1
|
Skelin J, Luk HY, Butorac D, Boon SS, Tomaić V. The effects of HPV oncoproteins on host communication networks: Therapeutic connotations. J Med Virol 2023; 95:e29315. [PMID: 38115222 DOI: 10.1002/jmv.29315] [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: 10/05/2023] [Revised: 11/13/2023] [Accepted: 11/29/2023] [Indexed: 12/21/2023]
Abstract
Human papillomavirus (HPV) infections are a leading cause of viral-induced malignancies worldwide, with a prominent association with cervical and head and neck cancers. The pivotal role of HPV oncoproteins, E5, E6, and E7, in manipulating cellular events, which contribute to viral pathogenesis in various ways, has been extensively documented. This article reviews the influence of HPV oncoproteins on cellular signaling pathways within the host cell, shedding light on the underlying molecular mechanisms. A comprehensive understanding of these molecular alterations is essential for the development of targeted therapies and strategies to combat HPV-induced premalignancies and prevent their progress to cancer. Furthermore, this review underscores the intricate interplay between HPV oncoproteins and some of the most important cellular signaling pathways: Notch, Wnt/β-catenin, MAPK, JAK/STAT, and PI3K AKT/mTOR. The treatment efficacies of the currently available inhibitors on these pathways in an HPV-positive context are also discussed. This review also highlights the importance of continued research to advance our knowledge and enhance therapeutic interventions for HPV-associated diseases.
Collapse
Affiliation(s)
- Josipa Skelin
- Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Ho Yin Luk
- Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong SAR
| | - Dražan Butorac
- Department of Gynecology and Obstetrics, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Siaw Shi Boon
- Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong SAR
| | - Vjekoslav Tomaić
- Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
2
|
Wang L, Trasanidis N, Wu T, Dong G, Hu M, Bauer DE, Pinello L. Dictys: dynamic gene regulatory network dissects developmental continuum with single-cell multiomics. Nat Methods 2023; 20:1368-1378. [PMID: 37537351 DOI: 10.1038/s41592-023-01971-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 07/05/2023] [Indexed: 08/05/2023]
Abstract
Gene regulatory networks (GRNs) are key determinants of cell function and identity and are dynamically rewired during development and disease. Despite decades of advancement, challenges remain in GRN inference, including dynamic rewiring, causal inference, feedback loop modeling and context specificity. To address these challenges, we develop Dictys, a dynamic GRN inference and analysis method that leverages multiomic single-cell assays of chromatin accessibility and gene expression, context-specific transcription factor footprinting, stochastic process network and efficient probabilistic modeling of single-cell RNA-sequencing read counts. Dictys improves GRN reconstruction accuracy and reproducibility and enables the inference and comparative analysis of context-specific and dynamic GRNs across developmental contexts. Dictys' network analyses recover unique insights in human blood and mouse skin development with cell-type-specific and dynamic GRNs. Its dynamic network visualizations enable time-resolved discovery and investigation of developmental driver transcription factors and their regulated targets. Dictys is available as a free, open-source and user-friendly Python package.
Collapse
Affiliation(s)
- Lingfei Wang
- Molecular Pathology Unit and Center for Cancer Research, Massachusetts General Hospital Research Institute, Department of Pathology, Harvard Medical School, Boston, MA, USA
- Gene Regulation Observatory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nikolaos Trasanidis
- Molecular Pathology Unit and Center for Cancer Research, Massachusetts General Hospital Research Institute, Department of Pathology, Harvard Medical School, Boston, MA, USA
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Ting Wu
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Guanlan Dong
- Molecular Pathology Unit and Center for Cancer Research, Massachusetts General Hospital Research Institute, Department of Pathology, Harvard Medical School, Boston, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Bioinformatics and Integrative Genomics PhD Program, Harvard Medical School, Boston, MA, USA
| | - Michael Hu
- Molecular Pathology Unit and Center for Cancer Research, Massachusetts General Hospital Research Institute, Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Daniel E Bauer
- Gene Regulation Observatory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Luca Pinello
- Molecular Pathology Unit and Center for Cancer Research, Massachusetts General Hospital Research Institute, Department of Pathology, Harvard Medical School, Boston, MA, USA.
- Gene Regulation Observatory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
3
|
Tazi N, Semlali A, Loubaki L, Alamri A, Rouabhia M. Cannabis smoke condensate induces human gingival epithelial cell damage through apoptosis, autophagy, and oxidative stress. Arch Oral Biol 2022; 141:105498. [PMID: 35810494 DOI: 10.1016/j.archoralbio.2022.105498] [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: 04/18/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 11/02/2022]
Abstract
OBJECTIVES This study aims to investigate the effects of cannabis smoke condensate (CSC) on the adhesion, growth, and signaling pathways of human gingival epithelial cells. DESIGN The effects of CSC on cell shape and adhesion, and viability were evaluated after 30 min, 60 min, 2 h, and 24 h of exposure using microscopic observation, cell metabolic activity, and lactate dehydrogenase activity assays. The effects of CSC on cell apoptosis, necrosis, autophagy, and oxidative stress were determined through flow cytometry, while apoptotic and autophagic gene expression were identified via an RT2-PCR array. Phosphorylated signaling pathway proteins were measured using flow cytometry. RESULTS CSC deregulated gingival epithelial cell shape and adhesion, decreased cell viability, and increased lactate dehydrogenase release. Its toxic effects included apoptosis, autophagy, and oxidative stress. Moreover, it modulated seven specific apoptotic and six autophagic genes. Furthermore, it decreased phosphorylation in signaling proteins, such as STAT5, ERK12, P38, and nuclear factor κB. CONCLUSIONS CSC has notable adverse effects on gingival epithelial cells. This finding indicates that cannabis smoke could impair gingival epithelial cell innate immune function, leading to gingivitis and periodontitis. Oral health professionals may need to document observed modifications in the oral cavity of patients who smoke cannabis and consider these potential changes during clinical care.
Collapse
Affiliation(s)
- Neftaha Tazi
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, Canada
| | - Abdelhabib Semlali
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, Canada
| | - Lionel Loubaki
- Medical Affairs and Innovation, Héma-Québec, 1070 Avenue des Sciences-de-la-Vie, Québec, Canada
| | - Abdullah Alamri
- Genome Research Chair, Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mahmoud Rouabhia
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, Canada.
| |
Collapse
|
4
|
Baek HS, Kwon TU, Shin S, Kwon YJ, Chun YJ. Steroid sulfatase deficiency causes cellular senescence and abnormal differentiation by inducing Yippee-like 3 expression in human keratinocytes. Sci Rep 2021; 11:20867. [PMID: 34675221 PMCID: PMC8531280 DOI: 10.1038/s41598-021-00051-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 10/05/2021] [Indexed: 12/15/2022] Open
Abstract
Human steroid sulfatase (STS) is an enzyme that catalyzes the hydrolysis of dehydroepiandrosterone sulfate (DHEAS), estrone sulfate (E1S), and cholesterol sulfate. Abnormal expression of STS causes several diseases including colorectal, breast, and prostate cancer and refractory skin disease. In particular, accumulation of intracellular cholesterol sulfate by STS deficiency leads to a skin disorder with abnormal keratinization called X-linked ichthyosis (XLI). To determine the detailed mechanisms of XLI, we performed RNA sequencing (RNA-seq) analysis using human keratinocyte HaCaT cells treated with cholesterol and cholesterol sulfate. Of the genes with expression changes greater than 1.5-fold, Yippee-like 3 (YPEL3), a factor expected to affect cell differentiation, was found. Induction of YPEL3 causes permanent growth arrest, cellular senescence, and inhibition of metastasis in normal and tumor cells. In this study, we demonstrate that YPEL3 expression was induced by STS deficiency and, using the CRISPR/Cas9 system, a partial knock-out (STS+/−) cell line was constructed to establish a disease model for XLI studies. Furthermore, we show that increased expression of YPEL3 in STS-deficient cell lines promoted cellular senescence and expression of keratinization-related proteins such as involucrin and loricrin. Our results suggest that upregulation of YPEL3 expression by STS deficiency may play a crucial role in inducing cellular senescence and abnormal differentiation in human keratinocytes.
Collapse
Affiliation(s)
- Hyoung-Seok Baek
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul, Republic of Korea, 06974
| | - Tae-Uk Kwon
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul, Republic of Korea, 06974
| | - Sangyun Shin
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul, Republic of Korea, 06974
| | - Yeo-Jung Kwon
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul, Republic of Korea, 06974
| | - Young-Jin Chun
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul, Republic of Korea, 06974.
| |
Collapse
|
5
|
Morgan EL, Macdonald A. Manipulation of JAK/STAT Signalling by High-Risk HPVs: Potential Therapeutic Targets for HPV-Associated Malignancies. Viruses 2020; 12:E977. [PMID: 32899142 PMCID: PMC7552066 DOI: 10.3390/v12090977] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/14/2022] Open
Abstract
Human papillomaviruses (HPVs) are small, DNA viruses that cause around 5% of all cancers in humans, including almost all cervical cancer cases and a significant proportion of anogenital and oral cancers. The HPV oncoproteins E5, E6 and E7 manipulate cellular signalling pathways to evade the immune response and promote virus persistence. The Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) pathway has emerged as a key mediator in a wide range of important biological signalling pathways, including cell proliferation, cell survival and the immune response. While STAT1 and STAT2 primarily drive immune signalling initiated by interferons, STAT3 and STAT5 have widely been linked to the survival and proliferative potential of a number of cancers. As such, the inhibition of STAT3 and STAT5 may offer a therapeutic benefit in HPV-associated cancers. In this review, we will discuss how HPV manipulates JAK/STAT signalling to evade the immune system and promote cell proliferation, enabling viral persistence and driving cancer development. We also discuss approaches to inhibit the JAK/STAT pathway and how these could potentially be used in the treatment of HPV-associated disease.
Collapse
Affiliation(s)
- Ethan L. Morgan
- Tumour Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA
| | - Andrew Macdonald
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, West Yorkshire, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, West Yorkshire, UK
| |
Collapse
|
6
|
Al-Eitan LN, Alghamdi MA, Tarkhan AH, Al-Qarqaz FA. Genome-wide identification of methylated CpG sites in nongenital cutaneous warts. BMC Med Genomics 2020; 13:100. [PMID: 32641122 PMCID: PMC7346436 DOI: 10.1186/s12920-020-00745-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 06/19/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Low-risk HPV infection has not been the subject of epigenetic investigation. The present study was carried out in order to investigate the methylation status of CpG sites in non-genital cutaneous warts. METHODS Genomic DNA was extracted from 24 paired epidermal samples of warts and normal skin. DNA samples were bisulfite converted and underwent genome-wide methylation profiling using the Infinium MethylationEPIC BeadChip Kit. RESULTS From a total of 844,234 CpG sites, 56,960 and 43,040 CpG sites were found to be hypo- and hypermethylated, respectively, in non-genital cutaneous warts. The most differentially methylated CpG sites in warts were located within the C10orf26, FAM83H-AS1, ZNF644, LINC00702, GSAP, STAT5A, HDAC4, NCALD, and EXOC4 genes. CONCLUSION Non-genital cutaneous warts exhibit a unique CpG methylation signature.
Collapse
Affiliation(s)
- Laith N Al-Eitan
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid, 22110, Jordan.
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Mansour A Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha, 61421, Saudi Arabia
- Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha, 61421, Saudi Arabia
| | - Amneh H Tarkhan
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Firas A Al-Qarqaz
- Department of Internal Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
- Division of Dermatology, Department of Internal Medicine, King Abdullah University Hospital Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
7
|
Crane JD, MacNeil LG, Lally JS, Ford RJ, Bujak AL, Brar IK, Kemp BE, Raha S, Steinberg GR, Tarnopolsky MA. Exercise-stimulated interleukin-15 is controlled by AMPK and regulates skin metabolism and aging. Aging Cell 2015; 14:625-34. [PMID: 25902870 PMCID: PMC4531076 DOI: 10.1111/acel.12341] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2015] [Indexed: 12/28/2022] Open
Abstract
Aging is commonly associated with a structural deterioration of skin that compromises its barrier function, healing, and susceptibility to disease. Several lines of evidence show that these changes are driven largely by impaired tissue mitochondrial metabolism. While exercise is associated with numerous health benefits, there is no evidence that it affects skin tissue or that endocrine muscle-to-skin signaling occurs. We demonstrate that endurance exercise attenuates age-associated changes to skin in humans and mice and identify exercise-induced IL-15 as a novel regulator of mitochondrial function in aging skin. We show that exercise controls IL-15 expression in part through skeletal muscle AMP-activated protein kinase (AMPK), a central regulator of metabolism, and that the elimination of muscle AMPK causes a deterioration of skin structure. Finally, we establish that daily IL-15 therapy mimics some of the anti-aging effects of exercise on muscle and skin in mice. Thus, we elucidate a mechanism by which exercise confers health benefits to skin and suggest that low-dose IL-15 therapy may prove to be a beneficial strategy to attenuate skin aging.
Collapse
Affiliation(s)
- Justin D Crane
- Department of Kinesiology, McMaster UniversityHamilton, Ontario, Canada
- Department of Pediatrics, McMaster UniversityHamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster UniversityHamilton, Ontario, Canada
| | - Lauren G MacNeil
- Department of Pediatrics, McMaster UniversityHamilton, Ontario, Canada
| | - James S Lally
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster UniversityHamilton, Ontario, Canada
| | - Rebecca J Ford
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster UniversityHamilton, Ontario, Canada
| | - Adam L Bujak
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster UniversityHamilton, Ontario, Canada
| | - Ikdip K Brar
- Department of Pediatrics, McMaster UniversityHamilton, Ontario, Canada
| | - Bruce E Kemp
- Department of Medicine, St. Vincent’s Institute of Medical Research, University of MelbourneFitzroy, Vic., Australia
| | - Sandeep Raha
- Department of Pediatrics, McMaster UniversityHamilton, Ontario, Canada
| | - Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster UniversityHamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster UniversityHamilton, Ontario, Canada
| | | |
Collapse
|
8
|
Danso MO, van Drongelen V, Mulder A, Gooris G, van Smeden J, El Ghalbzouri A, Bouwstra JA. Exploring the potentials of nurture: 2nd and 3rd generation explant human skin equivalents. J Dermatol Sci 2015; 77:102-9. [DOI: 10.1016/j.jdermsci.2014.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 11/21/2014] [Accepted: 12/17/2014] [Indexed: 01/03/2023]
|
9
|
Wang Z, Hou X, Qu B, Wang J, Gao X, Li Q. Pten regulates development and lactation in the mammary glands of dairy cows. PLoS One 2014; 9:e102118. [PMID: 25009983 PMCID: PMC4092105 DOI: 10.1371/journal.pone.0102118] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 06/15/2014] [Indexed: 12/31/2022] Open
Abstract
Pten is a tumor suppressor gene regulating many cellular processes, including growth, adhesion, and apoptosis. In the aim of investigating the role of Pten during mammary gland development and lactation of dairy cows, we analyzed Pten expression levels in the mammary glands of dairy cows by using western blotting, immunohistochemistry, and quantitative polymerase chain reaction (qPCR) assays. Dairy cow mammary epithelial cells (DCMECs) were used to study the function of Pten in vitro. We determined concentrations of β-casein, triglyceride, and lactose in the culture medium following Pten overexpression and siRNA inhibition. To determine whether Pten affected DCMEC viability and proliferation, cells were analyzed by CASY-TT and flow cytometry. Genes involved in lactation-related signaling pathways were detected. Pten expression was also assessed by adding prolactin and glucose to cell cultures. When Pten was overexpressed, proliferation of DCMECs and concentrations for β-casein, triglyceride, and lactose were significantly decreased. Overexpression of Pten down-regulated expression of MAPK, CYCLIN D1, AKT, MTOR, S6K1, STAT5, SREBP1, PPARγ, PRLR, and GLUT1, but up-regulated 4EBP1 in DCMECs. The Pten siRNA inhibition experiments revealed results that opposed those from the gene overexpression experiments. Introduction of prolactin (PRL) increased secretion of β-casein, triglyceride, and lactose, but decreased Pten expression levels. Introduction of glucose also increased β-casein and triglyceride concentrations, but did not significantly alter Pten expression levels. The Pten mRNA and protein expression levels were decreased 0.3- and 0.4-fold in mammary glands of lactating cows producing high quality milk (milk protein >3.0%, milk fat >3.5%), compared with those cows producing low quality milk (milk protein <3.0%, milk fat <3.5%). In conclusion, Pten functions as an inhibitor during mammary gland development and lactation in dairy cows. It can down-regulate DCMECs secretion of β-casein, triglyceride, and lactose, and plays a critical role in lactation related signaling pathways.
Collapse
Affiliation(s)
- Zhuoran Wang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xiaoming Hou
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Bo Qu
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Jie Wang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xuejun Gao
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Qingzhang Li
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, China
- * E-mail:
| |
Collapse
|
10
|
Tanabe T, Kanoh S, Moskowitz WB, Rubin BK. Cardiac asthma: transforming growth factor-β from the failing heart leads to squamous metaplasia in human airway cells and in the murine lung. Chest 2013; 142:1274-1283. [PMID: 22505689 DOI: 10.1378/chest.11-1710] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Cardiac asthma describes symptoms of airflow obstruction due to heart failure. Chronic heart failure is associated with decreased FEV 1 , and FEV 1 improves after heart transplantation. Fibrotic remodeling of the heart and airways is mediated, in part, through transforming growth factor (TGF)- β . Blood TGF- b 1 concentration correlates with ventricular remodeling in cardiac disease, and TGF- β decreases after repair. METHODS We established a coculture of normal human bronchial epithelial (NHBE) cells differentiated at air-liquid interface with submerged basal cardiomyoblasts. Airway cells were immunostained with cytokeratin, actin, and involucrin. TGF- β synthesis was assayed using enzyme-linked immunosorbent assay. Phosphorylation of Smad in NHBE cells was determined by Western blotting.Mice given doxorubicin developed cardiac failure, and their airways were histologically examined. RESULTS Coculture induced involucrin-positive squamous metaplasia of NHBE cells, and this was attenuated by TGF- β antibody. Total TGF- β 1 was increased in coculture conditioned medium( P < .001). After 14 days of exposure to recombinant TGF- β 1 , there was squamous transformation of NHBE cells. One week after removing cardiomyoblasts from culture, squamous metaplasia resolved into normal ciliated epithelia. Smad was phosphorylated in NHBE cells with cardiomyoblasts or with recombinant TGF- β 1 exposure. The airways of mice with heart failure also demonstrated involucrin-positive squamous transformation. CONCLUSIONS TGF- β from cardiomyoblasts or from the failing heart can cause airway squamous metaplasia via Smad signaling, and this is blocked by anti-TGF- b antibody and reversed when cardiac cells are removed from culture. This appears to be an important mechanism for airflow obstruction with heart failure, sometimes described as cardiac asthma.
Collapse
Affiliation(s)
- Tsuyoshi Tanabe
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Soichiro Kanoh
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - William B Moskowitz
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Bruce K Rubin
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA.
| |
Collapse
|
11
|
Sahu RP, DaSilva SC, Rashid B, Martel KC, Jernigan D, Mehta SR, Mohamed DR, Rezania S, Bradish JR, Armstrong AB, Warren S, Konger RL. Mice lacking epidermal PPARγ exhibit a marked augmentation in photocarcinogenesis associated with increased UVB-induced apoptosis, inflammation and barrier dysfunction. Int J Cancer 2012; 131:E1055-66. [PMID: 22467332 DOI: 10.1002/ijc.27562] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 02/16/2012] [Indexed: 12/21/2022]
Abstract
Recent studies suggest that peroxisome proliferator-activated receptor gamma (PPARγ) agonists may have cancer chemopreventive activity. Other studies have shown that loss of epidermal PPARγ results in enhanced chemical carcinogenesis in mice via unknown mechanisms. However, ultraviolet B (UVB) exposure represents the primary etiological agent for skin cancer formation and the role of PPARγ in photobiology and photocarcinogenesis is unknown. In previous studies, we demonstrated that UVB irradiation of cells results in the formation of oxidized glycerophosphocholines that exhibit PPARγ ligand activity. We therefore hypothesized that PPARγ would prove to be a chemopreventive target in photocarcinogenesis. We first showed that UVB irradiation of mouse skin causes generation of PPARγ agonist species in vivo. We then generated SKH-1 hairless, albino mice deficient in epidermal Pparg (Pparg-/-(epi)) using a cytokeratin 14 driven Cre-LoxP strategy. Using a chronic model of UVB photocarcinogenesis, we next showed that Pparg-/-(epi) mice exhibit an earlier onset of tumor formation, increased tumor burden and tumor progression. Increased tumor burden in Pparg-/-(epi) mice was accompanied by a significant increase in epidermal hyperplasia and p53 positive epidermal cells in surrounding skin lacking tumors. After acute UVB irradiation, Pparg-/-(epi) mice exhibited an augmentation of both UVB-induced Caspase 3/7 activity and inflammation. Increased apoptosis and inflammation was also observed after treatment with the PPARγ antagonist GW9662. With chronic UVB irradiation, Pparg-/-(epi) mice exhibited a sustained increase in erythema and transepidermal water loss relative to wildtype littermates. This suggests that PPARγ agonists could have possible chemopreventive activity in non-melanoma skin cancer.
Collapse
Affiliation(s)
- Ravi P Sahu
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
PPARγ mediates innate immunity by regulating the 1α,25-dihydroxyvitamin D3 induced hBD-3 and cathelicidin in human keratinocytes. J Dermatol Sci 2010; 60:179-86. [PMID: 20970965 DOI: 10.1016/j.jdermsci.2010.09.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 09/17/2010] [Accepted: 09/21/2010] [Indexed: 01/17/2023]
Abstract
BACKGROUND Production of antimicrobial peptides (AMPs) is the primary mechanism by which skin innate immunity protects against infection. Hormonally active vitamin D3 (1α,25-dihydroxyvitamin D3; 1,25D₃) is a vital regulator of skin innate immunity, and has been shown to increase the expression and function of AMPs. OBJECTIVE PPARγ is a ligand-activated nuclear receptor and plays a role in keratinocyte differentiation and cutaneous homeostasis. In this study, we investigate whether 1,25D₃-activated PPARγ signaling regulates AMP expression in keratinocytes. METHODS Subconfluent keratinocytes were treated with 1,25D₃ for the indicated times. The mRNA and protein levels of AMPs were detected by RT-PCR and Western blot, and the DNA binding activation of PPARγ, VDRE and AP-1 was investigated by EMSA. To examine the role of PPARγ, the recombinant adenovirus carrying a dominant-negative form of PPARγ (dn-PPARγ) was constructed and transfected into keratinocytes. RESULTS We show here that 1,25D₃ significantly enhances hBD-3 and cathelicidin expression in keratinocytes. Expression of dn-PPARγ did not affect binding to the vitamin D-responsive element (VDRE), which is crucial for cathelicidin induction by VD3; however, it did decrease 1,25D₃ induction of both hBD-3 and cathelicidin. Inhibition of the p38, ERK, and JNK signaling pathways blocked hBD-3 expression, whereas only p38 inhibition suppressed cathelicidin induction. dn-PPARγ had no effect on ERK and JNK activity, but inhibited p38 phosphorylation and suppressed 1,25D₃-induced AP-1 activation via effects on Fra1 and c-Fos proteins. CONCLUSIONS In conclusion, PPARγ regulates the 1,25D₃-induced hBD-3 and cathelicidin expression in keratinocytes through the regulation of AP-1 and p38 activity.
Collapse
|
13
|
Dentelli P, Trombetta A, Togliatto G, Zeoli A, Rosso A, Uberti B, Orso F, Taverna D, Pegoraro L, Brizzi MF. Formation of STAT5/PPARgamma transcriptional complex modulates angiogenic cell bioavailability in diabetes. Arterioscler Thromb Vasc Biol 2008; 29:114-20. [PMID: 18927468 DOI: 10.1161/atvbaha.108.172247] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Circulating angiogenic cells (CACs) expansion is a multistage process requiring sequential activation of transcriptional factors, including STAT5. STAT5, in concert with peroxisome proliferator-activated receptors (PPARs), seems to induce discrete biological responses in different tissues. In the present study we investigated the role of STAT5 and PPARgamma in regulating CAC expansion in normal and diabetic settings. METHODS AND RESULTS Normal and diabetic CACs were used. siRNA technology, EMSA, and chromatin immunoprecipitation (ChIP) assay as well as site-directed mutagenesis of the STAT5 response element in the PPARgamma promoter enabled us to demonstrate that STAT5 transcriptional activity controls PPARgamma expression. Moreover, FACS analysis, coimmunoprecipitation experiments, and ChIP assay revealed that a STAT5/PPARgamma transcriptional complex controls cyclin D1 expression and CAC progression into the cell-cycle. Conversely, PPARgamma agonists, by preventing the expression of STAT5 and the formation of the STAT5/PPARgamma heterodimeric complex failed to promote CAC expansion. Finally, we demonstrated that diabetic CAC functional capability can be recovered by molecules able to activate the STAT5/PPARgamma transcriptional complex. CONCLUSIONS Our data identify the STAT5/PPARgamma heterodimers as landmark of CAC expansion and provide evidences for a mechanism that partially rescues CAC bioavailability in diabetic setting.
Collapse
Affiliation(s)
- Patrizia Dentelli
- Department of Internal Medicine, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Huang CS, Ho WL, Lee WS, Sheu MT, Wang YJ, Tu SH, Chen RJ, Chu JS, Chen LC, Lee CH, Tseng H, Ho YS, Wu CH. SP1-regulated p27/Kip1 gene expression is involved in terbinafine-induced human A431 cancer cell differentiation: An in vitro and in vivo study. Biochem Pharmacol 2008; 75:1783-96. [DOI: 10.1016/j.bcp.2008.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2007] [Revised: 02/01/2008] [Accepted: 02/07/2008] [Indexed: 12/21/2022]
|
15
|
Schmuth M, Jiang YJ, Dubrac S, Elias PM, Feingold KR. Thematic review series: skin lipids. Peroxisome proliferator-activated receptors and liver X receptors in epidermal biology. J Lipid Res 2008; 49:499-509. [PMID: 18182682 DOI: 10.1194/jlr.r800001-jlr200] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The epidermis is a very active site of lipid metabolism, and all peroxisome proliferator-activated receptor (PPAR) and liver X receptor (LXR) isoforms are expressed in the epidermis. Activation of PPARalpha, -beta/delta, or -gamma or LXRs stimulates keratinocyte differentiation. Additionally, activation of these receptors also improves permeability barrier homeostasis by a number of mechanisms, including stimulating epidermal lipid synthesis, increasing lamellar body formation and secretion, and increasing the activity of enzymes required for the extracellular processing of lipids in the stratum corneum, leading to the formation of lamellar membranes that mediate permeability barrier function. The stimulation of keratinocyte differentiation and permeability barrier formation also occurs during fetal development, resulting in accelerated epidermal development. PPAR and LXR activation regulates keratinocyte proliferation and apoptosis, and studies have shown that these receptors play a role in cutaneous carcinogenesis. Lastly, PPAR and LXR activation is anti-inflammatory, reducing inflammation in animal models of allergic and irritant contact dermatitis. Because of their broad profile of beneficial effects on skin homeostasis, PPAR and LXR have great potential to serve as drug targets for common skin diseases such as psoriasis, atopic dermatitis, and skin cancer.
Collapse
Affiliation(s)
- Matthias Schmuth
- Department of Dermatology, University of California, San Francisco, CA, USA.
| | | | | | | | | |
Collapse
|