1
|
Qi D, Liu P, Wang Y, Tai X, Ma S, Wang Y. A multi-omic dissection of molecular hallmarks of accelerated aging in schizophrenia. Sci Rep 2025; 15:17856. [PMID: 40404850 PMCID: PMC12098977 DOI: 10.1038/s41598-025-98181-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 04/09/2025] [Indexed: 05/24/2025] Open
Abstract
Schizophrenia, a pervasive mental health condition, poses a global challenge to individual well-being. The intricate interplay of schizophrenia with the aging process is characterized by a shortened lifespan, underscoring the urgent need for an in-depth exploration of the underlying biological mechanisms that contribute to the unique aging trajectory within this specific patient population. Currently, this crucial aspect remains largely unexplored. To bridge this knowledge gap, in the present study, serum samples from of 29 subjects with schizophrenia were analysed via liquid chromatography-mass spectrometry (LC-MS) and compared with those of 30 nonpsychiatric controls. This exploratory analysis of circulating blood serum, albeit based on a limited sample size, provides valuable insights into the significantly altered molecular pathways linked to schizophrenia and clarifies the unique aspects of aging in the context of this particular disease. The data presented in this study constitute a compilation of molecular alterations associated with schizophrenia across the human lifespan. By distinguishing between schizophrenia patients and nonpsychiatric controls, we identified a panel of 342 differentially abundant metabolites and 544 differentially expressed proteins. Our pathway enrichment analysis was focused primarily on histone acetylation, the Wnt/β-catenin signaling pathway, the dopamine receptor signaling pathway, and fatty acid beta-oxidation, highlighting their pivotal roles in schizophrenia. Furthermore, we conducted a co-occurrence analysis of these metabolites and proteins, aiming to elucidate their connection with accelerated aging processes. This analysis revealed the involvement of metabolic pathways crucial for lipid metabolism, such as the peroxisome and peroxisome proliferator-activated receptor (PPAR) signaling pathways, as well as neurotransmitter-related metabolic pathways influencing tryptophan metabolism and the dopaminergic synapse pathway. The findings significantly increase our understanding of the intricate molecular mechanisms underlying schizophrenia and its co-occurring comorbidities across the spectrum of aging, providing insights into the dynamics of the progression of this complex disorder. However, this study has certain limitations, particularly the lack of its findings and the relatively small sample size, especially in the proteomic analysis.
Collapse
Affiliation(s)
- DongDong Qi
- Basic and Clinical laboratory of Mental illness, The Third People's Hospital of Hulunbuir City (The Mental Health Center of Hulunbuir City), Yakeshi, InnerMongolia, China
| | - Peng Liu
- Basic and Clinical laboratory of Mental illness, The Third People's Hospital of Hulunbuir City (The Mental Health Center of Hulunbuir City), Yakeshi, InnerMongolia, China
| | - YiMeng Wang
- Basic and Clinical laboratory of Mental illness, The Third People's Hospital of Hulunbuir City (The Mental Health Center of Hulunbuir City), Yakeshi, InnerMongolia, China
| | - XuGuang Tai
- Basic and Clinical laboratory of Mental illness, The Third People's Hospital of Hulunbuir City (The Mental Health Center of Hulunbuir City), Yakeshi, InnerMongolia, China
| | - ShiFa Ma
- Basic and Clinical laboratory of Mental illness, The Third People's Hospital of Hulunbuir City (The Mental Health Center of Hulunbuir City), Yakeshi, InnerMongolia, China
| | - Yue Wang
- Basic and Clinical laboratory of Mental illness, The Third People's Hospital of Hulunbuir City (The Mental Health Center of Hulunbuir City), Yakeshi, InnerMongolia, China.
| |
Collapse
|
2
|
Gao Q, Shi X, Liu Y, Han Y, Yuan Z, Zhang D, Zhang H, Weng Q. 1,25(OH)₂D₃ regulates androgen synthesis via transcriptional control of steroidogenic enzymes and LHR in the scented glands of muskrats (Ondatra zibethicus). Free Radic Biol Med 2025; 229:82-95. [PMID: 39827922 DOI: 10.1016/j.freeradbiomed.2025.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/19/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Photoperiodic changes induce seasonal variations in vitamin D3 levels, which can affect reproductive function. The muskrat, a seasonal breeder, possesses a pair of scented glands that secrete musky substances to attract mates. The scented glands can also synthesize androgens, which regulate their function through autocrine or paracrine signaling. This study aimed to investigate whether active vitamin D3 was synthesized locally and to explore how seasonal changes affected the specific mechanisms of androgen synthesis in the scented glands. The scented glands showed significant seasonal changes in morphology and histology. Transcriptomic analysis revealed that differentially expressed genes were enriched in pathways related to sex steroid hormone synthesis and calcium signaling. Quantitative analyses using qPCR and Western blotting demonstrated significant seasonal variations in the expressions of vitamin D3 receptors (VDR) and key synthetic and metabolic enzymes. Seasonal fluctuations in 1,25(OH)₂D₃ levels were positively correlated with the expressions of steroidogenic enzymes and androgen concentrations in the scented glands. Additionally, 1,25(OH)₂D₃ enhanced the expressions of steroidogenic enzymes in cultured primary cells. ChIP-seq analysis revealed that VD/VDR directly regulated the transcription of Cyp11a1, Cyp17a1, and Lhr by binding to their promoter regions. Furthermore, elevated androgen levels were observed when hCG was combined with 1 nM 1,25(OH)₂D₃. This study suggests that the scented glands can produce 1,25(OH)₂D₃ in autocrine or paracrine forms, with levels varying seasonally. VD/VDR enhanced androgen synthesis through a dual mechanism: (1) directly up-regulating the transcription of Cyp11a1 and Cyp17a1 to increase steroidogenic enzyme levels, and (2) indirectly promoting the expressions of steroidogenic enzymes by modulating Lhr transcription.
Collapse
Affiliation(s)
- Qingjing Gao
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Xinjing Shi
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Yuning Liu
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Yingying Han
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Zhengrong Yuan
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Dong Zhang
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, China
| | - Haolin Zhang
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.
| | - Qiang Weng
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.
| |
Collapse
|
3
|
Aminizadeh S, Moslemizadeh AH, Sheibani S, Sedighi-Khovidak Z, Roholamini Z, Jafarinejad-Farsangi S, Kheirandish R, Sheibani V, Bashiri H. Preventive effect of MitoQ supplementation and endurance training on glioblastoma and its consequences: TLR4/CREB/ NF-κβ /IL-1β pathway and behaviors. Int Immunopharmacol 2025; 145:113756. [PMID: 39662270 DOI: 10.1016/j.intimp.2024.113756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 11/17/2024] [Accepted: 11/27/2024] [Indexed: 12/13/2024]
Abstract
OBJECTIVE The present study investigated the preventive effect of MitoQ supplementation and endurance training (ET) on the TLR4/CREB/ NF-κβ signaling pathway, antioxidant indices, and behaviors in C6-induced glioblastoma (GBM) in rats. METHODS 60 male Wistar rats were randomly divided into five groups (n = 12); Sham, Tumor, MitoQ, ET, and MitoQ + ET. Rats in the training groups performed endurance training (5 days per week), and MitoQ at the dose of 250 µM/L daily was administered in drinking water for 8 weeks. At the end of the protocol, all groups except the sham group received 1*106 tumor cells /10 µl culture medium. Two weeks after tumor induction, behavioral tests were performed, and then brain tissue was collected for the histopathology, measurement of antioxidant and inflammatory factors, TLR4, NF-κB proteins, and TLR4, NF-κβ, CREB, IL-1ß, TNF-a, IL-10, Bax, Bcl-2, and Caspase-3 gene expression. RESULTS The increased level of TLR4 and NF-κβ protein expression in GBM rats decreased in the treatment groups. Gene expression of TLR4, NF-κβ, CREB, TNF-a, IL-10, and Bcl-2 increased in the tumor groups, and treatment groups decreased TLR4, NF-κB, Bcl-2, and CREB. In addition, social behaviors, balance, and memory were impaired in the tumor group, which combination group could improve these behaviors. CONCLUSION In sum, the preventive effects of MitoQ as a beneficial immune reactive agent and exercise training in rats with C6-induced glioblastoma may be mediated via modulating oxidative stress, inflammatory factors, and down-regulation of the expression of TLR4.
Collapse
Affiliation(s)
- Soheil Aminizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Sara Sheibani
- Department of Behavioral and Molecular Neurobiology, Regensburg Center for Neuroscience, University of Regensburg, Regensburg, Germany
| | - Zahra Sedighi-Khovidak
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Zahrasadat Roholamini
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | | | - Reza Kheirandish
- Department of Pathology, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
4
|
Souza CB, Menezes GRO, Gondo A, Egito AA, Ramos PVB, Gomes RC, Ribas MN, Fernandes Júnior JA, Guimarães SEF. Estimation of Genetic Parameters and GWAS on Water Efficiency Traits in the Senepol Cattle. J Anim Breed Genet 2024. [PMID: 39726399 DOI: 10.1111/jbg.12920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/20/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024]
Abstract
The need for producing in environmentally resilient system drives new research to achieve sustainable beef production. Water footprint of the beef supply chain is a concern that must be addressed, aiming to improve water use within the production chain. One approach is genetic selection of beef cattle for water efficiency. However, it is essential to understand the genetic architecture and mechanisms involved in the expression of this phenotype to choose the best selection criteria. Thus, our study aimed to estimate genetic parameters for water efficiency traits, conduct a genome-wide association study (GWAS) and identify the genetic networks and biological processes involved. A population of 1762 purebred Senepol cattle was phenotyped for the following water efficiency traits: water intake (WI), gross water efficiency (GWE), water conversion ratio (WCR), residual water intake based on average daily gain (RWIADG) and residual water intake based on dry matter intake (RWIDMI). A subset of 1342 animals was genotyped using GGP Bovine 50 K SNP Chip with (734 animals) or 100 K (508 animals), and imputation from 50 K to 100 K was performed with Beagle software. The heritability estimates were 0.36 ± 0.06, 0.26 ± 0.05, 0.22 ± 0.05, 0.24 ± 0.05 and 0.20 ± 0.05 for WI, GWE, WCR, RWIADG and RWIDMI, respectively. Unlike the raw measures of WI, the phenotypic correlations between average daily gain (ADG) and the residuals (RWIDMI and RWIADG) were zero. All water efficiency traits were moderately to highly correlated with each other. GWAS were used to estimate the effect of 79,860 single nucleotide polymorphisms (SNPs), and significant SNPs were only observed for WCR. Enrichment analysis of genes in the significant regions revealed the involvement of different biological processes, such as saliva production, water transport, renal system and immune system. Genetic selection of Senepol cattle for water efficiency traits is feasible and can reduce water requirements for meat production. Water efficiency measures are polygenic traits, and different biological processes act simultaneously on the expression of related phenotypes.
Collapse
Affiliation(s)
- Christhian B Souza
- Departament of Animal Science, Federal University of Viçosa, Viçosa, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Pérez-de-Oliveira ME, Wagner VP, Bingle CD, Vargas PA, Bingle L. Disruption of oncogenic pathways in mucoepidermoid carcinoma: CREB inhibitor 666.15 as a potential therapeutic agent. Oral Oncol 2024; 159:107029. [PMID: 39332274 DOI: 10.1016/j.oraloncology.2024.107029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/07/2024] [Indexed: 09/29/2024]
Abstract
OBJECTIVES Mucoepidermoid carcinoma (MEC) is the most common malignant salivary gland tumour with around 50 % of cases carrying the CRTC1-MAML2 translocation. The CREB pathway has been associated with the transforming activity of this translocation. The aim of this study was to determine the effects of CREB inhibition on MEC cell behaviour in vitro. MATERIAL AND METHODS Two translocation-positive (UM-HMC-2 and H292) and one translocation-negative (H253) MEC cell lines were treated with 666.15, a CREB inhibitor. Drug IC50 doses were determined for each cell line. Clonogenic and spheroid assays were used to assess survival, including percentage of cancer stem cells, and transwell and scratch assays evaluated invasive and migratory capacities, respectively. Immunofluorescence staining was used to determine E-cadherin expression. RESULTS CREB inhibition significantly reduced the number of surviving colonies and spheroids and delayed cell invasion in all cell lines, but this was more significant in the fusion positive, UM-HMC-2 cells. The expression of E-cadherin was significantly higher in treated UM-HMC-2 and H292 cells. CONCLUSION CREB inhibition with 666.15 impaired key MEC oncogenic behaviours associated with metastasis and drug resistance, including cell invasion and survival.
Collapse
Affiliation(s)
- Maria Eduarda Pérez-de-Oliveira
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, São Paulo, Brazil; School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Vivian Petersen Wagner
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, São Paulo, Brazil; School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom; Department of Oral Medicine, School of Dentistry, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Colin D Bingle
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom
| | - Pablo Agustin Vargas
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, São Paulo, Brazil
| | - Lynne Bingle
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
6
|
Herring JA, Crabtree JE, Hill JT, Tessem JS. Loss of glucose-stimulated β-cell Nr4a1 expression impairs insulin secretion and glucose homeostasis. Am J Physiol Cell Physiol 2024; 327:C1111-C1124. [PMID: 39219449 PMCID: PMC11482045 DOI: 10.1152/ajpcell.00315.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/15/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
A central aspect of type 2 diabetes is decreased functional β-cell mass. The orphan nuclear receptor Nr4a1 is critical for fuel utilization, but little is known regarding its regulation and function in the β-cell. Nr4a1 expression is decreased in type 2 diabetes rodent β-cells and type 2 diabetes patient islets. We have shown that Nr4a1-deficient mice have reduced β-cell mass and that Nr4a1 knockdown impairs glucose-stimulated insulin secretion (GSIS) in INS-1 832/13 β-cells. Here, we demonstrate that glucose concentration directly regulates β-cell Nr4a1 expression. We show that 11 mM glucose increases Nr4a1 expression in INS-1 832/13 β-cells and primary mouse islets. We show that glucose functions through the cAMP/PKA/CREB pathway to regulate Nr4a1 mRNA and protein expression. Using Nr4a1-/- animals, we show that Nr4a1 is necessary for GSIS and systemic glucose handling. Using RNA-seq, we define Nr4a1-regulated pathways in response to glucose in the mouse islet, including Glut2 expression. Our data suggest that Nr4a1 plays a critical role in the β-cells response to the fed state.NEW & NOTEWORTHY Nr4a1 has a key role in fuel metabolism and β-cell function, but its exact role is unclear. Nr4a1 expression is regulated by glucose concentration using cAMP/PKA/CREB pathway. Nr4a1 regulates Glut2, Ndufa4, Ins1, In2, Sdhb, and Idh3g expression in response to glucose treatment. These results suggest that Nr4a1 is necessary for proper insulin secretion both through glucose uptake and metabolism machinery.
Collapse
Affiliation(s)
- Jacob A Herring
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah, United States
| | - Jacqueline E Crabtree
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, Provo, Utah, United States
| | - Jonathon T Hill
- Department of Cell Biology and Physiology, Brigham Young University, Provo, Utah, United States
| | - Jeffery S Tessem
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah, United States
| |
Collapse
|
7
|
Cao ZM, Fu S, Dong C, Yang TY, Liu XK, Zhang CL, Li DZ. DSCR1-1 attenuates osteoarthritis-associated chondrocyte injury by regulating the CREB1/ALDH2/Wnt/β-catenin axis: An in vitro and in vivo study. Cell Signal 2024; 121:111287. [PMID: 38969191 DOI: 10.1016/j.cellsig.2024.111287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/27/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
The progression of osteoarthritis (OA) includes the initial inflammation, subsequent degradation of the extracellular matrix (ECM), and chondrocyte apoptosis. Down syndrome candidate region 1 (DSCR1) is a stress-responsive gene and expresses in varied types of cells, including chondrocytes. Bioinformatics analysis of GSE103416 and GSE104739 datasets showed higher DSCR1 expression in the inflamed cartilage tissues and chondrocytes of OA. DSCR1 had two major isoforms, isoform 1 (DSCR1-1) and isoform 4 (DSCR1-4). We found that DSCR1-1 had a faster (in vitro) and higher expression (in vivo) response to OA compared to DSCR1-4. IL-1β-induced apoptosis, inflammation, and ECM degradation in chondrocytes were attenuated by DSCR1-1 overexpression. DSCR1-1 triggered the phosphorylation of cAMP response element-binding 1 (CREB1) at 133 serine sites by decreasing calcineurin activity. Moreover, activated CREB1 moved into the cell nucleus and combined in the promoter regions of aldehyde dehydrogenase 2 (ALDH2), thus enhancing its gene transcription. ALDH2 could recover Wnt/β-catenin signaling transduction by enhancing phosphorylation of β-catenin at 33/37 serine sites and inhibiting the migration of β-catenin protein from the cellular matrix to the nucleus. In vivo, adenoviruses (1 × 108 PFU) overexpressing DSCR1-1 were injected into the articular cavity of C57BL/6 mice with medial meniscus surgery-induced OA, and it showed that DSCR1-1 overexpression ameliorated cartilage injury. Collectively, our study demonstrates that DSCR1-1 may be a potential therapeutic target of OA.
Collapse
Affiliation(s)
- Zheng-Ming Cao
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, China.
| | - Su Fu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, China
| | - Chao Dong
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, China
| | - Teng-Yue Yang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, China
| | - Xiao-Kang Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, China
| | - Chun-Lin Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, China
| | - Dong-Zhe Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, China.
| |
Collapse
|
8
|
Lambourne L, Mattioli K, Santoso C, Sheynkman G, Inukai S, Kaundal B, Berenson A, Spirohn-Fitzgerald K, Bhattacharjee A, Rothman E, Shrestha S, Laval F, Yang Z, Bisht D, Sewell JA, Li G, Prasad A, Phanor S, Lane R, Campbell DM, Hunt T, Balcha D, Gebbia M, Twizere JC, Hao T, Frankish A, Riback JA, Salomonis N, Calderwood MA, Hill DE, Sahni N, Vidal M, Bulyk ML, Fuxman Bass JI. Widespread variation in molecular interactions and regulatory properties among transcription factor isoforms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584681. [PMID: 38617209 PMCID: PMC11014633 DOI: 10.1101/2024.03.12.584681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Most human Transcription factors (TFs) genes encode multiple protein isoforms differing in DNA binding domains, effector domains, or other protein regions. The global extent to which this results in functional differences between isoforms remains unknown. Here, we systematically compared 693 isoforms of 246 TF genes, assessing DNA binding, protein binding, transcriptional activation, subcellular localization, and condensate formation. Relative to reference isoforms, two-thirds of alternative TF isoforms exhibit differences in one or more molecular activities, which often could not be predicted from sequence. We observed two primary categories of alternative TF isoforms: "rewirers" and "negative regulators", both of which were associated with differentiation and cancer. Our results support a model wherein the relative expression levels of, and interactions involving, TF isoforms add an understudied layer of complexity to gene regulatory networks, demonstrating the importance of isoform-aware characterization of TF functions and providing a rich resource for further studies.
Collapse
Affiliation(s)
- Luke Lambourne
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kaia Mattioli
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Clarissa Santoso
- Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - Gloria Sheynkman
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sachi Inukai
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Babita Kaundal
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anna Berenson
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| | - Kerstin Spirohn-Fitzgerald
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Anukana Bhattacharjee
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Elisabeth Rothman
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Florent Laval
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- TERRA Teaching and Research Centre, University of Liège, Gembloux, Belgium
- Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège, Belgium
| | - Zhipeng Yang
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Deepa Bisht
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jared A Sewell
- Department of Biology, Boston University, Boston, MA, USA
| | - Guangyuan Li
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anisa Prasad
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Harvard College, Cambridge MA, USA
| | - Sabrina Phanor
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ryan Lane
- Department of Biology, Boston University, Boston, MA, USA
| | | | - Toby Hunt
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Dawit Balcha
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Marinella Gebbia
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario, Canada
| | - Jean-Claude Twizere
- TERRA Teaching and Research Centre, University of Liège, Gembloux, Belgium
- Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège, Belgium
| | - Tong Hao
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Adam Frankish
- Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège, Belgium
| | - Josh A Riback
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nathan Salomonis
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael A Calderwood
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David E Hill
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nidhi Sahni
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marc Vidal
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Martha L Bulyk
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Juan I Fuxman Bass
- Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| |
Collapse
|
9
|
Falcón D, Calderón-Sánchez EM, Mayoral-González I, Martín-Bórnez M, Dominguez-Rodriguez A, Gutiérrez-Carretero E, Ordóñez-Fernández A, Rosado JA, Smani T. Inhibition of adenylyl cyclase 8 prevents the upregulation of Orai1 channel, which improves cardiac function after myocardial infarction. Mol Ther 2024; 32:646-662. [PMID: 38291755 PMCID: PMC10928147 DOI: 10.1016/j.ymthe.2024.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/14/2023] [Accepted: 01/19/2024] [Indexed: 02/01/2024] Open
Abstract
The upregulation of Orai1 and subsequent store-operated Ca2+ entry (SOCE) has been associated with adverse cardiac remodeling and heart failure (HF). However, the mechanism underlying Orai1 upregulation and its role in myocardial infarction remains unclear. Our study investigated the role of Orai1 in activating adenylyl cyclase 8 (AC8) and cyclic AMP (cAMP) response element-binding protein (CREB), as well as its contribution to cardiac dysfunction induced by ischemia and reperfusion (I/R). We found that I/R evoked an increase in the expression of Orai1 and AC8 in rats' hearts, resulting in a substantial rise in diastolic Ca2+ concentration ([Ca2+]i), and reduced ventricular contractions. The expression of Orai1 and AC8 was also increased in ventricular biopsies of post-ischemic HF patients. Mechanistically, we demonstrate that I/R activation of Orai1 stimulated AC8, which produced cAMP and phosphorylated CREB. Subsequently, p-CREB activated the ORAI1 promoter, resulting in Orai1 upregulation and SOCE exacerbation. Intramyocardial administration of AAV9 carrying AC8 short hairpin RNA decreased the expression of AC8, Orai1 and CREB, which restored diastolic [Ca2+]i and improved cardiac contraction. Therefore, our data suggests that the axis composed by Orai1/AC8/CREB plays a critical role in I/R-induced cardiac dysfunction, representing a potential new therapeutic target to limit the progression of the disease toward HF.
Collapse
Affiliation(s)
- Débora Falcón
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocio/University of Seville/CSIC, 41013 Seville, Spain; Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain.
| | - Eva M Calderón-Sánchez
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocio/University of Seville/CSIC, 41013 Seville, Spain; Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| | - Isabel Mayoral-González
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocio/University of Seville/CSIC, 41013 Seville, Spain
| | - Marta Martín-Bórnez
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocio/University of Seville/CSIC, 41013 Seville, Spain; Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| | - Alejandro Dominguez-Rodriguez
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocio/University of Seville/CSIC, 41013 Seville, Spain
| | - Encarnación Gutiérrez-Carretero
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocio/University of Seville/CSIC, 41013 Seville, Spain; Department of Surgery, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| | - Antonio Ordóñez-Fernández
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocio/University of Seville/CSIC, 41013 Seville, Spain
| | - Juan Antonio Rosado
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - Tarik Smani
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocio/University of Seville/CSIC, 41013 Seville, Spain; Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain.
| |
Collapse
|
10
|
Liang J, Bi G, Huang Y, Zhao G, Sui Q, Zhang H, Bian Y, Yin J, Wang Q, Chen Z, Zhan C. MAFF confers vulnerability to cisplatin-based and ionizing radiation treatments by modulating ferroptosis and cell cycle progression in lung adenocarcinoma. Drug Resist Updat 2024; 73:101057. [PMID: 38266355 DOI: 10.1016/j.drup.2024.101057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 12/22/2023] [Accepted: 01/16/2024] [Indexed: 01/26/2024]
Abstract
AIMS Lung cancer is the leading cause of cancer mortality and lung adenocarcinoma (LUAD) accounts for more than half of all lung cancer cases. Tumor elimination is mostly hindered by drug resistance and the mechanisms remain to be explored in LUAD. METHODS CRISPR screens in cell and murine models and single-cell RNA sequencing were conducted, which identified MAF bZIP transcription factor F (MAFF) as a critical factor regulating tumor growth and treatment resistance in LUAD. RNA and ChIP sequencing analyses were performed for transcriptional target expression and specific binding sites of MAFF. Functions of MAFF in inhibiting tumor growth and promoting cisplatin or irradiation efficacy were investigated using cellular and xenograft models. RESULTS Patients with lung adenocarcinoma and reduced MAFF expression had worse clinical outcomes. MAFF inhibited tumor cell proliferation by regulating the expression of SLC7A11, CDK6, and CDKN2C, promoting ferroptosis and preventing cell cycle progression from G1 to S. MAFF also conferred tumor cells vulnerable to cisplatin-based or ionizing radiation treatments. MAFF reduction was a final event in the acquisition of cisplatin resistance of LUAD cells. The intracellular cAMP/PKA/CREB1 pathway upregulated MAFF in response to cisplatin-based or ionizing radiation treatments. CONCLUSIONS MAFF suppresses tumor growth, and pharmacological agonists targeting MAFF may improve cisplatin or irradiation therapies for lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Guangyin Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Qihai Sui
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Huan Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Jiacheng Yin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China.
| | - Zhencong Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China.
| |
Collapse
|
11
|
Cai Y, Dong J, Huang J, He J, Hu Y, Sui Z, Tang P. The cyclic AMP (cAMP) phosphodiesterase CpdA required for growth, biofilm formation, motility and pathogenicity of Edwardsiella piscicida. Microb Pathog 2024; 188:106545. [PMID: 38244636 DOI: 10.1016/j.micpath.2024.106545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/09/2024] [Accepted: 01/14/2024] [Indexed: 01/22/2024]
Abstract
Edwardsiella piscicida is a severe fish pathogen with wide host range, causing the huge economic losses in the aquaculture industry. Cyclic adenosine monophosphate (cAMP) as an important second messenger regulates the physiological and behavioral responses to environmental cues in eukaryotic and prokaryotic. The intracellular level of cAMP for effective activity is tightly controlled by the synthesis of adenylate cyclase, excretion and degradation of phosphodiesterase. In this study, we identified and characterized a class III cAMP phosphodiesterase, named as CpdA, in the E. piscicida. To investigate the role of CpdA in the physiology and pathogenicity, we constructed the in-frame deletion mutant of cpdA of E. piscicida, TX01ΔcpdA. The results showed that TX01ΔcpdA accumulated the higher intracellular cAMP concentration than TX01, indicating that CpdA exerted the hydrolysis of cAMP. In addition, compared to the TX01, the TX01ΔcpdA slowed growth rate, diminished biofilm formation and lost motility. More importantly, pathogenicity analysis confirmed that TX01ΔcpdA significantly impaired the ability of invading the epithelial cells, reproduction in macrophages, tissues dissemination and lethality for healthy tilapias. The most of lost properties of TX01ΔcpdA were restored partially or fully by the introduction of cpdA gene. These results suggest that cpdA is required for regulation of the physiology and virulence of E. piscicida.
Collapse
Affiliation(s)
- Yidong Cai
- School of Life and Health, Hainan University, Haikou, 570228, China; Key Laboratory of Biology and Genetic Resources of Tropical Crops, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; Key Laboratory of Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute for Tropical Agricultural Resources, Haikou, 571101, China
| | - Jinggang Dong
- Key Laboratory of Biology and Genetic Resources of Tropical Crops, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; Key Laboratory of Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute for Tropical Agricultural Resources, Haikou, 571101, China
| | - Jianqiang Huang
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, 524013, China
| | - Jiaojiao He
- School of Life and Health, Hainan University, Haikou, 570228, China; Key Laboratory of Biology and Genetic Resources of Tropical Crops, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; Key Laboratory of Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute for Tropical Agricultural Resources, Haikou, 571101, China
| | - Yonghua Hu
- Key Laboratory of Biology and Genetic Resources of Tropical Crops, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; Key Laboratory of Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute for Tropical Agricultural Resources, Haikou, 571101, China; Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Haikou, 571101, China; Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, 524013, China
| | - Zhihai Sui
- School of Life Science, Linyi University, Linyi, 276000, China.
| | - Ping Tang
- State Key Laboratory of Conservation and Utilization of Biologícal Resources of Yunnan, College of Plant Protection, Yunnan Agricultural University, Kunming, 650201, China.
| |
Collapse
|
12
|
Jin Z, Cao Y, Wen Q, Zhang H, Fang Z, Zhao Q, Xi Y, Luo Z, Jiang H, Zhang Z, Hang J. Dapagliflozin ameliorates diabetes-induced spermatogenic dysfunction by modulating the adenosine metabolism along the gut microbiota-testis axis. Sci Rep 2024; 14:641. [PMID: 38182877 PMCID: PMC10770392 DOI: 10.1038/s41598-024-51224-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024] Open
Abstract
Male infertility is one of the most common complications of diabetes mellitus (DM). Dapagliflozin is widely used to manage the type II DM. This study aimed to assess the dapagliflozin's effects on the spermatogenesis by administering either dapagliflozin (Dapa) or vehicle (db) to male db/db mice, and using littermate male db/m mice as the control (Con). We further performed the integrative analyses of the cecal shotgun metagenomics, cecal/plasmatic/testicular metabolomics, and testicular proteomics. We found that dapagliflozin treatment significantly alleviated the diabetes-induced spermatogenic dysfunction by improving sperm quality, including the sperm concentration and sperm motility. The overall microbial composition was reshaped in Dapa mice and 13 species (such as Lachnospiraceae bacterium 3-1) were regarded as potential beneficial bacteria. Metabolites exhibited modified profiles, in which adenosine, cAMP, and 2'-deoxyinosine being notably altered in the cecum, plasma, and testis, respectively. Testicular protein expression patterns were similar between the Dapa and Con mice. In vivo results indicated that when compared with db group, dapagliflozin treatment alleviated apoptosis and oxidative stress in testis tissues by down-regulating 2'-deoxyinosine. This was further validated by in vitro experiments using GC-2 cells. Our findings support the potential use of dapagliflozin to prevent the diabetes-induced impaired sperm quality and to treat diabetic male infertility.
Collapse
Affiliation(s)
- Zirun Jin
- Department of Urology, Center for Reproductive Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
- Department of Urology, Department of Andrology, Peking University First Hospital, No. 8, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yalei Cao
- Department of Urology, Center for Reproductive Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Qi Wen
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, No.49 North Garden Road, Haidian District, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
| | - Haitao Zhang
- Department of Urology, Center for Reproductive Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, No.49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Zhuofan Fang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, No.49 North Garden Road, Haidian District, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
| | - Qiancheng Zhao
- Department of Urology, Center for Reproductive Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Yu Xi
- Department of Urology, Center for Reproductive Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Zhichao Luo
- Department of Urology, Center for Reproductive Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Hui Jiang
- Department of Urology, Center for Reproductive Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
- Department of Urology, Department of Andrology, Peking University First Hospital, No. 8, Xishiku Street, Xicheng District, Beijing, 100034, China.
| | - Zhe Zhang
- Department of Urology, Center for Reproductive Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, No.49 North Garden Road, Haidian District, Beijing, 100191, China.
| | - Jing Hang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, No.49 North Garden Road, Haidian District, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China.
| |
Collapse
|
13
|
Sahay S, Henkel ND, Vargas CFA, McCullumsmith RE, O’Donovan SM. Activity of Protein Kinase A in the Frontal Cortex in Schizophrenia. Brain Sci 2023; 14:13. [PMID: 38248228 PMCID: PMC10813263 DOI: 10.3390/brainsci14010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/21/2023] [Accepted: 12/02/2023] [Indexed: 01/23/2024] Open
Abstract
Schizophrenia is a serious cognitive disorder characterized by disruptions in neurotransmission, a process requiring the coordination of multiple kinase-mediated signaling events. Evidence suggests that the observed deficits in schizophrenia may be due to imbalances in kinase activity that propagate through an intracellular signaling network. Specifically, 3'-5'-cyclic adenosine monophosphate (cAMP)-associated signaling pathways are coupled to the activation of neurotransmitter receptors and modulate cellular functions through the activation of protein kinase A (PKA), an enzyme whose function is altered in the frontal cortex in schizophrenia. In this study, we measured the activity of PKA in human postmortem anterior cingulate cortex (ACC) and dorsolateral prefrontal cortex (DLPFC) tissue from schizophrenia and age- and sex-matched control subjects. No significant differences in PKA activity were observed in male and female individuals in either brain region; however, correlation analyses indicated that PKA activity in the ACC may be influenced by tissue pH in all subjects and by age and tissue pH in females. Our data provide novel insights into the function of PKA in the ACC and DLPFC in schizophrenia.
Collapse
Affiliation(s)
- Smita Sahay
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (N.D.H.); (C.F.-A.V.); (R.E.M.)
| | - Nicholas Daniel Henkel
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (N.D.H.); (C.F.-A.V.); (R.E.M.)
| | - Christina Flora-Anabelle Vargas
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (N.D.H.); (C.F.-A.V.); (R.E.M.)
| | - Robert Erne McCullumsmith
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (N.D.H.); (C.F.-A.V.); (R.E.M.)
- Neuroscience Institute, Promedica, Toledo, OH 43606, USA
| | - Sinead Marie O’Donovan
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (N.D.H.); (C.F.-A.V.); (R.E.M.)
| |
Collapse
|
14
|
Chakraborty S, Anand S, Coe S, Reh B, Bhandari RK. The PCOS-NAFLD Multidisease Phenotype Occurred in Medaka Fish Four Generations after the Removal of Bisphenol A Exposure. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:12602-12619. [PMID: 37581432 PMCID: PMC10469501 DOI: 10.1021/acs.est.3c01922] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/16/2023]
Abstract
As a heterogeneous reproductive disorder, polycystic ovary syndrome (PCOS) can be caused by genetic, diet, and environmental factors. Bisphenol A (BPA) can induce PCOS and nonalcoholic fatty liver disease (NAFLD) due to direct exposure; however, whether these phenotypes persist in future unexposed generations is not currently understood. In a previous study, we observed that transgenerational NAFLD persisted in female medaka for five generations (F4) after exposure to an environmentally relevant concentration (10 μg/L) of BPA. Here, we demonstrate PCOS in the same F4 generation female medaka that developed NAFLD. The ovaries contained immature follicles, restricted follicular progression, and degenerated follicles, which are characteristics of PCOS. Untargeted metabolomic analysis revealed 17 biomarkers in the ovary of BPA lineage fish, whereas transcriptomic analysis revealed 292 genes abnormally expressed, which were similar to human patients with PCOS. Metabolomic-transcriptomic joint pathway analysis revealed activation of the cancerous pathway, arginine-proline metabolism, insulin signaling, AMPK, and HOTAIR regulatory pathways, as well as upstream regulators esr1 and tgf signaling in the ovary. The present results suggest that ancestral BPA exposure can lead to PCOS phenotypes in the subsequent unexposed generations and warrant further investigations into potential health risks in future generations caused by initial exposure to EDCs.
Collapse
Affiliation(s)
- Sourav Chakraborty
- Department of Biology, University of North Carolina at Greensboro, Greensboro 27412 North Carolina, United
States
| | - Santosh Anand
- Department of Biology, University of North Carolina at Greensboro, Greensboro 27412 North Carolina, United
States
| | - Seraiah Coe
- Department of Biology, University of North Carolina at Greensboro, Greensboro 27412 North Carolina, United
States
| | - Beh Reh
- Department of Biology, University of North Carolina at Greensboro, Greensboro 27412 North Carolina, United
States
| | - Ramji Kumar Bhandari
- Department of Biology, University of North Carolina at Greensboro, Greensboro 27412 North Carolina, United
States
| |
Collapse
|
15
|
Chowdhury MAR, An J, Jeong S. The Pleiotropic Face of CREB Family Transcription Factors. Mol Cells 2023; 46:399-413. [PMID: 37013623 PMCID: PMC10336275 DOI: 10.14348/molcells.2023.2193] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 04/05/2023] Open
Abstract
cAMP responsive element-binding protein (CREB) is one of the most intensively studied phosphorylation-dependent transcription factors that provide evolutionarily conserved mechanisms of differential gene expression in vertebrates and invertebrates. Many cellular protein kinases that function downstream of distinct cell surface receptors are responsible for the activation of CREB. Upon functional dimerization of the activated CREB to cis-acting cAMP responsive elements within the promoters of target genes, it facilitates signal-dependent gene expression. From the discovery of CREB, which is ubiquitously expressed, it has been proven to be involved in a variety of cellular processes that include cell proliferation, adaptation, survival, differentiation, and physiology, through the control of target gene expression. In this review, we highlight the essential roles of CREB proteins in the nervous system, the immune system, cancer development, hepatic physiology, and cardiovascular function and further discuss a wide range of CREB-associated diseases and molecular mechanisms underlying the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Md. Arifur Rahman Chowdhury
- Division of Life Sciences (Molecular Biology Major), Department of Bioactive Material Sciences, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Korea
| | - Jungeun An
- Division of Life Sciences (Life Sciences Major), Jeonbuk National University, Jeonju 54896, Korea
| | - Sangyun Jeong
- Division of Life Sciences (Molecular Biology Major), Department of Bioactive Material Sciences, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
16
|
Shen L, He Y, Chen S, He L, Zhang Y. PTHrP Modulates the Proliferation and Osteogenic Differentiation of Craniofacial Fibrous Dysplasia-Derived BMSCs. Int J Mol Sci 2023; 24:ijms24087616. [PMID: 37108778 PMCID: PMC10146947 DOI: 10.3390/ijms24087616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Fibrous dysplasia (FD) is a skeletal stem cell disease caused by mutations in the guanine nucleotide-binding protein, alpha-stimulating activity polypeptide (GNAS) gene, which results in the abnormal accumulation of cyclic adenosine monophosphate (cAMP) and hyperactivation of downstream signaling pathways. Parathyroid hormone-related protein (PTHrP) is secreted by the osteoblast lineage and is involved in various physiological and pathological activities of bone. However, the association between the abnormal expression of PTHrP and FD, as well as its underlying mechanism, remains unclear. In this study, we discovered that FD patient-derived bone marrow stromal cells (FD BMSCs) expressed significantly higher levels of PTHrP during osteogenic differentiation and exhibited greater proliferation capacity but impaired osteogenic ability compared to normal control patient-derived BMSCs (NC BMSCs). Continuous exogenous PTHrP exposure on the NC BMSCs promoted the FD phenotype in both in vitro and in vivo experiments. Through the PTHrP/cAMP/PKA axis, PTHrP could partially influence the proliferation and osteogenesis capacity of FD BMSCs via the overactivation of the Wnt/β-Catenin signaling pathway. Furthermore, PTHrP not only directly modulated cAMP/PKA/CREB transduction but was also demonstrated as a transcriptional target of CREB. This study provides novel insight into the possible pathogenesis involved in the FD phenotype and enhances the understanding of its molecular signaling pathways, offering theoretical evidence for the feasibility of potential therapeutic targets for FD.
Collapse
Affiliation(s)
- Lihang Shen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yang He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Shuo Chen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Linhai He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
- First Clinical Division, Peking University School and Hospital of Stomatology, Beijing 100034, China
| | - Yi Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| |
Collapse
|
17
|
Inhibition of late mRNA synthesis in the hippocampus impairs consolidation and reconsolidation of spatial memory in male rats. Neurobiol Learn Mem 2022; 195:107687. [DOI: 10.1016/j.nlm.2022.107687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/19/2022]
|
18
|
Hamed O, Joshi R, Mostafa MM, Giembycz MA. α and β Catalytic Subunits of cAMP-dependent Protein Kinase Regulate Formoterol-induced Inflammatory Gene Expression Changes in Human Bronchial Epithelial Cells. Br J Pharmacol 2022; 179:4593-4614. [PMID: 35735057 DOI: 10.1111/bph.15901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/27/2022] [Accepted: 06/18/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND & PURPOSE It has been proposed that genomic mechanisms contribute to the adverse-effects that are often experienced by asthmatic subjects who take regular, inhaled β2 -adrenoceptor agonists as a monotherapy. Moreover, data from preclinical models of asthma suggest that these gene expression changes are mediated by β-arrestin-2 rather than PKA. Herein, we tested this hypothesis by comparing the genomic effects of formoterol, a β2 -adrenoceptor agonist, with forskolin in human primary bronchial epithelial cells (HBEC). EXPERIMENTAL APPROACH Gene expression changes were determined by RNA-sequencing. Gene silencing and genome editing were employed to explore the roles of β-arrestin-2 and PKA. KEY RESULTS The formoterol-regulated transcriptome in HBEC treated concurrently with TNFα, was defined by 1480 unique gene expression changes. TNFα-induced transcripts modulated by formoterol were annotated with enriched gene ontology terms related to inflammation and proliferation, notably "GO:0070374~positive regulation of ERK1 and ERK2 cascade", which is an established β-arrestin-2 target. However, expression of the formoterol- and forskolin-regulated transcriptomes were highly rank-order correlated and the effects of formoterol on TNFα-induced inflammatory genes were abolished by an inhibitor of PKA. Furthermore, formoterol-induced gene expression changes in BEAS-2B bronchial epithelial cell clones deficient in β-arrestin-2 were comparable to those expressed by their parental counterparts. Contrariwise, gene expression was partially inhibited in clones lacking the α-catalytic subunit (Cα) of PKA and abolished following the additional knockdown of the β-catalytic subunit (Cβ) paralogue. CONCLUSIONS The effects of formoterol on inflammatory gene expression in airway epithelia are mediated by PKA and involve the cooperation of PKA-Cα and PKA-Cβ.
Collapse
Affiliation(s)
- Omar Hamed
- Airways Inflammation Research Group, Department of Physiology & Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Radhika Joshi
- Airways Inflammation Research Group, Department of Physiology & Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mahmoud M Mostafa
- Airways Inflammation Research Group, Department of Physiology & Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mark A Giembycz
- Airways Inflammation Research Group, Department of Physiology & Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
19
|
Zhao YY, Zhang LJ, Liang XY, Zhang XC, Chang JR, Shi M, Liu H, Zhou Y, Sun Z, Zhao YF. Pyruvate Upregulates Hepatic FGF21 Expression by Activating PDE and Inhibiting cAMP–Epac–CREB Signaling Pathway. Int J Mol Sci 2022; 23:ijms23105490. [PMID: 35628302 PMCID: PMC9141208 DOI: 10.3390/ijms23105490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) functions as a polypeptide hormone to regulate glucose and lipid metabolism, and its expression is regulated by cellular metabolic stress. Pyruvate is an important intermediate metabolite that acts as a key hub for cellular fuel metabolism. However, the effect of pyruvate on hepatic FGF21 expression and secretion remains unknown. Herein, we examined the gene expression and protein levels of FGF21 in human hepatoma HepG2 cells and mouse AML12 hepatocytes in vitro, as well as in mice in vivo. In HepG2 and AML12 cells, pyruvate at concentrations above 0.1 mM significantly increased FGF21 expression and secretion. The increase in cellular cAMP levels by adenylyl cyclase activation, phosphodiesterase (PDE) inhibition and 8-Bromo-cAMP administration significantly restrained pyruvate-stimulated FGF21 expression. Pyruvate significantly increased PDE activities, reduced cAMP levels and decreased CREB phosphorylation. The inhibition of exchange protein directed activated by cAMP (Epac) and cAMP response element binding protein (CREB) upregulated FGF21 expression, upon which pyruvate no longer increased FGF21 expression. The increase in plasma pyruvate levels in mice induced by the intraperitoneal injection of pyruvate significantly increased FGF21 gene expression and PDE activity with a reduction in cAMP levels and CREB phosphorylation in the mouse liver compared with the control. In conclusion, pyruvate activates PDEs to reduce cAMP and then inhibits the cAMP–Epac–CREB signaling pathway to upregulate FGF21 expression in hepatocytes.
Collapse
|
20
|
Xiong Z, Wang M, You S, Chen X, Lin J, Wu J, Shi X. Transcription Regulation of Tceal7 by the Triple Complex of Mef2c, Creb1 and Myod. BIOLOGY 2022; 11:biology11030446. [PMID: 35336819 PMCID: PMC8945367 DOI: 10.3390/biology11030446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary We have previously reported a striated muscle-specific gene during embryogenesis, Tceal7. Our studies have characterized the 0.7 kb promoter of the Tceal7 gene, which harbors important E-box motifs driving the LacZ reporter in the myogenic lineage. However, the underlying mechanism regulating the dynamic expression of Tceal7 during skeletal muscle regeneration is still elusive. In the present work, we have defined a cluster of Mef2#3–CRE#3–E#4 motifs through bioinformatic analysis and transcription assays. Our studies suggested that the triple complex of Mef2c, Creb1 and Myod binds to the Mef2#3–CRE#3–E#4 cluster region, therefore driving the dynamic expression of Tceal7 during skeletal muscle regeneration. The novel mechanism may throw new light on understanding transcription regulation in skeletal muscle myogenesis. Abstract Tceal7 has been identified as a direct, downstream target gene of MRF in the skeletal muscle. The overexpression of Tceal7 represses myogenic proliferation and promotes cell differentiation. Previous studies have defined the 0.7 kb upstream fragment of the Tceal7 gene. In the present study, we have further determined two clusters of transcription factor-binding motifs in the 0.7 kb promoter: CRE#2–E#1–CRE#1 in the proximal region and Mef2#3–CRE#3–E#4 in the distal region. Utilizing transcription assays, we have also shown that the reporter containing the Mef2#3–CRE#3–E#4 motifs is synergistically transactivated by Mef2c and Creb1. Further studies have mapped out the protein–protein interaction between Mef2c and Creb1. In summary, our present studies support the notion that the triple complex of Mef2c, Creb1 and Myod interacts with the Mef2#3–CRE#3–E#4 motifs in the distal region of the Tceal7 promoter, thereby driving Tceal7 expression during skeletal muscle development and regeneration.
Collapse
Affiliation(s)
- Zhenzhen Xiong
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; (Z.X.); (M.W.); (S.Y.); (X.C.); (J.W.)
| | - Mengni Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; (Z.X.); (M.W.); (S.Y.); (X.C.); (J.W.)
| | - Shanshan You
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; (Z.X.); (M.W.); (S.Y.); (X.C.); (J.W.)
| | - Xiaoyan Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; (Z.X.); (M.W.); (S.Y.); (X.C.); (J.W.)
| | - Jiangguo Lin
- Research Department of Medical Sciences, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China;
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jianhua Wu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; (Z.X.); (M.W.); (S.Y.); (X.C.); (J.W.)
| | - Xiaozhong Shi
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; (Z.X.); (M.W.); (S.Y.); (X.C.); (J.W.)
- Correspondence: ; Tel.: +86-20-39380620
| |
Collapse
|
21
|
Exogenous Parathyroid Hormone Alleviates Intervertebral Disc Degeneration through the Sonic Hedgehog Signalling Pathway Mediated by CREB. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9955677. [PMID: 35265269 PMCID: PMC8898813 DOI: 10.1155/2022/9955677] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/19/2022]
Abstract
As an important hormone that regulates the balance of calcium and phosphorus, parathyroid hormone (PTH) has also been found to have an important function in intervertebral disc degeneration (IVDD). Our aim was to investigate the mechanism by which PTH alleviates IVDD. In this study, the PTH 1 receptor was found to be highly expressed in severely degenerated human nucleus pulposus (NP) cells. We found in the mouse model of IVDD that supplementation with exogenous PTH alleviated the narrowing of the intervertebral space and the degradation of the extracellular matrix (ECM) caused by tail suspension (TS). In addition, inflammation, oxidative stress, and apoptosis levels were significantly increased in the intervertebral disc tissues of TS-induced mice, and the activity of NP cells was decreased. TS also led to the downregulation of Sonic hedgehog (SHH) signalling pathway-related signal molecules in NP cells such as SHH, Smoothened, and GLI1. However, supplementation with exogenous PTH can reverse these changes. In vitro, PTH also promotes the activity of NP cells and the secretion of ECM. However, the antagonist of the SHH signalling pathway can inhibit the therapeutic effect of PTH on NP cells. In addition, a cAMP-response element-binding protein, as an important transcription factor, was found to mediate the promotion of PTH on the SHH signalling pathway. Our results revealed that PTH can alleviate IVDD by inhibiting inflammation, oxidative stress, and apoptosis and improving the activity of NP cells via activating the SHH signalling pathway.
Collapse
|
22
|
Mori Y, Tsuchihira A, Yoshida T, Yoshida S, Fujiuchi A, Ohmi M, Isogai Y, Sakaguchi T, Eguchi S, Tsuda T, Kato K, Ohashi K, Ouchi N, Park HM, Murohara T, Takefuji M. Corticotropin releasing hormone receptor 2 antagonist, RQ-00490721, for the prevention of pressure overload-induced cardiac dysfunction. Biomed Pharmacother 2021; 146:112566. [PMID: 34954642 DOI: 10.1016/j.biopha.2021.112566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/14/2021] [Accepted: 12/19/2021] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND G protein-coupled receptors (GPCRs) regulate the pathological and physiological functions of the heart. GPCR antagonists are widely used in the treatment of chronic heart failure. Despite therapeutic advances in the treatments for cardiovascular diseases, heart failure is a major clinical health problem, with significant mortality and morbidity. Corticotropin releasing hormone receptor 2 (CRHR2) is highly expressed in cardiomyocytes, and cardiomyocyte-specific deletion of the genes encoding CRHR2 suppresses pressure overload-induced cardiac dysfunction. This suggests that the negative modulation of CRHR2 may prevent the progression of heart failure. However, there are no systemic drugs against CRHR2. FINDINGS We developed a novel, oral, small molecule antagonist of CRHR2, RQ-00490721, to investigate the inhibition of CRHR2 as a potential therapeutic approach for the treatment of heart failure. In vitro, RQ-00490721 decreased CRHR2 agonist-induced 3', 5'-cyclic adenosine monophosphate (cAMP) production. In vivo, RQ-00490721 showed sufficient oral absorption and better distribution to peripheral organs than to the central nervous system. Oral administration of RQ-00490721 inhibited the CRHR2 agonist-induced phosphorylation of cAMP-response element binding protein (CREB) in the heart, which regulates a transcription activator involved in heart failure. RQ-00490721 administration was not found to affect basal heart function in mice but protected them from pressure overload-induced cardiac dysfunction. INTERPRETATION Our results suggest that RQ-00490721 is a promising agent for use in the treatment of chronic heart failure.
Collapse
Affiliation(s)
- Yu Mori
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | | | - Tatsuya Yoshida
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Satoya Yoshida
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Akiyoshi Fujiuchi
- Discovery Research, RaQualia Pharma Inc., Nagoya, Japan; RaQualia Pharma Industry-Academia Collaborative Research Center, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Masashi Ohmi
- Discovery Research, RaQualia Pharma Inc., Nagoya, Japan
| | - Yumi Isogai
- Discovery Research, RaQualia Pharma Inc., Nagoya, Japan
| | - Teruhiro Sakaguchi
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Shunsuke Eguchi
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Takuma Tsuda
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Katsuhiro Kato
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Koji Ohashi
- Department of Molecular Medicine and Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Noriyuki Ouchi
- Department of Molecular Medicine and Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Hyi-Man Park
- Discovery Research, RaQualia Pharma Inc., Nagoya, Japan; RaQualia Pharma Industry-Academia Collaborative Research Center, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Mikito Takefuji
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan.
| |
Collapse
|
23
|
Robinson K, Yang Q, Li H, Zhang L, Aylward B, Arsenault RJ, Zhang G. Butyrate and Forskolin Augment Host Defense, Barrier Function, and Disease Resistance Without Eliciting Inflammation. Front Nutr 2021; 8:778424. [PMID: 34778349 PMCID: PMC8579826 DOI: 10.3389/fnut.2021.778424] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/04/2021] [Indexed: 12/13/2022] Open
Abstract
Host defense peptides (HDPs) are an integral part of the innate immune system with both antimicrobial and immunomodulatory activities. Induction of endogenous HDP synthesis is being actively explored as an antibiotic-alternative approach to disease control and prevention. Butyrate, a short-chain fatty acid, and forskolin, a phytochemical, have been shown separately to induce HDP gene expression in human cells. Here, we investigated the ability of butyrate and forskolin to induce the expressions of chicken HDP genes and the genes involved in barrier function such as mucin 2 and claudin 1 both in vitro and in vivo. We further evaluated their efficacy in protecting chickens from Clostridium perfringens-induced necrotic enteritis. Additionally, we profiled the transcriptome and global phosphorylation of chicken HD11 macrophage cells in response to butyrate and forskolin using RNA sequencing and a kinome peptide array, respectively. Our results showed a strong synergy between butyrate and forskolin in inducing the expressions of several, but not all, HDP genes. Importantly, dietary supplementation of butyrate and a forskolin-containing plant extract resulted in significant alleviation of intestinal lesions and the C. perfringens colonization in a synergistic manner in a chicken model of necrotic enteritis. RNA sequencing revealed a preferential increase in HDP and barrier function genes with no induction of proinflammatory cytokines in response to butyrate and forskolin. The antiinflammatory and barrier protective properties of butyrate and forskolin were further confirmed by the kinome peptide array. Moreover, we demonstrated an involvement of inducible cAMP early repressor (ICER)-mediated negative feedback in HDP induction by butyrate and forskolin. Overall, these results highlight a potential for developing butyrate and forskolin, two natural products, as novel antibiotic alternatives to enhance intestinal health and disease resistance in poultry and other animals.
Collapse
Affiliation(s)
- Kelsy Robinson
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States.,Poultry Production and Product Safety Research Unit, United States Department of Agriculture (USDA)-Agricultural Research Service, Fayetteville, AR, United States
| | - Qing Yang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Hong Li
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States.,College of Animal Science and Technology, Henan Agriculture University, Zhengzhou, China
| | - Long Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States.,Institute of Ecology, China West Normal University, Nanchong, China
| | - Bridget Aylward
- Department of Animal and Food Sciences, University of Delaware, Newark, DE, United States
| | - Ryan J Arsenault
- Department of Animal and Food Sciences, University of Delaware, Newark, DE, United States
| | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
24
|
Kim W, LeBlanc B, Matthews WL, Zhang ZY, Zhang Y. Advancements in chemical biology targeting the kinases and phosphatases of RNA polymerase II-mediated transcription. Curr Opin Chem Biol 2021; 63:68-77. [PMID: 33714893 PMCID: PMC8384638 DOI: 10.1016/j.cbpa.2021.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/21/2021] [Accepted: 02/01/2021] [Indexed: 11/30/2022]
Abstract
Phosphorylation of RNA polymerase II (RNAP II) coordinates the temporal progression of eukaryotic transcription. The development and application of chemical genetic methods have enhanced our ability to investigate the intricate and intertwined pathways regulated by the kinases and phosphatases targeting RNAP II to ensure transcription accuracy and efficiency. Although identifying small molecules that modulate these enzymes has been challenging due to their highly conserved structures, powerful new chemical biology strategies such as targeted covalent inhibitors and small molecule degraders have significantly improved chemical probe specificity. The recent success in discovering phosphatase holoenzyme activators and inhibitors, which demonstrates the feasibility of selective targeting of individual phosphatase complexes, opens up new avenues into the study of transcription. Herein, we summarize how chemical biology is used to delineate kinases' identities involved in RNAP II regulation and new concepts in inhibitor/activator design implemented for kinases/phosphatases involved in modulating RNAP II-mediated transcription.
Collapse
Affiliation(s)
- Wantae Kim
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Blase LeBlanc
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, 78712, USA
| | - Wendy L Matthews
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, 78712, USA
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Department of Chemistry, and Institute for Drug Discovery, Purdue University, West Lafayette, IN, 47907, USA
| | - Yan Zhang
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, 78712, USA; The Institute for Cellular and Molecular Biology. University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
25
|
Fu Y, Xin Z, Ling Z, Xie H, Xiao T, Shen X, Lin J, Xu L, Jiang H. A CREB1-miR-181a-5p loop regulates the pathophysiologic features of bone marrow stromal cells in fibrous dysplasia of bone. Mol Med 2021; 27:81. [PMID: 34294046 PMCID: PMC8296714 DOI: 10.1186/s10020-021-00341-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Fibrous dysplasia (FD) is a bone marrow stromal cell (BMSC) disease caused by activating mutations of guanine nucleotide-binding protein alpha-stimulating activity polypeptide (GNAS) and is characterized by increased proliferative activity and disrupted osteogenesis of BMSCs. However, the molecular mechanisms regulating the pathophysiologic features of BMSCs in FD remain unknown. This study aimed to identify and verify the roles of the CREB1-miR-181a-5p regulatory loop in FD pathophysiology. METHODS MicroRNA (miRNA) sequencing analysis was used to identify the possible miRNAs implicated in FD. The proliferation, apoptosis, and osteogenic differentiation of BMSCs, as well as the osteoclast-induced phenotype, were measured and compared after exogenous miR-181a-5p transfection into FD BMSCs or miR-181a-5p inhibitor transfection into normal BMSCs. Chromatin immunoprecipitation and luciferase reporter assays were performed to verify the interactions between CREB1 and miR-181a-5p and their effects on the FD pathological phenotype. RESULTS Compared to normal BMSCs, FD BMSCs showed decreased miR-181a-5p levels and exhibited increased proliferative activity, decreased apoptotic capacity, and impaired osteogenesis. FD BMSCs also showed a stronger osteoclast activation effect. miR-181a-5p overexpression reversed the pathophysiologic features of FD BMSCs, whereas miR-181a-5p suppression induced an FD-like phenotype in normal BMSCs. Mechanistically, miR-181a-5p was the downstream target of CREB1, and CREB1 was posttranscriptionally regulated by miR-181a-5p. CONCLUSIONS Our study identifies that the interaction loop between CREB1 and miR-181a-5p plays a crucial role in regulating the pathophysiologic features of FD BMSCs. MiR-181a-5p may be a potential therapeutic target for the treatment of FD.
Collapse
Affiliation(s)
- Yu Fu
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China. .,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China. .,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China.
| | - Zhili Xin
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Ziji Ling
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Hanyu Xie
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Tao Xiao
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Xin Shen
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Jialin Lin
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China
| | - Ling Xu
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China
| | - Hongbing Jiang
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China. .,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China. .,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
26
|
Li Y, Chen L, Zhao W, Sun L, Zhang R, Zhu S, Xie K, Feng X, Wu X, Sun Z, Shu G, Wang S, Gao P, Zhu X, Wang L, Jiang Q. Food reward depends on TLR4 activation in dopaminergic neurons. Pharmacol Res 2021; 169:105659. [PMID: 33971268 DOI: 10.1016/j.phrs.2021.105659] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/30/2021] [Accepted: 04/30/2021] [Indexed: 01/07/2023]
Abstract
The rising prevalence of obesity and being overweight is a worldwide health concern. Food reward dysregulation is the basic factor for the development of obesity. Dopamine (DA) neurons in the ventral tegmental area (VTA) play a vital role in food reward. Toll-like receptor 4 (TLR4) is a transmembrane pattern recognition receptor that can be activated by saturated fatty acids. Here, we show that the deletion of TLR4 specifically in DA neurons increases body weight, increases food intake, and decreases food reward. Conditional deletion of TLR4 also decreased the activity of DA neurons while suppressing the expression of tyrosine hydroxylase (TH) in the VTA, which regulates the concentration of DA in the nucleus accumbens (NAc) to affect food reward. Meanwhile, AAV-Cre-GFP mediated VTA-specific TLR4-deficient mice recapitulates food reward of DAT-TLR4-KO mice. Food reward could be rescued by re-expressing TLR4 in VTA DA neurons. Moreover, effects of intra-VTA infusion of lauric acid (a saturated fatty acid with 12 carbon) on food reward were abolished in mice lacking TLR4 in DA neurons. Our study demonstrates the critical role of TLR4 signaling in regulating the activity of VTA DA neurons and the normal function of the mesolimbic DA system that may contribute to food reward.
Collapse
Affiliation(s)
- Yongxiang Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Lvshuang Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Weijie Zhao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Lijuan Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Ruixue Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Shuqing Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Kailai Xie
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Xiajie Feng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Xin Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Zhonghua Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Xiaotong Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.
| |
Collapse
|
27
|
Pentoxifylline Enhances Antioxidative Capability and Promotes Mitochondrial Biogenesis in D-Galactose-Induced Aging Mice by Increasing Nrf2 and PGC-1 α through the cAMP-CREB Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6695613. [PMID: 34257818 PMCID: PMC8245236 DOI: 10.1155/2021/6695613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/21/2021] [Accepted: 05/13/2021] [Indexed: 02/07/2023]
Abstract
Aging is a complex phenomenon associated with oxidative stress and mitochondrial dysfunction. The objective of this study was to investigate the potential ameliorative effects of the phosphodiesterase inhibitor pentoxifylline (PTX) on the aging process and its underlying mechanisms. We treated D-galactose- (D-gal-) induced aging mice with PTX and measured the changes in behavior, degree of oxidative damage, and mitochondrial ultrastructure and content as well as the expression of nuclear factor erythroid 2-related factor 2- (Nrf2-) mediated antioxidant genes and peroxisome proliferator-activated receptor-gamma coactivator 1-alpha- (PGC-1α-) dependent mitochondrial biogenesis genes. The results demonstrated that PTX improved cognitive deficits, reduced oxidative damage, ameliorated abnormal mitochondrial ultrastructure, increased mitochondrial content and Nrf2 activation, and upregulated antioxidant and mitochondrial biogenesis gene expression in the hippocampus of wild-type aging mice. However, the above antiaging effects of PTX were obviously decreased in the brains of Nrf2-deficient D-gal-induced aging mice. Moreover, in hydrogen peroxide-treated SH-SY5Y cells, we found that cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB) and Nrf2/PGC-1α act in a linear way by CREB siRNA transfection. Thus, PTX administration improved the aging-related decline in brain function by enhancing antioxidative capability and promoting mitochondrial biogenesis, which might depend on increasing Nrf2 and PGC-1α by activating the cAMP-CREB pathway.
Collapse
|
28
|
Biringer RG. A Review of Prostanoid Receptors: Expression, Characterization, Regulation, and Mechanism of Action. J Cell Commun Signal 2021; 15:155-184. [PMID: 32970276 PMCID: PMC7991060 DOI: 10.1007/s12079-020-00585-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022] Open
Abstract
Prostaglandin signaling controls a wide range of biological processes from blood pressure homeostasis to inflammation and resolution thereof to the perception of pain to cell survival. Disruption of normal prostanoid signaling is implicated in numerous disease states. Prostaglandin signaling is facilitated by G-protein-coupled, prostanoid-specific receptors and the array of associated G-proteins. This review focuses on the expression, characterization, regulation, and mechanism of action of prostanoid receptors with particular emphasis on human isoforms.
Collapse
Affiliation(s)
- Roger G Biringer
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Blvd, Bradenton, FL, 34211, USA.
| |
Collapse
|
29
|
Deng Y, Liu B, Fu C, Gao L, Shen Y, Liu K, Li Q, Cao J, Mao W. Lipopolysaccharide stimulates bovine endometrium explants through toll‑like receptor 4 signaling and PGE 2 synthesis. Prostaglandins Leukot Essent Fatty Acids 2021; 168:102272. [PMID: 33895679 DOI: 10.1016/j.plefa.2021.102272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/09/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023]
Abstract
Bovine endometrium infection with gram-negative bacteria commonly causes uterine diseases. Previous studies indicate that prostaglandin E2 (PGE2) is an inflammatory mediator in bacterial endometritis. However, the mechanism underlying lipopolysaccharide (LPS)-induced inflammatory response regulation in bovine endometrial explants remains elusive. In the present study, bovine explants were pre-treated with 15-hydroxyprostaglandin dehydrogenase (15-PGDH) inhibitors before LPS stimulation. PGE2 secretion, prostaglandin synthetase, pro-inflammatory factor, damage-associated molecular pattern (DAMP), and related signaling pathway factor levels were evaluated. Using 15-PGDH inhibitors pre-treatment, LPS-treated bovine endometrial explants exhibited augmentation of PGE2 and DAMP expression, and upregulation of various signaling pathway factors. Protein kinase A (PKA), extracellular-signal-regulated kinase, and c-Jun N-terminal kinase phosphorylation and degradation of nuclear transcription factor-κB (NF-κB) inhibitors were induced in the pre-treated endometrial explants. The mechanism underlying LPS-induced PGE2 accumulation acting as a pro-inflammatory mediator through toll-like receptor 4 signaling in bovine explants could involve the PKA, mitogen-activated protein kinase, and NF-κB pathways.
Collapse
Affiliation(s)
- Yang Deng
- School of Public Health, Baotou Medical College, 014040, Baotou, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, 010018, Hohhot, China
| | - Bo Liu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, 010018, Hohhot, China
| | - Changqi Fu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, 010018, Hohhot, China
| | - Long Gao
- School of Public Health, Baotou Medical College, 014040, Baotou, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, 010018, Hohhot, China
| | - Yuan Shen
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, 010018, Hohhot, China
| | - Kun Liu
- School of Public Health, Baotou Medical College, 014040, Baotou, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, 010018, Hohhot, China
| | - Qianru Li
- School of Public Health, Baotou Medical College, 014040, Baotou, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, 010018, Hohhot, China
| | - Jinshan Cao
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, 010018, Hohhot, China.
| | - Wei Mao
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, 010018, Hohhot, China.
| |
Collapse
|
30
|
Cysteinyl-specialized proresolving mediators link resolution of infectious inflammation and tissue regeneration via TRAF3 activation. Proc Natl Acad Sci U S A 2021; 118:2013374118. [PMID: 33649212 PMCID: PMC7958394 DOI: 10.1073/pnas.2013374118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The recently elucidated proresolving conjugates in tissue regeneration (CTR) maresin-CTR (MCTR), protectin-CTR (PCTR), and resolvin-CTR (RCTR), termed cysteinyl-specialized proresolving mediators (cys-SPMs) each promotes regeneration, controls infection, and accelerates resolution of inflammation. Here, we sought evidence for cys-SPM activation of primordial pathways in planaria (Dugesia japonica) regeneration that might link resolution of inflammation and regeneration. On surgical resection, planaria regeneration was enhanced with MCTR3, PCTR3, or RCTR3 (10 nM), each used for RNA sequencing. The three cys-SPMs shared up-regulation of 175 known transcripts with fold-change > 1.25 and combined false discovery rate (FDR) < 0.002, and 199 canonical pathways (FDR < 0.25), including NF-κB pathways and an ortholog of human TRAF3 (TNFR-associated factor 3). Three separate pathway analyses converged on TRAF3 up-regulation by cys-SPMs. With human macrophages, three cys-SPMs each dose-dependently increased TRAF3 expression in a cAMP-PKA-dependent manner. TRAF3 overexpression in macrophages enhanced Interleukin-10 (IL-10) and phagocytosis of Escherichia coli IL-10 also increased phagocytosis in a dose-dependent manner. Silencing of mouse TRAF3 in vivo significantly reduced IL-10 and macrophage phagocytosis. TRAF3 silencing in vivo also relieved cys-SPMs' actions in limiting polymorphonuclear neutrophil in E. coli exudates. These results identify cys-SPM-regulated pathways in planaria regeneration, uncovering a role for TRAF3/IL-10 in regulating mammalian phagocyte functions in resolution. Cys-SPM activation of TRAF3 signaling is a molecular component of both regeneration and resolution of infectious inflammation.
Collapse
|
31
|
Androgen Receptor Stimulates Hexokinase 2 and Induces Glycolysis by PKA/CREB Signaling in Hepatocellular Carcinoma. Dig Dis Sci 2021; 66:802-813. [PMID: 32274668 DOI: 10.1007/s10620-020-06229-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/20/2020] [Indexed: 01/26/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) escapes growth inhibition by upregulating hexokinase 2 (HK2); however, the mechanism by which tumor cells upregulate HK2 remains unclear. AIM We aimed to investigate the role of androgen receptor (AR) signalling in promoting HK2 expression in HCC. METHODS The expressions of AR and HK2 in HCC tissues were analyzed by immunohistochemistry. Cell proliferation was determined using the CCK-8 assay, and the molecular mechanism of AR in the regulation of HK2 was evaluated by immunoblotting and luciferase assays. RESULTS AR expression is positively correlated with HK2 staining by an immunohistochemical analysis. The manipulation of AR expression changed HK2 expression and glycolysis. AR signaling promoted the growth of HCC by enhancing HK2-mediated glycolysis. Moreover, AR stimulated HK2 levels and glycolysis by potentiating protein kinase A/cyclic adenosine monophosphate response element-binding (CREB) protein signaling. CREB silencing decreased HK2 expression and inhibited AR-mediated HCC glycolysis. AR affected the sensitivity of HCC cells to glycolysis inhibitors by regulating downstream phosphorylated (p)-CREB. CONCLUSIONS These results indicate that AR at least partially induced glycolysis via p-CREB regulation of HK2 in HCC cells. Thus, this pathway should be considered for the design of novel therapeutic methods to target AR-overexpressing HCC.
Collapse
|
32
|
Huang W, Zhu L, Cao G, Xie P, Song Y, Huang J, Chen X, Cai Z. Integrated Proteomics and Metabolomics Assessment Indicated Metabolic Alterations in Hypothalamus of Mice Exposed to Triclosan. Chem Res Toxicol 2021; 34:1319-1328. [PMID: 33611912 DOI: 10.1021/acs.chemrestox.0c00514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Triclosan (TCS) is a ubiquitous antimicrobial used in many daily consumer products. It has been reported to induce endocrine disrupting effects at low doses in mammals, disturbing sex hormone function and thyroid function. The hypothalamus plays a crucial role in the maintenance of neuroendocrine function and energy homeostasis. We speculated that the adverse effects of TCS might be related to the disturbance of metabolic processes in hypothalamus. The present study aimed at investigating the effects of TCS exposure on the protein and metabolite profiles in hypothalamus of mice. Male C57BL/6 mice were orally exposed to TCS at the dosage of 10 mg/kg/d for 13 weeks. The hypothalamus was isolated and processed for mass spectrometry (MS)-based proteomics and metabolomics analyses. The results showed that a 10.6% decrease (P = 0.066) in body weight gain was observed in the TCS exposure group compared with vehicle control group. Differential analysis defined 52 proteins and 57 metabolites that delineated TCS exposed mice from vehicle controls. Among the differential features, multiple proteins and metabolites were found to play vital roles in neuronal signaling and function. Bioinformatics analysis revealed that these differentially expressed proteins and metabolites were involved in four major biological processes, including glucose metabolism, purine metabolism, neurotransmitter release, and neural plasticity, suggesting the disturbance of homeostasis in energy metabolism, mitochondria function, neurotransmitter system, and neuronal function. Our results may provide insights into the neurotoxicity of TCS and extend our understanding of the biological effects induced by TCS exposure.
Collapse
Affiliation(s)
- Wei Huang
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China.,School of Environment, Jinan University, Guangzhou 510632, China
| | - Lin Zhu
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China
| | - Guodong Cao
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China
| | - Peisi Xie
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yuanyuan Song
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jialing Huang
- School of Environment, Jinan University, Guangzhou 510632, China
| | - Xiangfeng Chen
- Shandong Analysis and Test Center, Qilu University of Technology, Jinan, Shandong, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
33
|
Al-Hakeim HK, Al-Mayali HH, Moustafa SR, Maes M. Cytokine dependent hematopoietic cell linker (CLNK) is highly elevated in blood transfusion dependent beta-thalassemia major patients. Transfus Clin Biol 2021; 28:194-198. [PMID: 33453373 DOI: 10.1016/j.tracli.2021.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/23/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Transfusion-dependent β-thalassemia (TDT) is a severe form of thalassemia caused by mutations in the β-globin gene, resulting in partial or complete deficiency of β-globin chains. This deficiency results in oxidative stress, dyserythropoiesis, and chronic anemia. Cytokine-dependent hematopoietic cell linker (CLNK) belongs to adaptor proteins that have the capacity to interact with multiple signalling proteins and function in the organisation of the molecular components required for signal transduction. OBJECTIVES This is the first study which measured serum CLNK in TDT patients and examines the correlation between CLNK and iron overload biomarkers. PATIENTS AND METHODS Sixty children with TDT and 30 normal children (aged 3-12 years old) participated in the present study. The patients were on blood transfusion as a part of their treatment regimen. Serum C-reactive protein was negative in all samples. RESULTS The results showed significantly higher (P<0.001) serum CLNK levels in TDT patients as compared with controls. The TDT diagnosis explained 19.4% of the variance in CLNK levels. The increased levels of CLNK were significantly associated with indicants of iron overload, namely increased ferritin levels. CONCLUSIONS Increased CLNK levels in TDT may be explained by reciprocal effects between immune signalling and immature erythrocytes, which release soluble receptors and signalling molecules, including CLNK, in the blood.
Collapse
Affiliation(s)
- H K Al-Hakeim
- Department of Chemistry, College of Science, University of Kufa, Najaf, Iraq.
| | - H H Al-Mayali
- Department of Chemistry, College of Science, University of Kufa, Najaf, Iraq.
| | - S R Moustafa
- Clinical analysis department, College of Pharmacy, Hawler Medical University, Havalan City, Erbil, Iraq.
| | - M Maes
- Department of psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of psychiatry, Medical University Plovdiv, Plovdiv, Bulgaria; IMPACT research centre, Deakin University, Geelong, Australia.
| |
Collapse
|
34
|
Gao X, Zhang D, Xu C, Li H, Caron KM, Frenette PS. Nociceptive nerves regulate haematopoietic stem cell mobilization. Nature 2021; 589:591-596. [PMID: 33361809 PMCID: PMC7856173 DOI: 10.1038/s41586-020-03057-y] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/19/2020] [Indexed: 11/17/2022]
Abstract
Haematopoietic stem cells (HSCs) reside in specialized microenvironments in the bone marrow-often referred to as 'niches'-that represent complex regulatory milieux influenced by multiple cellular constituents, including nerves1,2. Although sympathetic nerves are known to regulate the HSC niche3-6, the contribution of nociceptive neurons in the bone marrow remains unclear. Here we show that nociceptive nerves are required for enforced HSC mobilization and that they collaborate with sympathetic nerves to maintain HSCs in the bone marrow. Nociceptor neurons drive granulocyte colony-stimulating factor (G-CSF)-induced HSC mobilization via the secretion of calcitonin gene-related peptide (CGRP). Unlike sympathetic nerves, which regulate HSCs indirectly via the niche3,4,6, CGRP acts directly on HSCs via receptor activity modifying protein 1 (RAMP1) and the calcitonin receptor-like receptor (CALCRL) to promote egress by activating the Gαs/adenylyl cyclase/cAMP pathway. The ingestion of food containing capsaicin-a natural component of chili peppers that can trigger the activation of nociceptive neurons-significantly enhanced HSC mobilization in mice. Targeting the nociceptive nervous system could therefore represent a strategy to improve the yield of HSCs for stem cell-based therapeutic agents.
Collapse
Affiliation(s)
- Xin Gao
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Dachuan Zhang
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,These authors contributed equally
| | - Chunliang Xu
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,These authors contributed equally
| | - Huihui Li
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kathleen M. Caron
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Paul S. Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
35
|
Verbenko DA, Karamova AE, Artamonova OG, Deryabin DG, Rakitko A, Chernitsov A, Krasnenko A, Elmuratov A, Solomka VS, Kubanov AA. Apremilast Pharmacogenomics in Russian Patients with Moderate-to-Severe and Severe Psoriasis. J Pers Med 2020; 11:jpm11010020. [PMID: 33383665 PMCID: PMC7823747 DOI: 10.3390/jpm11010020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/18/2020] [Accepted: 12/25/2020] [Indexed: 12/14/2022] Open
Abstract
One of the target drugs for plaque psoriasis treatment is apremilast, which is a selective phosphodiesterase 4 (PDE4) inhibitor. In this study, 34 moderate-to-severe and severe plaque psoriasis patients from Russia were treated with apremilast for 26 weeks. This allowed us to observe the effectiveness of splitting patient cohorts based on clinical outcomes, which were assessed using the Psoriasis Area Severity Index (PASI). In total, 14 patients (41%) indicated having an advanced outcome with delta PASI 75 after treatment; 20 patients indicated having moderate or no effects. Genome variability was investigated using the Illumina Infinium Global Screening Array. Genome-wide analysis revealed apremilast therapy clinical outcome associations at three compact genome regions with undefined functions situated on chromosomes 2, 4, and 5, as well as on a single single-nucleotide polymorphism (SNP) on chromosome 23. Pre-selected SNP sets were associated with psoriasis vulgaris analysis, which was used to identify four SNP-associated targeted therapy efficiencies: IL1β (rs1143633), IL4 (IL13) (rs20541), IL23R (rs2201841), and TNFα (rs1800629) genes. Moreover, we showed that the use of the global polygenic risk score allowed for the prediction of onset psoriasis in Russians. Therefore, these results can serve as a starting point for creating a predictive model of apremilast therapy response in the targeted therapy of patients with psoriasis vulgaris.
Collapse
Affiliation(s)
- Dmitry A. Verbenko
- State Research Center of Dermatovenereology and Cosmetology, Korolenko St., 3, bldg 6, 107076 Moscow, Russia; (A.E.K.); (O.G.A.); (D.G.D.); (V.S.S.); (A.A.K.)
- Correspondence:
| | - Arfenya E. Karamova
- State Research Center of Dermatovenereology and Cosmetology, Korolenko St., 3, bldg 6, 107076 Moscow, Russia; (A.E.K.); (O.G.A.); (D.G.D.); (V.S.S.); (A.A.K.)
| | - Olga G. Artamonova
- State Research Center of Dermatovenereology and Cosmetology, Korolenko St., 3, bldg 6, 107076 Moscow, Russia; (A.E.K.); (O.G.A.); (D.G.D.); (V.S.S.); (A.A.K.)
| | - Dmitry G. Deryabin
- State Research Center of Dermatovenereology and Cosmetology, Korolenko St., 3, bldg 6, 107076 Moscow, Russia; (A.E.K.); (O.G.A.); (D.G.D.); (V.S.S.); (A.A.K.)
| | - Alexander Rakitko
- Genotek Ltd., Nastavnicheskiipereulok 17/1, 105120 Moscow, Russia; (A.R.); (A.C.); (A.K.); (A.E.)
| | - Alexandr Chernitsov
- Genotek Ltd., Nastavnicheskiipereulok 17/1, 105120 Moscow, Russia; (A.R.); (A.C.); (A.K.); (A.E.)
| | - Anna Krasnenko
- Genotek Ltd., Nastavnicheskiipereulok 17/1, 105120 Moscow, Russia; (A.R.); (A.C.); (A.K.); (A.E.)
| | - Artem Elmuratov
- Genotek Ltd., Nastavnicheskiipereulok 17/1, 105120 Moscow, Russia; (A.R.); (A.C.); (A.K.); (A.E.)
| | - Victoria S. Solomka
- State Research Center of Dermatovenereology and Cosmetology, Korolenko St., 3, bldg 6, 107076 Moscow, Russia; (A.E.K.); (O.G.A.); (D.G.D.); (V.S.S.); (A.A.K.)
| | - Alexey A. Kubanov
- State Research Center of Dermatovenereology and Cosmetology, Korolenko St., 3, bldg 6, 107076 Moscow, Russia; (A.E.K.); (O.G.A.); (D.G.D.); (V.S.S.); (A.A.K.)
| |
Collapse
|
36
|
Sapio L, Salzillo A, Ragone A, Illiano M, Spina A, Naviglio S. Targeting CREB in Cancer Therapy: A Key Candidate or One of Many? An Update. Cancers (Basel) 2020; 12:3166. [PMID: 33126560 PMCID: PMC7693618 DOI: 10.3390/cancers12113166] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Intratumor heterogeneity (ITH) is considered the major disorienting factor in cancer treatment. As a result of stochastic genetic and epigenetic alterations, the appearance of a branched evolutionary shape confers tumor plasticity, causing relapse and unfavorable clinical prognosis. The growing evidence in cancer discovery presents to us "the great paradox" consisting of countless potential targets constantly discovered and a small number of candidates being effective in human patients. Among these, cyclic-AMP response element-binding protein (CREB) has been proposed as proto-oncogene supporting tumor initiation, progression and metastasis. Overexpression and hyperactivation of CREB are frequently observed in cancer, whereas genetic and pharmacological CREB downregulation affects proliferation and apoptosis. Notably, the present review is designed to investigate the feasibility of targeting CREB in cancer therapy. In particular, starting with the latest CREB evidence in cancer pathophysiology, we evaluate the advancement state of CREB inhibitor design, including the histone lysine demethylases JMJD3/UTX inhibitor GSKJ4 that we newly identified as a promising CREB modulator in leukemia cells. Moreover, an accurate analysis of strengths and weaknesses is also conducted to figure out whether CREB can actually represent a therapeutic candidate or just one of the innumerable preclinical cancer targets.
Collapse
Affiliation(s)
| | | | | | | | | | - Silvio Naviglio
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (L.S.); (A.S.); (A.R.); (M.I.); (A.S.)
| |
Collapse
|
37
|
Zhou N, Chen X, Xi J, Ma B, Leimena C, Stoll S, Qin G, Wang C, Qiu H. Novel genomic targets of valosin-containing protein in protecting pathological cardiac hypertrophy. Sci Rep 2020; 10:18098. [PMID: 33093614 PMCID: PMC7582185 DOI: 10.1038/s41598-020-75128-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 10/12/2020] [Indexed: 12/22/2022] Open
Abstract
Pressure overload-induced cardiac hypertrophy, such as that caused by hypertension, is a key risk factor for heart failure. However, the underlying molecular mechanisms remain largely unknown. We previously reported that the valosin-containing protein (VCP), an ATPase-associated protein newly identified in the heart, acts as a significant mediator of cardiac protection against pressure overload-induced pathological cardiac hypertrophy. Still, the underlying molecular basis for the protection is unclear. This study used a cardiac-specific VCP transgenic mouse model to understand the transcriptomic alterations induced by VCP under the cardiac stress caused by pressure overload. Using RNA sequencing and comprehensive bioinformatic analysis, we found that overexpression of the VCP in the heart was able to normalize the pressure overload-stimulated hypertrophic signals by activating G protein-coupled receptors, particularly, the olfactory receptor family, and inhibiting the transcription factor controlling cell proliferation and differentiation. Moreover, VCP overexpression restored pro-survival signaling through regulating alternative splicing alterations of mitochondrial genes. Together, our study revealed a novel molecular regulation mediated by VCP under pressure overload that may bring new insight into the mechanisms involved in protecting against hypertensive heart failure.
Collapse
Affiliation(s)
- Ning Zhou
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA.,Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Xin Chen
- Center for Genomics and Department of Basic Sciences, School of Medicine, Loma Linda University, 11021 Campus Street, AH 120/104, Loma Linda, CA, 92350, USA
| | - Jing Xi
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Ben Ma
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA.,Center of Molecular and Translational Medicine, Institution of Biomedical Science, Georgia State University, Petit Research Center, Room 588, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Christiana Leimena
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Shaunrick Stoll
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama At Birmingham, Birmingham, AL, 35294, USA
| | - Charles Wang
- Center for Genomics and Department of Basic Sciences, School of Medicine, Loma Linda University, 11021 Campus Street, AH 120/104, Loma Linda, CA, 92350, USA.
| | - Hongyu Qiu
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA. .,Center of Molecular and Translational Medicine, Institution of Biomedical Science, Georgia State University, Petit Research Center, Room 588, 100 Piedmont Ave, Atlanta, GA, 30303, USA.
| |
Collapse
|
38
|
van der Horst J, Greenwood IA, Jepps TA. Cyclic AMP-Dependent Regulation of Kv7 Voltage-Gated Potassium Channels. Front Physiol 2020; 11:727. [PMID: 32695022 PMCID: PMC7338754 DOI: 10.3389/fphys.2020.00727] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/04/2020] [Indexed: 01/08/2023] Open
Abstract
Voltage-gated Kv7 potassium channels, encoded by KCNQ genes, have major physiological impacts cardiac myocytes, neurons, epithelial cells, and smooth muscle cells. Cyclic adenosine monophosphate (cAMP), a well-known intracellular secondary messenger, can activate numerous downstream effector proteins, generating downstream signaling pathways that regulate many functions in cells. A role for cAMP in ion channel regulation has been established, and recent findings show that cAMP signaling plays a role in Kv7 channel regulation. Although cAMP signaling is recognized to regulate Kv7 channels, the precise molecular mechanism behind the cAMP-dependent regulation of Kv7 channels is complex. This review will summarize recent research findings that support the mechanisms of cAMP-dependent regulation of Kv7 channels.
Collapse
Affiliation(s)
- Jennifer van der Horst
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Iain A Greenwood
- Molecular and Clinical Sciences Institute, St. George's University of London, London, United Kingdom
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
39
|
Illiano M, Conte M, Salzillo A, Ragone A, Spina A, Nebbioso A, Altucci L, Sapio L, Naviglio S. The KDM Inhibitor GSKJ4 Triggers CREB Downregulation via a Protein Kinase A and Proteasome-Dependent Mechanism in Human Acute Myeloid Leukemia Cells. Front Oncol 2020; 10:799. [PMID: 32582541 PMCID: PMC7289982 DOI: 10.3389/fonc.2020.00799] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/23/2020] [Indexed: 01/02/2023] Open
Abstract
Acute myeloid leukemia (AML) is a progressive hematopoietic-derived cancer arising from stepwise genetic mutations of the myeloid lineage. cAMP response element-binding protein (CREB) is a nuclear transcription factor, which plays a key role in the multistep process of leukemogenesis, thus emerging as an attractive potential drug target for AML treatment. Since epigenetic dysregulations, such as DNA methylation, histone modifications, as well as chromatin remodeling, are a frequent occurrence in AML, an increasing and selective number of epi-drugs are emerging as encouraging therapeutic agents. Here, we demonstrate that the histone lysine demethylases (KDMs) JMJD3/UTX inhibitor GSKJ4 results in both proliferation decrease and CREB protein downregulation in AML cells. We found that GSKJ4 clearly decreases CREB protein, but not CREB mRNA levels. By cycloheximide assay, we provide evidence that GSKJ4 reduces CREB protein stability; moreover, proteasome inhibition largely counteracts the GSKJ4-induced CREB downregulation. Very interestingly, a rapid CREB phosphorylation at the Ser133 residue precedes CREB protein decrease in response to GSKJ4 treatment. In addition, protein kinase A (PKA) inhibition, but not extracellular signal-regulated kinase (ERK)1/2 inhibition, almost completely prevents both GSKJ4-induced p-Ser133-CREB phosphorylation and CREB protein downregulation. Overall, our study enforces the evidence regarding CREB as a potential druggable target, identifies the small epigenetic molecule GSKJ4 as an "inhibitor" of CREB, and encourages the design of future GSKJ4-based studies for the development of innovative approaches for AML therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Luigi Sapio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Silvio Naviglio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
40
|
Wu Y, Li Y, Liu J, Chen M, Li W, Chen Y, Xu M. The diagnostic value of extracellular protein kinase A (ECPKA) in serum for gastric and colorectal cancer. Transl Cancer Res 2020; 9:3870-3878. [PMID: 35117754 PMCID: PMC8797401 DOI: 10.21037/tcr-20-764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/24/2020] [Indexed: 01/26/2023]
Abstract
Background For the biomarkers of cancer, many chromosomal and genetic alterations have been examined as possible. However, some tumors do not display a clear molecular and genetic signature. While there are some cellular processes regulated by second messenger intracellular pathways indeed involved in carcinogenesis. The first intracellular second messenger was described as cyclic adenosine 3',5'-monophosphate (cAMP), and cAMP-dependent protein kinase (PKA) play a crucial role in several biological processes; The dysregulation of PKA-mediated signaling in several types of cancer should be investigated. More interesting, the alpha catalytic subunit of PKA (PKACα) could be secreted into the conditioned medium by different types of cancer cells, and it also existed in the serum of some cancer patients, defined as extracellular protein kinase A (ECPKA). Methods The levels of serum PKACα from healthy people, gastric cancer and colon cancer patients were detected by ELISA kits. Western blotting was used to detect the expression of PKACα in cancer tissue and the adjacent mucosa. Mann-Whitney test was applied to analyze the patients’ characteristics and serum PKACα. ROC analyses were performed to further evaluate the utility of PKACα in cancer diagnosis. The correlation of serum PKACα and T stage, age, and tumor markers were analyzed by Spearman rank and Pearson correlation analysis, respectively. Results There were significant differences of PKACα in serum between the volunteers and the gastric cancers (P<0.01), but not the colorectal cancers (P>0.05). ROC analyses evaluated the utility of PKACα for gastric cancer with 61.90% sensitivities and 87.50% specificities. The serum PKACα was correlated with tumor marker CA50, while there was no significant difference of PKACα expression between the gastric/colorectal cancer tissue and the adjacent mucosa. Conclusions The above results implied that PKACα levels might be a potential biomarker for the early screening of gastric cancers. Moreover, further research is still needed to investigate the role of secreted PKACα and the regulatory mechanism in tumor progression.
Collapse
Affiliation(s)
- Yan Wu
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yafang Li
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Junqiang Liu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Mengjiao Chen
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Wei Li
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yongchang Chen
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
41
|
Gao ZZ, Li YC, Shao CY, Xiao J, Shen Y, Zhou L. EPAC Negatively Regulates Myelination via Controlling Proliferation of Oligodendrocyte Precursor Cells. Neurosci Bull 2020; 36:639-648. [PMID: 32303914 DOI: 10.1007/s12264-020-00495-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 01/05/2020] [Indexed: 12/17/2022] Open
Abstract
Increasing evidence suggests that a cyclic adenosine monophosphate (cAMP)-dependent intracellular signal drives the process of myelination. Yet, the signal transduction underlying the action of cAMP on central nervous system myelination remains undefined. In the present work, we sought to determine the role of EPAC (exchange protein activated by cAMP), a downstream effector of cAMP, in the development of the myelin sheath using EPAC1 and EPAC2 double-knockout (EPACdKO) mice. The results showed an age-dependent regulatory effect of EPAC1 and EPAC2 on myelin development, as their deficiency caused more myelin sheaths in postnatal early but not late adult mice. Knockout of EPAC promoted the proliferation of oligodendrocyte precursor cells and had diverse effects on myelin-related transcription factors, which in turn increased the expression of myelin-related proteins. These results indicate that EPAC proteins are negative regulators of myelination and may be promising targets for the treatment of myelin-related diseases.
Collapse
Affiliation(s)
- Zhen-Zhen Gao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ying-Cong Li
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Chong-Yu Shao
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ying Shen
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Liang Zhou
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Key Laboratory of Brain Science, Guizhou Institution of Higher Education, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
42
|
Jiang K, Yao G, Hu L, Yan Y, Liu J, Shi J, Chang Y, Zhang Y, Liang D, Shen D, Zhang G, Meng S, Piao H. MOB2 suppresses GBM cell migration and invasion via regulation of FAK/Akt and cAMP/PKA signaling. Cell Death Dis 2020; 11:230. [PMID: 32286266 PMCID: PMC7156523 DOI: 10.1038/s41419-020-2381-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 12/20/2022]
Abstract
Mps one binder 2 (MOB2) regulates the NDR kinase family, however, whether and how it is implicated in cancer remain unknown. Here we show that MOB2 functions as a tumor suppressor in glioblastoma (GBM). Analysis of MOB2 expression in glioma patient specimens and bioinformatic analyses of public datasets revealed that MOB2 was downregulated at both mRNA and protein levels in GBM. Ectopic MOB2 expression suppressed, while depletion of MOB2 enhanced, the malignant phenotypes of GBM cells, such as clonogenic growth, anoikis resistance, and formation of focal adhesions, migration, and invasion. Moreover, depletion of MOB2 increased, while overexpression of MOB2 decreased, GBM cell metastasis in a chick chorioallantoic membrane model. Overexpression of MOB2-mediated antitumor effects were further confirmed in mouse xenograft models. Mechanistically, MOB2 negatively regulated the FAK/Akt pathway involving integrin. Notably, MOB2 interacted with and promoted PKA signaling in a cAMP-dependent manner. Furthermore, the cAMP activator Forskolin increased, while the PKA inhibitor H89 decreased, MOB2 expression in GBM cells. Functionally, MOB2 contributed to the cAMP/PKA signaling-regulated inactivation of FAK/Akt pathway and inhibition of GBM cell migration and invasion. Collectively, these findings suggest a role of MOB2 as a tumor suppressor in GBM via regulation of FAK/Akt signaling. Additionally, we uncover MOB2 as a novel regulator in cAMP/PKA signaling. Given that small compounds targeting FAK and cAMP pathway have been tested in clinical trials, we suggest that interference with MOB2 expression and function may support a theoretical and therapeutic basis for applications of these compounds.
Collapse
Affiliation(s)
- Ke Jiang
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China.,Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9 Lvshun Road South, 116044, Dalian, China
| | - Gang Yao
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9 Lvshun Road South, 116044, Dalian, China
| | - Lulu Hu
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9 Lvshun Road South, 116044, Dalian, China
| | - Yumei Yan
- The First Department of Ultrasound, the First Affiliated Hospital to Dalian Medical University, No. 222 Zhongshan Road, 116021, Dalian, China
| | - Jia Liu
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Ji Shi
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Youwei Chang
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Ye Zhang
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Dapeng Liang
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9 Lvshun Road South, 116044, Dalian, China
| | - Dachuan Shen
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, 116004, Dalian, China
| | - Guirong Zhang
- Central laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China.
| | - Songshu Meng
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9 Lvshun Road South, 116044, Dalian, China.
| | - Haozhe Piao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China.
| |
Collapse
|
43
|
Petralia MC, Ciurleo R, Saraceno A, Pennisi M, Basile MS, Fagone P, Bramanti P, Nicoletti F, Cavalli E. Meta-Analysis of Transcriptomic Data of Dorsolateral Prefrontal Cortex and of Peripheral Blood Mononuclear Cells Identifies Altered Pathways in Schizophrenia. Genes (Basel) 2020; 11:genes11040390. [PMID: 32260267 PMCID: PMC7230488 DOI: 10.3390/genes11040390] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/13/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia (SCZ) is a psychiatric disorder characterized by both positive and negative symptoms, including cognitive dysfunction, decline in motivation, delusion and hallucinations. Antipsychotic agents are currently the standard of care treatment for SCZ. However, only about one-third of SCZ patients respond to antipsychotic medications. In the current study, we have performed a meta-analysis of publicly available whole-genome expression datasets on Brodmann area 46 of the brain dorsolateral prefrontal cortex in order to prioritize potential pathways underlying SCZ pathology. Moreover, we have evaluated whether the differentially expressed genes in SCZ belong to specific subsets of cell types. Finally, a cross-tissue comparison at both the gene and functional level was performed by analyzing the transcriptomic pattern of peripheral blood mononuclear cells of SCZ patients. Our study identified a robust disease-specific set of dysfunctional biological pathways characterizing SCZ patients that could in the future be exploited as potential therapeutic targets.
Collapse
Affiliation(s)
| | - Rosella Ciurleo
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (R.C.); (P.B.)
| | - Andrea Saraceno
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
- Correspondence: ; Tel.: +39-095-4781284
| | - Placido Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (R.C.); (P.B.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| | - Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| |
Collapse
|
44
|
Diana T, Olivo PD, Chang YH, Wüster C, Kanitz M, Kahaly GJ. Comparison of a Novel Homogeneous Cyclic Amp Assay and a Luciferase Assay for Measuring Stimulating Thyrotropin-Receptor Autoantibodies. Eur Thyroid J 2020; 9:67-72. [PMID: 32257955 PMCID: PMC7109431 DOI: 10.1159/000504509] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/02/2019] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Stimulating thyrotropin-receptor antibodies (TSAb) cause Graves' disease (GD). We tested a novel homogeneous fluorescent 3',5' cyclic adenine monophosphate (cAMP) assay for the detection of TSAb in a bioassay. METHODS Chinese hamster ovary (CHO) cell lines expressing either a chimeric (MC4) or wild-type (WT) TSH-R were incubated with the adenyl cyclase activator forskolin, a human TSAb monoclonal antibody (M22), and with sera from GD patients. Intracellular cAMP levels were measured using a Bridge-It® cAMP assay, and the results were compared with a luciferase-based bioassay. RESULTS Both cell lines were stimulated with forskolin concentrations (0.006-200 µM) in a dose-dependent manner. The linear range in the MC4 and WT cells was 0.8-25 and 3.1-50 µM, respectively. Levels of cAMP and luciferase in forskolin-treated MC4 and WT cells were positively correlated (r = 0.91 and 0.84, both p < 0.001). The 50% maximum stimulatory concentration of forskolin was more than 16-fold higher for the CHO-WT cells than the CHO-MC4 cells in the cAMP assay and 4-fold higher in the luciferase assay. Incubation of both cell lines with M22 (0.006-50 ng/mL) resulted in a dose-dependent increase in cAMP levels with linear ranges for the MC4 and WT cells of 0.8-12.5 and 0.2-3.125 ng/mL, respectively. Comparison of cAMP and luciferase levels in M22-treated MC4 and WT cells also showed a positive correlation (r = 0.88, p < 0.001 and 0.75, p = 0.002). A positive correlation was also noted when using patient samples (r = 0.96, p < 0.001) that were all TSH-R-Ab binding assay positive. CONCLUSION The novel, rapid, simple-to-perform cAMP assay provides TSAb-mediated stimulatory results comparable to a luciferase-based bioassay.
Collapse
Affiliation(s)
- Tanja Diana
- Molecular Thyroid Research Laboratory, Department of Medicine I, Johannes Gutenberg University (JGU) Medical Center, Mainz, Germany
| | - Paul D. Olivo
- Department of Molecular Microbiology, Washington University Medical School, St. Louis, Missouri, USA
| | | | | | - Michael Kanitz
- Molecular Thyroid Research Laboratory, Department of Medicine I, Johannes Gutenberg University (JGU) Medical Center, Mainz, Germany
| | - George J. Kahaly
- Molecular Thyroid Research Laboratory, Department of Medicine I, Johannes Gutenberg University (JGU) Medical Center, Mainz, Germany
- *Prof. George J. Kahaly, JGU Medical Center, Langenbeckstrasse 1, DE–55131 Mainz (Germany), E-Mail
| |
Collapse
|
45
|
Huff TC, Camarena V, Sant DW, Wilkes Z, Van Booven D, Aron AT, Muir RK, Renslo AR, Chang CJ, Monje PV, Wang G. Oscillatory cAMP signaling rapidly alters H3K4 methylation. Life Sci Alliance 2019; 3:3/1/e201900529. [PMID: 31882444 PMCID: PMC6935296 DOI: 10.26508/lsa.201900529] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 01/02/2023] Open
Abstract
This work explores how GPCR-cAMP signaling dynamically influences histone methylation by altering intracellular labile Fe(II) levels and subsequently modulating histone demethylase activity. Epigenetic variation reflects the impact of a dynamic environment on chromatin. However, it remains elusive how environmental factors influence epigenetic events. Here, we show that G protein–coupled receptors (GPCRs) alter H3K4 methylation via oscillatory intracellular cAMP. Activation of Gs-coupled receptors caused a rapid decrease of H3K4me3 by elevating cAMP, whereas stimulation of Gi-coupled receptors increased H3K4me3 by diminishing cAMP. H3K4me3 gradually recovered towards baseline levels after the removal of GPCR ligands, indicating that H3K4me3 oscillates in tandem with GPCR activation. cAMP increased intracellular labile Fe(II), the cofactor for histone demethylases, through a non-canonical cAMP target—Rap guanine nucleotide exchange factor-2 (RapGEF2), which subsequently enhanced endosome acidification and Fe(II) release from the endosome via vacuolar H+-ATPase assembly. Removing Fe(III) from the media blocked intracellular Fe(II) elevation after stimulation of Gs-coupled receptors. Iron chelators and inhibition of KDM5 demethylases abolished cAMP-mediated H3K4me3 demethylation. Taken together, these results suggest a novel function of cAMP signaling in modulating histone demethylation through labile Fe(II).
Collapse
Affiliation(s)
- Tyler C Huff
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Vladimir Camarena
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David W Sant
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Zachary Wilkes
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Derek Van Booven
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Allegra T Aron
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Ryan K Muir
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Adam R Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Christopher J Chang
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.,Department of Chemistry, University of California, Berkeley, CA, USA.,Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
| | - Paula V Monje
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gaofeng Wang
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA .,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
46
|
Chakrabarti S, Jana M, Roy A, Pahan K. Upregulation of Suppressor of Cytokine Signaling 3 in Microglia by Cinnamic Acid. Curr Alzheimer Res 2019; 15:894-904. [PMID: 29732971 DOI: 10.2174/1567205015666180507104755] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/15/2018] [Accepted: 04/23/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Neuroinflammation plays an important role in the pathogenesis of various neurodegenerative diseases including Alzheimer's disease (AD). Suppressor of cytokine signaling 3 (SOCS3) is an anti-inflammatory molecule that suppresses cytokine signaling and inflammatory gene expression in different cells including microglia. OBJECTIVE The pathways through which SOCS3 could be upregulated are poorly described. Cinnamic acid is a metabolite of cinnamon, a natural compound that is being widely used all over the world as a spice or flavoring agent. Here, we examined if cinnamic acid could upregulate SOCS3 in microglia. METHOD Microglia and astroglia isolated from mouse brain as well as BV-2 microglial cells were treated with cinnamic acid followed by monitoring the level of SOCS3 and different proinflammatory molecules by RT-PCR and real-time PCR. To nail down the mechanism, we also performed ChIP analysis to monitore the recruitment of cAMP response element binding (CREB) to the socs3 gene promoter and carried out siRNA knockdown of CREB. RESULTS Cinnamic acid upregulated the expression of SOCS3 mRNA and protein in mouse BV-2 microglial cells in dose- and time-dependent manner. Accordingly, cinnamic acid also increased the level of SOCS3 and suppressed the expression of inducible nitric oxide synthase and proinflammatory cytokines (TNFα, IL-1β and IL-6) in LPSstimulated BV-2 microglial cells. Similar to BV-2 microglial cells, cinnamic acid also increased the expression of SOCS3 in primary mouse microglia and astrocytes. We have seen that cAMP response element is present in the promoter of socs3 gene, that cinnamic acid induces the activation of CREB, that siRNA knockdown of CREB abrogates cinnamic acid-mediated upregulation of SOCS3, and that cinnamic acid treatment leads to the recruitment of CREB to the socs3 gene. CONCLUSIONS These studies suggest that cinnamic acid upregulates the expression of SOCS3 in glial cells via CREB pathway, which may be of importance in neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Sudipta Chakrabarti
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL-60612, United States
| | - Malabendu Jana
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL-60612, United States
| | - Avik Roy
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL-60612, United States
| | - Kalipada Pahan
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL-60612, United States
| |
Collapse
|
47
|
Regulation of Hedgehog signaling Offers A Novel Perspective for Bone Homeostasis Disorder Treatment. Int J Mol Sci 2019; 20:ijms20163981. [PMID: 31426273 PMCID: PMC6719140 DOI: 10.3390/ijms20163981] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/10/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023] Open
Abstract
The hedgehog (HH) signaling pathway is central to the regulation of bone development and homeostasis. HH signaling is not only involved in osteoblast differentiation from bone marrow mesenchymal stem cells (BM-MSCs), but also acts upstream within osteoblasts via the OPG/RANK/RANKL axis to control the expression of RANKL. HH signaling has been found to up-regulate parathyroid hormone related protein (PTHrP) expression in osteoblasts, which in turn activates its downstream targets nuclear factor of activated T cells (NFAT) and cAMP responsive element binding protein (CREB), and as a result CREB and NFAT cooperatively increase RANKL expression and osteoclastogenesis. Osteoblasts must remain in balance with osteoclasts in order to avoid excessive bone formation or resorption, thereby maintaining bone homeostasis. This review systemically summarizes the mechanisms whereby HH signaling induces osteoblast development and controls RANKL expression through PTHrP in osteoblasts. Proper targeting of HH signaling may offer a therapeutic option for treating bone homeostasis disorders.
Collapse
|
48
|
Giuliodori A, Beffagna G, Marchetto G, Fornetto C, Vanzi F, Toppo S, Facchinello N, Santimaria M, Vettori A, Rizzo S, Della Barbera M, Pilichou K, Argenton F, Thiene G, Tiso N, Basso C. Loss of cardiac Wnt/β-catenin signalling in desmoplakin-deficient AC8 zebrafish models is rescuable by genetic and pharmacological intervention. Cardiovasc Res 2019. [PMID: 29522173 DOI: 10.1093/cvr/cvy057] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aims Arrhythmogenic cardiomyopathy (AC) is an inherited heart disease characterized by life-threatening ventricular arrhythmias and fibro-fatty replacement of the myocardium. More than 60% of AC patients show pathogenic mutations in genes encoding for desmosomal proteins. By focusing our attention on the AC8 form, linked to the junctional protein desmoplakin (DSP), we present here a zebrafish model of DSP deficiency, exploited to identify early changes of cell signalling in the cardiac region. Methods and results To obtain an embryonic model of Dsp deficiency, we first confirmed the orthologous correspondence of zebrafish Dsp genes (dspa and dspb) to the human DSP counterpart. Then, we verified their cardiac expression, at embryonic and adult stages, and subsequently we targeted them by antisense morpholino strategy, confirming specific and disruptive effects on desmosomes, like those identified in AC patients. Finally, we exploited our Dsp-deficient models for an in vivo cell signalling screen, using pathway-specific reporter transgenes. Out of nine considered, three pathways (Wnt/β-catenin, TGFβ/Smad3, and Hippo/YAP-TAZ) were significantly altered, with Wnt as the most dramatically affected. Interestingly, under persistent Dsp deficiency, Wnt signalling is rescuable both by a genetic and a pharmacological approach. Conclusion Our data point to Wnt/β-catenin as the final common pathway underlying different desmosomal AC forms and support the zebrafish as a suitable model for detecting early signalling pathways involved in the pathogenesis of DSP-associated diseases, possibly responsive to pharmacological or genetic rescue.
Collapse
Affiliation(s)
- Alice Giuliodori
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| | - Giorgia Beffagna
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| | - Giulia Marchetto
- European Laboratory for Non-Linear Spectroscopy, via N. Carrara, 1, Sesto Fiorentino (FI) 50019, Italy
| | - Chiara Fornetto
- European Laboratory for Non-Linear Spectroscopy, via N. Carrara, 1, Sesto Fiorentino (FI) 50019, Italy
| | - Francesco Vanzi
- European Laboratory for Non-Linear Spectroscopy, via N. Carrara, 1, Sesto Fiorentino (FI) 50019, Italy.,Department of Biology, University of Florence, via Madonna del Piano, 6, Sesto Fiorentino (FI) 50019, Italy
| | - Stefano Toppo
- Department of Molecular Medicine University of Padova, viale G. Colombo, 3, Padova 35131, Italy; and
| | - Nicola Facchinello
- Department of Biology, University of Padova, via U. Bassi, 58/B, Padova 35131, Italy
| | - Mattia Santimaria
- Department of Biology, University of Padova, via U. Bassi, 58/B, Padova 35131, Italy
| | - Andrea Vettori
- Department of Biology, University of Padova, via U. Bassi, 58/B, Padova 35131, Italy
| | - Stefania Rizzo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| | - Mila Della Barbera
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| | - Kalliopi Pilichou
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| | - Francesco Argenton
- Department of Biology, University of Padova, via U. Bassi, 58/B, Padova 35131, Italy
| | - Gaetano Thiene
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| | - Natascia Tiso
- Department of Biology, University of Padova, via U. Bassi, 58/B, Padova 35131, Italy
| | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| |
Collapse
|
49
|
Akizuki R, Eguchi H, Endo S, Matsunaga T, Ikari A. ZO-2 Suppresses Cell Migration Mediated by a Reduction in Matrix Metalloproteinase 2 in Claudin-18-Expressing Lung Adenocarcinoma A549 Cells. Biol Pharm Bull 2019; 42:247-254. [PMID: 30713254 DOI: 10.1248/bpb.b18-00670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abnormal expression of the tight junctional components claudins (CLDNs) is observed in various malignant tissues. We reported recently that CLDN18 expression is down-regulated in human lung adenocarcinoma tissues. In the present study, we investigated the biological functions of CLDN18 using lung adenocarcinoma A549 cells. Microarray analysis showed that CLDN18 increases zonula occludens (ZO)-2 expression in A549 cells. The ectopic expression of CLDN18 increased nuclear ZO-2 levels, which were inhibited by N-[2-[[3-(4-bromophenyl)-2-propen-1-yl]amino]ethyl]5-isoquinolinesulfonamide (H-89), a nonspecific protein kinase A (PKA) inhibitor, but not by a PKA inhibitor 14-22 amide. In addition, dibutyryl cyclic adenosine monophosphate, an analogue of PKA, did not increase ZO-2 levels. These results suggest that H-89 sensitive factors without PKA are involved in the CLDN18-induced elevation of ZO-2. The cell cycle was affected by neither ZO-2 knockdown in CLDN18-expresssing A549 (CLDN18/A549) cells nor ZO-2 overexpression in A549 cells, suggesting that ZO-2 does not play an important role in the regulation of cell proliferation. The introduction of ZO-2 small interfering RNA (siRNA) into CLDN18/A549 cells increased migration, the expression and activity of matrix metalloproteinase 2 (MMP2), and the reporter activity of an MMP2 promoter construct. Furthermore, H-89 enhanced both mRNA levels and reporter activity of MMP2 in CLDN18/A549 cells. These results suggested that a reduction in CLDN18-dependent ZO-2 expression enhances MMP2 expression in lung adenocarcinoma cells, resulting in the promotion of the cell migration. CLDN18 may be a novel marker for metastasis in lung adenocarcinoma.
Collapse
Affiliation(s)
- Risa Akizuki
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences,Gifu Pharmaceutical University
| | - Hiroaki Eguchi
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences,Gifu Pharmaceutical University
| | - Satoshi Endo
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences,Gifu Pharmaceutical University
| | - Toshiyuki Matsunaga
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences,Gifu Pharmaceutical University
| | - Akira Ikari
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences,Gifu Pharmaceutical University
| |
Collapse
|
50
|
Lim SH, Lee J. Supplementation with psyllium seed husk reduces myocardial damage in a rat model of ischemia/reperfusion. Nutr Res Pract 2019; 13:205-213. [PMID: 31214288 PMCID: PMC6548711 DOI: 10.4162/nrp.2019.13.3.205] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/13/2019] [Accepted: 03/23/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND/OBJECTIVES Myocardial infarction (MI) is caused by extensive myocardial damage attributed to the occlusion of coronary arteries. Our previous study in a rat model of ischemia/reperfusion (I/R) demonstrated that administration of arabinoxylan (AX), comprising arabinose and xylose, protects against myocardial injury. In this study, we undertook to investigate whether psyllium seed husk (PSH), a safe dietary fiber containing a high level of AX (> 50%), also imparts protection against myocardial injury in the same rat model. MATERIALS/METHODS Rats were fed diets supplemented with PSH (1, 10, or 100 mg/kg/d) for 3 d. The rats were then subjected to 30 min ischemia through ligation of the left anterior descending coronary artery, followed by 3 h reperfusion through release of the ligation. The hearts were harvested and cut into four slices. To assess infarct size (IS), an index representing heart damage, the slices were stained with 2,3,5-triphenyltetrazolium chloride (TTC). To elucidate underlying mechanisms, Western blotting was performed for the slices. RESULTS Supplementation with 10 or 100 mg/kg/d of PSH significantly reduces the IS. PSH supplementation (100 mg/kg/d) tends to reduce caspase-3 generation and increase BCL-2/BAX ratio. PSH supplementation also upregulates the expression of nuclear factor erythroid 2-related factor 2 (NRF2), and its target genes including antioxidant enzymes such as glutathione S-transferase mu 2 (GSTM2) and superoxide dismutase 2 (SOD2). PSH supplementation upregulates some sirtuins (NAD+-dependent deacetylases) including SIRT5 (a mitochondrial sirtuin) and SIRT6 and SIRT7 (nuclear sirtuins). Finally, PSH supplementation upregulates the expression of protein kinase A (PKA), and increases phosphorylated cAMP response element-binding protein (CREB) (pCREB), a target protein of PKA. CONCLUSIONS The results from this study indicate that PSH consumption reduces myocardial I/R injury in rats by inhibiting the apoptotic cascades through modulation of gene expression of several genes located upstream of apoptosis. Therefore, we believe that PSH can be developed as a functional food that would be beneficial in the prevention of MI.
Collapse
Affiliation(s)
- Sun Ha Lim
- Department of Biochemistry, School of Medicine, Catholic University of Daegu, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea
| | - Jongwon Lee
- Department of Biochemistry, School of Medicine, Catholic University of Daegu, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea
| |
Collapse
|