that forced expression of HOXA1 in immortalized human mammary epithelial cells significantly increased levels of signal transducer and activator of transcription (STAT) 3, 5A, and 5B mRNA by transcriptional up-regulation. The protein levels of STAT3 and 5B, but not STAT5A, and protein phosphorylation levels of STAT3 and 5B were significantly increased by forced expression of HOXA1. Forced expression of STAT3 or STAT5B was sufficient to transform oncogenically an immortalized human mammary epithelial cell line. Accordingly, inhibition of STAT3 or STAT5B activity with dominant negative STAT3 or STAT5B abrogated the ability of HOXA1 to stimulate cell proliferation, survival, oncogenic transformation, and generation of large disorganized multiacinar structures in three-dimensional culture. These results suggest that HOXA1 partially mediates oncogenic transformation of the immortalized human mammary epithelial cell through modulation of the STAT3 and STAT5B pathways.
Collapse
Affiliation(s)
- Kumarasamypet M Mohankumar
- The Liggins Institute and National Research Centre for Growth and Development, University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | | |
Collapse
|
20
|
Shafiei F, Rahnama F, Pawella L, Mitchell MD, Gluckman PD, Lobie PE. DNMT3A and DNMT3B mediate autocrine hGH repression of plakoglobin gene transcription and consequent phenotypic conversion of mammary carcinoma cells. Oncogene 2008; 27:2602-12. [PMID: 17998942 DOI: 10.1038/sj.onc.1210917] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 10/05/2007] [Accepted: 10/08/2007] [Indexed: 12/23/2022]
Abstract
Directed by microarray analyses, we report that autocrine human growth hormone (hGH) increased the mRNA and protein expression of DNA methyltransferase 1 (DNMT1), DNMT3A and DNMT3B in mammary carcinoma cells. Autocrine hGH stimulation of DNMT3A and DNMT3B expression was mediated by JAK2 and Src kinases, and treatment of mammary carcinoma cells with the DNMT inhibitor, 5'-aza-2'-deoxycytidine (AZA), abrogated autocrine hGH-stimulated cellular proliferation, apoptosis and anchorage-independent growth. AZA reversed the epitheliomesenchymal transition of mammary carcinoma cells induced by autocrine hGH, to an epithelioid morphology and abrogated cell migration stimulated by autocrine hGH. Autocrine hGH-stimulated hypermethylation of the first exon of the PLAKOGLOBIN gene and AZA abrogated the ability of autocrine hGH to repress plakoglobin gene transcription. Small interfering RNA (siRNA)-mediated depletion of the individual DNMT molecules did not release autocrine hGH repression of PLAKOGLOBIN promoter activity nor did individual DNMT depletion affect autocrine hGH-stimulated migration. However, concomitant siRNA-mediated depletion of both DNMT3A and DNMT3B abrogated hypermethylation of the PLAKOGLOBIN gene stimulated by autocrine hGH and subsequent repression of plakoglobin gene transcription and increased cell migration. Thus, the autocrine hGH-stimulated increases in DNMT3A and DNMT3B expression mediate repression of plakoglobin gene transcription by direct hypermethylation of its promoter and consequent phenotypic conversion of mammary carcinoma cells. Autocrine hGH, therefore, utilizes DNA methylation as a mechanism to exert its oncogenic effects in mammary carcinoma cells.
Collapse
MESH Headings
- Antimetabolites, Antineoplastic/pharmacology
- Apoptosis/drug effects
- Apoptosis/genetics
- Autocrine Communication/drug effects
- Autocrine Communication/genetics
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Cell Movement/drug effects
- Cell Movement/genetics
- Cell Proliferation/drug effects
- Cytidine Monophosphate/analogs & derivatives
- Cytidine Monophosphate/pharmacology
- DNA (Cytosine-5-)-Methyltransferase 1
- DNA (Cytosine-5-)-Methyltransferases/biosynthesis
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA Methylation/drug effects
- DNA Methyltransferase 3A
- DNA, Neoplasm/genetics
- DNA, Neoplasm/metabolism
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Growth Hormone/antagonists & inhibitors
- Growth Hormone/genetics
- Growth Hormone/metabolism
- Humans
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Phenotype
- Promoter Regions, Genetic/genetics
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Neoplasm/biosynthesis
- RNA, Neoplasm/genetics
- RNA, Small Interfering/genetics
- gamma Catenin/biosynthesis
- gamma Catenin/genetics
- DNA Methyltransferase 3B
Collapse
Affiliation(s)
- F Shafiei
- National Research Centre for Growth and Development and the Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | | | | | | | | |
Collapse
|
21
|
Wang X, Carré W, Saxton AM, Cogburn LA. Manipulation of thyroid status and/or GH injection alters hepatic gene expression in the juvenile chicken. Cytogenet Genome Res 2007; 117:174-88. [PMID: 17675858 DOI: 10.1159/000103178] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2006] [Accepted: 01/19/2007] [Indexed: 11/19/2022] Open
Abstract
Both thyroid hormone (T3) and growth hormone (GH) are important regulators of somatic growth in birds and mammals. Although T3-mediated gene transcription is well known, the molecular basis of T3 interaction with GH on growth and development of birds remains unknown. In earlier studies, we discovered that exogenous GH alone increased accumulation of visceral fat in young chickens, while the combination of GH injections and dietary T3 worked synergistically to deplete body fat. In the present study, cDNA microarray and quantitative RT-PCR analyses enabled us to examine hepatic gene expression in young chickens after chronic manipulation of thyroid status and GH injection alone or in combination with T3. Thyroid status modulates expression of common and unique sets of genes involved in a wide range of molecular functions (i.e., energy metabolism, storage and transport, signal transduction, protein turnover and drug detoxification). Hepatic expression of 35 genes was altered by hypothyroidism (e.g., ADFP, ANGPTL3, GSTalpha, CAT, PPARG, HMGCL, GHR, IGF1, STAT3, THRSPalpha), whereas hyperthyroidism affected expression of another cluster of 13 genes (e.g., IGFBP1, KHK, LDHB, BAIA2L1, SULT1B, TRIAD3). Several genes were identified which have not been previously ascribed as T3 responsive (e.g., DEFB9, EPS8L2, ARHGAP1, LASS2, INHBC). Exogenous GH altered expression of 17 genes (e.g., CCAR1, CYP2C45, GYS2, ENOB, HK1, FABP1, SQLE, SOCS2, UPG2). The T3+GH treatment depleted the greatest amount of body fat, where 34 differentially expressed genes were unique to this group (e.g., C/EBP, CDC42EP1, SYDE2, PCK2, PIK4CA, TH1L, GPT2, BHMT). The marked reduction in body fat brought about by the T3+GH synergism could involve modulation of hormone signaling via altered activity of the Ras superfamily of molecular switches, which control diverse biological processes. In conclusion, this study provides the first global analysis of endocrine (T3 and GH) regulation of hepatic gene transcription in the chicken.
Collapse
Affiliation(s)
- X Wang
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19717, USA
| | | | | | | |
Collapse
|
22
|
Boyault C, Sadoul K, Pabion M, Khochbin S. HDAC6, at the crossroads between cytoskeleton and cell signaling by acetylation and ubiquitination. Oncogene 2007; 26:5468-76. [PMID: 17694087 DOI: 10.1038/sj.onc.1210614] [Citation(s) in RCA: 288] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Histone deacetylase 6 (HDAC6) is a unique enzyme with specific structural and functional features. It is actively or stably maintained in the cytoplasm and is the only member, within the histone deacetylase family, that harbors a full duplication of its deacetylase homology region followed by a specific ubiquitin-binding domain at the C-terminus end. Accordingly, this deacetylase functions at the heart of a cellular regulatory mechanism capable of coordinating various cellular functions largely relying on the microtubule network. Moreover, HDAC6 action as a regulator of the HSP90 chaperone activity adds to the multifunctionality of the protein, and allows us to propose a critical role for HDAC6 in mediating and coordinating various cellular events in response to different stressful stimuli.
Collapse
Affiliation(s)
- C Boyault
- INSERM, U823, Equipe Epigénétique et Signalisation Cellulaire, Institut Albert Bonniot, Université Joseph Fourier, Domaine de la Merci, Grenoble, La Tronche Cedex, France
| | | | | | | |
Collapse
|
23
|
Ceseña TI, Cui TX, Piwien-Pilipuk G, Kaplani J, Calinescu AA, Iñiguez-Lluhí JA, Kwok R, Schwartz J. Multiple mechanisms of growth hormone-regulated gene transcription. Mol Genet Metab 2007; 90:126-33. [PMID: 17129742 PMCID: PMC1986646 DOI: 10.1016/j.ymgme.2006.10.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2006] [Revised: 10/21/2006] [Accepted: 10/21/2006] [Indexed: 12/21/2022]
Abstract
Diverse physiological actions of growth hormone (GH) are mediated by changes in gene transcription. Transcription can be regulated at several levels, including post-translational modification of transcription factors, and formation of multiprotein complexes involving transcription factors, co-regulators and additional nuclear proteins; these serve as targets for regulation by hormones and signaling pathways. Evidence that GH regulates transcription at multiple levels is exemplified by analysis of the proto-oncogene c-fos. Among the GH-regulated transcription factors on c-fos, C/EBPbeta appears to be key, since depletion of C/EBPbeta by RNA interference blocks the stimulation of c-fos by GH. The phosphorylation state of C/EBPbeta and its ability to activate transcription are regulated by GH through MAPK and PI3K/Akt-mediated signaling cascades. The acetylation of C/EBPbeta also contributes to its ability to activate c-fos transcription. These and other post-translational modifications of C/EBPbeta appear to be integrated for regulation of transcription by GH. The formation of nuclear proteins into complexes associated with DNA-bound transcription factors is also regulated by GH. Both C/EBPbeta and the co-activator p300 are recruited to c-fos in response to GH, altering c-fos promoter activation. In addition, GH rapidly induces spatio-temporal re-localization of C/EBPbeta within the nucleus. Thus, GH-regulated gene transcription mediated by C/EBPbeta reflects the integration of diverse mechanisms including post-translational modifications, modulation of protein complexes associated with DNA and re-localization of gene regulatory proteins. Similar integration involving other transcription factors, including Stats, appears to be a feature of regulation by GH of other gene targets.
Collapse
Affiliation(s)
- Teresa I. Ceseña
- Cellular & Molecular Biology Program, University Michigan, Ann Arbor, MI 48109
| | - Tracy Xiao Cui
- Dept. of Molecular & Integrative Physiology, University Michigan, Ann Arbor, MI 48109
| | | | - Julianne Kaplani
- Dept. of Molecular & Integrative Physiology, University Michigan, Ann Arbor, MI 48109
| | | | | | - Roland Kwok
- Depts of Biological Chemistry and Ob/Gyn, University Michigan, Ann Arbor, MI 48109
| | - Jessica Schwartz
- Cellular & Molecular Biology Program, University Michigan, Ann Arbor, MI 48109
- Dept. of Molecular & Integrative Physiology, University Michigan, Ann Arbor, MI 48109
| |
Collapse
|
24
|
Vickers MH, Hofman PL, Gluckman PD, Lobie PE, Cutfield WS. Combination therapy with acipimox enhances the effect of growth hormone treatment on linear body growth in the normal and small-for-gestational-age rat. Am J Physiol Endocrinol Metab 2006; 291:E1212-9. [PMID: 16803850 DOI: 10.1152/ajpendo.00614.2005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Growth hormone (GH) therapy is often associated with adverse side effects, including impaired insulin sensitivity. GH treatment of children with idiopathic short stature does not lead to an optimized final adult height. It has been demonstrated that FFA reduction induced by pharmacological antilipolysis can stimulate GH secretion per se in both normal subjects and those with GH deficiency. However, to date, no investigation has been undertaken to establish efficacy of combination treatment with GH and FFA regulators on linear body growth. Using a model of maternal undernutrition in the rat to induce growth-restricted offspring, we investigated the hypothesis that combination treatment with GH and FFA regulators can enhance linear body growth above that of GH alone. At postnatal day 28, male offspring of normally nourished mothers (controls) and offspring born with low birth weight [small for gestational age (SGA)] were treated with saline, GH, or GH (5 mg.kg(-1).day(-1)) in combination with acipimox (GH + acipimox, 20 mg.kg(-1).day(-1)) or fenofibrate (GH + fenofibrate, 30 mg.kg(-1).day(-1)) for 40 days. GH plus acipimox treatment significantly enhanced linear body growth in the control and SGA animals above that of GH, as quantified by tibial and total body length. Treatment with GH significantly increased fasting plasma insulin, insulin-to-glucose ratio, and plasma volumes in control and SGA animals but was not significantly different between saline and GH-plus-acipimox-treated animals. GH-induced lipolysis was blocked by GH plus acipimox treatment in both control and SGA animals, concomitant with a significant reduction in fasting plasma FFA and insulin concentrations. This is the first study to show that GH plus acipimox combination therapy, via pharmacological blocking of lipolysis during GH exposure, can significantly enhance the efficacy of GH in linear growth promotion and ameliorate unwanted metabolic side effects.
Collapse
Affiliation(s)
- M H Vickers
- Liggins Institute, University of Auckland, Auckland, New Zealand.
| | | | | | | | | |
Collapse
|
25
|
Jayne S, Zwartjes C, Van Schaik F, Timmers H. Involvement of the SMRT/NCoR-HDAC3 complex in transcriptional repression by the CNOT2 subunit of the human Ccr4-Not complex. Biochem J 2006; 398:461-7. [PMID: 16712523 PMCID: PMC1559471 DOI: 10.1042/bj20060406] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In eukaryotic cells, the Ccr4-Not complex can regulate mRNA metabolism at various levels. Previously, we showed that promoter targeting of the CNOT2 subunit resulted in strong repression of RNA polymerase II transcription, which was sensitive to the HDAC (histone deacetylase) inhibitor, trichostatin A [Zwartjes, Jayne, van den Berg and Timmers (2004) J. Biol. Chem. 279, 10848-10854]. In the present study, the cofactor requirement for CNOT2-mediated repression was investigated. We found that coexpression of SMRT (silencing mediator for retinoic acid receptor and thyroid-hormone receptor) or NCoR (nuclear hormone receptor co-repressor) in combination with HDAC3 (or HDAC5 and HDAC6) augmented the repression by CNOT2. This repressive effect is mediated by the conserved Not-Box, which resides at the C-terminus of CNOT2 proteins. We observed physical interactions of CNOT2 with several subunits of the SMRT/NCoR-HDAC3 complex. Our results show that the SMRT/NCoR-HDAC3 complex is a cofactor of CNOT2-mediated repression and suggest that transcriptional regulation by the Ccr4-Not complex involves regulation of chromatin modification.
Collapse
Affiliation(s)
- Sandrine Jayne
- Department of Physiological Chemistry, Stratenum STR 3.211, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | - Carin G. M. Zwartjes
- Department of Physiological Chemistry, Stratenum STR 3.211, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | - Frederik M. A. Van Schaik
- Department of Physiological Chemistry, Stratenum STR 3.211, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | - H. Th. Marc Timmers
- Department of Physiological Chemistry, Stratenum STR 3.211, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
- To whom correspondence should be addressed (email )
| |
Collapse
|
26
|
Alliston T, Ko TC, Cao Y, Liang YY, Feng XH, Chang C, Derynck R. Repression of Bone Morphogenetic Protein and Activin-inducible Transcription by Evi-1. J Biol Chem 2005; 280:24227-37. [PMID: 15849193 DOI: 10.1074/jbc.m414305200] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Smads, key effectors of transforming growth factor (TGF)-beta, activin, and bone morphogenetic protein (BMP) signaling, regulate gene expression and interact with coactivators and corepressors that modulate Smad activity. The corepressor Evi-1 exerts its oncogenic effects by repressing TGF-beta/Smad3-mediated transcription, thereby blocking TGF-beta-induced growth arrest. Because Evi-1 interacts with the highly conserved MH2 domain of Smad3, we investigated the physical and functional interaction of Evi-1 with Smad1 and Smad2, downstream targets of BMP and activin signaling, respectively. Evi-1 interacted with and repressed the receptor-activated transcription through Smad1 and Smad2, similarly to Smad3. In addition, Evi-1 repressed BMP/Smad1- and activin/Smad2-mediated induction of endogenous Xenopus gene expression, suggesting a role of repression of BMP and activin signals by Evi-1 in vertebrate embryogenesis. Evi-1 also repressed the induction of endogenous Smad7 expression by TGF-beta family ligands. In the course of these studies, we observed Evi-1 repression of Smad transactivation even when Smad binding to DNA was kept constant. We therefore explored the mechanism of Evi-1 repression of TGF-beta family-inducible transcription. Evi-1 repression did not result from displacement of Smad binding to DNA or to CREB-binding protein but from the recruitment of Evi-1 by Smad3 and CREB-binding protein to DNA. Following TGF-beta stimulation, Evi-1 and the associated corepressor CtBP were recruited to the endogenous Smad7 promoter. Evi-1 recruitment to the promoter decreased TGF-beta-induced histone acetylation, coincident with its repression of Smad7 gene expression. In this way, Evi-1 acts as a general Smad corepressor to inhibit TGF-beta-, activin-, and BMP-inducible transcription.
Collapse
Affiliation(s)
- Tamara Alliston
- Department of Cell and Tissue Biology, University of California at San Francisco, San Francisco, California 94143-0512, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Growth hormone (GH) has numerous effects in the body and is most commonly known for its role in regulating metabolism and body growth. Because GH is involved in many aspects of cell function, its signaling is tightly controlled by several pathways at both the extracellular and intracellular level. Suppressor of cytokine signaling-2 (SOCS2) is one such intracellular regulator of GH signal transduction. Expression of SOCS2 is tightly regulated and alteration of its levels leads to marked abnormalities in metabolism and growth. Unexpectedly, GH and SOCS2 have been recently shown to regulate neural development, neural stem cell differentiation and neuronal growth -- functions that might have important therapeutic implications for both repairing nervous system injuries and treating neurological disease.
Collapse
Affiliation(s)
- Ann M Turnley
- Centre for Neuroscience, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|