1
|
Ma Y, Shi J, Liu Y, Cui W, Pan R, Qiu H, Han F, Hou N, Sun X. Identification of Potential Indicators for Survival in Patients with Thyroid Cancer Based on Expression of FAM3 Members. Crit Rev Eukaryot Gene Expr 2023; 33:39-52. [PMID: 37183945 DOI: 10.1615/critreveukaryotgeneexpr.2022044417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Thyroid cancer (THCA) is a common head and neck malignancy. The family with sequence similarity 3 (FAM3) is a cytokine-like gene family with four members, which is presumed to participate in the development of many cancer types. However, the expression patterns of FAM3s in THCA and their prognostic values, have not yet been established. We investigated differential expressions of FAM3 mRNA and protein in THCA, then validated the findings for FAM3B by immunohistochemistry. We also investigated survival data with respect to FAM3 expression patterns in patients with THCA. FAM3s information regarding their relationships with clinical pathological parameters were obtained and FAM3 mutations were assessed. KEGG and GO pathway regarding FAM3C were obtained using online databases. To investigate potential correlations between FAM3s and immune cell infiltration, we investigated the roles of FAM3s in immune cells of patients with THCA. The mRNA expression of FAM3C were significantly elevated in THCA tissues; high expression levels of FAM3C protein were also observed in THCA tissues. A significant association between the pathological stage and the expression of FAM3C was found in patients with THCA. Patients with THCA who had high mRNA expression levels of FAM3C exhibited significantly more favorable prognosis, compared with patients who had low mRNA expression levels of FAM3C. Overall, FAM3C may play vital roles in the pathogenesis and development of THCA, and these findings constitute novel insights for biomarkers of immunotherapeutic targeted agents and may aid in the identification of prognostic biomarkers for THCA.
Collapse
Affiliation(s)
- Yuting Ma
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Junfeng Shi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang Weifang, China
| | - Yongping Liu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Weiming Cui
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ruiyan Pan
- Department of Pharmacy, Weifang Medical University, Weifang, China
| | - Hongyan Qiu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang Weifang, China
| |
Collapse
|
2
|
López-Bermudo L, Luque-Sierra A, Maya-Miles D, Gallego-Durán R, Ampuero J, Romero-Gómez M, Berná G, Martín F. Contribution of Liver and Pancreatic Islet Crosstalk to β-Cell Function/Dysfunction in the Presence of Fatty Liver. Front Endocrinol (Lausanne) 2022; 13:892672. [PMID: 35651973 PMCID: PMC9148952 DOI: 10.3389/fendo.2022.892672] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
Tissue-to-tissue crosstalk regulates organ function, according to growing data. This phenomenon is relevant for pancreatic β-cells and the liver, as both tissues are involved in glucose homeostasis and lipid metabolism. The ability to fine-tune regulation and adaptive responses is enabled through communication between pancreatic β-cells and the liver. However, the crosstalk between both tissues changes when metabolic dysregulation is present. Factors and cargo from extracellular vesicles (EVs) released by liver and pancreatic β-cells that reach the circulation form the words of this interaction. The molecules released by the liver are called hepatokines and are usually secreted in response to the metabolic state. When hepatokines reach the pancreatic islets several mechanisms are initiated for their protection or damage. In the case of the crosstalk between pancreatic β-cells and the liver, only one factor has been found to date. This protein, pancreatic derived factor (PANDER) has been proposed as a novel linker between insulin resistance (IR) and type 2 diabetes mellitus (T2D) and could be considered a biomarker for non-alcoholic fatty liver disease (NAFLD) and T2D. Furthermore, the cargo released by EVs, mainly miRNAs, plays a significant role in this crosstalk. A better knowledge of the crosstalk between liver and pancreatic β-cells is essential to understand both diseases and it could lead to better prevention and new therapeutic options.
Collapse
Affiliation(s)
- Lucía López-Bermudo
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), University Pablo Olavide, University of Seville, CSIC, Seville, Spain
- Biomedical Research Network on Diabetes and Related Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Amparo Luque-Sierra
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), University Pablo Olavide, University of Seville, CSIC, Seville, Spain
| | - Douglas Maya-Miles
- Hospital Universitario Virgen del Rocío de Sevilla, Instituto de Biomedicina de Sevilla, Universidad de Sevilla, Sevilla, Spain
- Biomedical Research Network on Hepatic and Digestive Diseases (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Rocío Gallego-Durán
- Hospital Universitario Virgen del Rocío de Sevilla, Instituto de Biomedicina de Sevilla, Universidad de Sevilla, Sevilla, Spain
- Biomedical Research Network on Hepatic and Digestive Diseases (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Ampuero
- Hospital Universitario Virgen del Rocío de Sevilla, Instituto de Biomedicina de Sevilla, Universidad de Sevilla, Sevilla, Spain
- Biomedical Research Network on Hepatic and Digestive Diseases (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Romero-Gómez
- Hospital Universitario Virgen del Rocío de Sevilla, Instituto de Biomedicina de Sevilla, Universidad de Sevilla, Sevilla, Spain
- Biomedical Research Network on Hepatic and Digestive Diseases (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Genoveva Berná
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), University Pablo Olavide, University of Seville, CSIC, Seville, Spain
- Biomedical Research Network on Diabetes and Related Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Franz Martín, ; Genoveva Berná,
| | - Franz Martín
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), University Pablo Olavide, University of Seville, CSIC, Seville, Spain
- Biomedical Research Network on Diabetes and Related Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Franz Martín, ; Genoveva Berná,
| |
Collapse
|
3
|
The cytokine FAM3B/PANDER is an FGFR ligand that promotes posterior development in Xenopus. Proc Natl Acad Sci U S A 2021; 118:2100342118. [PMID: 33975953 PMCID: PMC8158011 DOI: 10.1073/pnas.2100342118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
How distinct body regions form along the anterior–posterior axis in vertebrate embryos is a fascinating and incompletely understood developmental process. FAM3B/PANDER is a secreted protein involved in glucose metabolism and type 2 diabetes pathogenesis in mammals, but its receptor has been unknown. Here, we report that FAM3B binds to transmembrane fibroblast growth factor receptors (FGFRs) and activates their downstream signaling pathway. In frog embryos, gain-of-function of FAM3B impairs head development and induces ectopic tail-like structures, whereas loss-of-function of FAM3B promotes head development. FGFR is required downstream of FAM3B for head-to-tail patterning. Our results reveal that FAM3B functions by activating the FGFR pathway in frog embryos and mammalian cells and shed light on its possible role in human diseases. Fibroblast growth factor (FGF)/extracellular signal-regulated kinase (ERK) signaling plays a crucial role in anterior–posterior (A–P) axial patterning of vertebrate embryos by promoting posterior development. In our screens for novel developmental regulators in Xenopus embryos, we identified Fam3b as a secreted factor regulated in ectodermal explants. Family with sequence similarity 3 member B (FAM3B)/PANDER (pancreatic-derived factor) is a cytokine involved in glucose metabolism, type 2 diabetes, and cancer in mammals. However, the molecular mechanism of FAM3B action in these processes remains poorly understood, largely because its receptor is still unidentified. Here we uncover an unexpected role of FAM3B acting as a FGF receptor (FGFR) ligand in Xenopus embryos. fam3b messenger RNA (mRNA) is initially expressed maternally and uniformly in the early Xenopus embryo and then in the epidermis at neurula stages. Overexpression of Xenopus fam3b mRNA inhibited cephalic structures and induced ectopic tail-like structures. Recombinant human FAM3B protein was purified readily from transfected tissue culture cells and, when injected into the blastocoele cavity, also caused outgrowth of tail-like structures at the expense of anterior structures, indicating FGF-like activity. Depletion of fam3b by specific antisense morpholino oligonucleotides in Xenopus resulted in macrocephaly in tailbud tadpoles, rescuable by FAM3B protein. Mechanistically, FAM3B protein bound to FGFR and activated the downstream ERK signaling in an FGFR-dependent manner. In Xenopus embryos, FGFR activity was required epistatically downstream of Fam3b to mediate its promotion of posterior cell fates. Our findings define a FAM3B/FGFR/ERK-signaling pathway that is required for axial patterning in Xenopus embryos and may provide molecular insights into FAM3B-associated human diseases.
Collapse
|
4
|
He S, Wang W, Yang Y, Li E, Xu L, Chen L. FAM3B promotes progression of oesophageal carcinoma via regulating the AKT-MDM2-p53 signalling axis and the epithelial-mesenchymal transition. J Cell Mol Med 2019; 23:1375-1385. [PMID: 30565387 PMCID: PMC6349344 DOI: 10.1111/jcmm.14040] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 10/19/2018] [Accepted: 10/30/2018] [Indexed: 02/05/2023] Open
Abstract
FAM3B has been suggested to play important roles in the progression of many cancers, such as gastric, oral, colon and prostate cancer. However, little is known about the role of FAM3B in human esophageal squamous cell carcinoma (ESCC). In the present study, we found that FAM3B expression was higher in ESCC tissues than in adjacent normal tissues. Using quantitative real-time polymerase chain reaction, we found similar results in cell lines. FAM3B expression was significantly related to T/TNM stage. Importantly, Kaplan-Meier analysis revealed that a high expression level of FAM3B predicted a poor outcome for ESCC patients. Overexpression of FAM3B inhibits ESCC cell death, increases oesophageal tumour growth in xenografted nude mice, and promotes ESCC cell migration and invasion. Further studies confirmed that FAM3B regulates the AKT-MDM2-p53 pathway and two core epithelial-to-mesenchymal transition process markers, Snail and E-cadherin. Our results provide new insights into the role of FAM3B in the progression of ESCC and suggest that FAM3B may be a promising molecular target and diagnostic marker for ESCC.
Collapse
Affiliation(s)
- Song‐Lin He
- Department of Thoracic SurgeryWest China Hospital of Sichuan UniversityChengduChina
- North Sichuan Medical CollegeNanchongChina
| | - Wen‐Ping Wang
- Department of Thoracic SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Yu‐Sang Yang
- Department of Thoracic SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - En‐Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouChina
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeShantouChina
| | - Li‐Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouChina
- Institute of Oncologic PathologyShantou University Medical CollegeShantouChina
| | - Long‐Qi Chen
- Department of Thoracic SurgeryWest China Hospital of Sichuan UniversityChengduChina
| |
Collapse
|
5
|
Wang H, Yu F, Zhang Z, Hou Y, Teng W, Shan Z, Lai Y. Effects of circulating member B of the family with sequence similarity 3 on the risk of developing metabolic syndrome and its components: A 5-year prospective study. J Diabetes Investig 2018; 9:782-788. [PMID: 29178453 PMCID: PMC6031514 DOI: 10.1111/jdi.12780] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/06/2017] [Accepted: 11/20/2017] [Indexed: 12/01/2022] Open
Abstract
AIMS/INTRODUCTION Member B of the family with sequence similarity 3 (FAM3B), also known as pancreatic-derived factor, is mainly synthesized and secreted by islet β-cells, and plays a role in abnormal metabolism of glucose and lipids. However, the prospective association of FAM3B with metabolic disorders remains unclear. The present study aimed to reveal the predictive relationship between pancreas-specific cytokine and metabolic syndrome (MetS). MATERIALS AND METHODS A total of 210 adults (88 men and 122 women) without MetS, aged between 40 and 65 years, were recruited and received a comprehensive health examination. Baseline serum FAM3B levels were determined by sandwich enzyme-linked immunosorbent assay. Subsequently, all participants underwent a follow-up examination after 5 years. MetS was identified in accordance with the International Diabetes Federation criteria. RESULTS During follow up, 35.7% participants developed MetS. In comparison with the non-MetS group, participants with MetS had an increased serum FAM3B at baseline (21.85 ng/mL [19.38, 24.17 ng/mL] vs 28.56 ng/mL [25.32, 38.10 ng/mL], P < 0.001). Moreover, serum FAM3B was significantly associated with variations in fasting plasma insulin (r = -0.306, P < 0.001), homeostasis model assessment of β-cell function (r = -0.328, P < 0.001) and homeostasis model assessment of insulin resistance (r = -0.191, P = 0.006). Furthermore, a positive correlation between baseline FAM3B and the incidence of MetS was observed, even after multivariable adjustment (relative risk 1.23 [1.15, 1.31], P < 0.001). Furthermore, the optimal cut-off values of FAM3B was 23.98 ng/mL for predicting MetS based on the Youden Index. CONCLUSIONS Elevated circulating FAM3B might be considered as a predictor of newly-onset MetS and its progression.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of Endocrinology and MetabolismInstitute of EndocrinologyLiaoning Provincial Key Laboratory of Endocrine DiseasesThe First Affiliated Hospital of China Medical UniversityChina Medical UniversityShenyangLiaoningChina
| | - Fadong Yu
- Department of Endocrinology and MetabolismInstitute of EndocrinologyLiaoning Provincial Key Laboratory of Endocrine DiseasesThe First Affiliated Hospital of China Medical UniversityChina Medical UniversityShenyangLiaoningChina
| | - Zhuo Zhang
- Department of Endocrinology and MetabolismInstitute of EndocrinologyLiaoning Provincial Key Laboratory of Endocrine DiseasesThe First Affiliated Hospital of China Medical UniversityChina Medical UniversityShenyangLiaoningChina
| | - Yuanyuan Hou
- Department of Endocrinology and MetabolismInstitute of EndocrinologyLiaoning Provincial Key Laboratory of Endocrine DiseasesThe First Affiliated Hospital of China Medical UniversityChina Medical UniversityShenyangLiaoningChina
| | - Weiping Teng
- Department of Endocrinology and MetabolismInstitute of EndocrinologyLiaoning Provincial Key Laboratory of Endocrine DiseasesThe First Affiliated Hospital of China Medical UniversityChina Medical UniversityShenyangLiaoningChina
| | - Zhongyan Shan
- Department of Endocrinology and MetabolismInstitute of EndocrinologyLiaoning Provincial Key Laboratory of Endocrine DiseasesThe First Affiliated Hospital of China Medical UniversityChina Medical UniversityShenyangLiaoningChina
| | - Yaxin Lai
- Department of Endocrinology and MetabolismInstitute of EndocrinologyLiaoning Provincial Key Laboratory of Endocrine DiseasesThe First Affiliated Hospital of China Medical UniversityChina Medical UniversityShenyangLiaoningChina
| |
Collapse
|
6
|
Zhang X, Yang W, Wang J, Meng Y, Guan Y, Yang J. FAM3 gene family: A promising therapeutical target for NAFLD and type 2 diabetes. Metabolism 2018; 81:71-82. [PMID: 29221790 DOI: 10.1016/j.metabol.2017.12.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/08/2017] [Accepted: 12/01/2017] [Indexed: 12/15/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) and diabetes are severe public health issues worldwide. The Family with sequence similarity 3 (FAM3) gene family consists of four members designated as FAM3A, FAM3B, FAM3C and FAM3D, respectively. Recently, there had been increasing evidence that FAM3A, FAM3B and FAM3C are important regulators of glucose and lipid metabolism. FAM3A expression is reduced in the livers of diabetic rodents and NAFLD patients. Hepatic FAM3A restoration activates ATP-P2 receptor-Akt and AMPK pathways to attenuate steatosis and hyperglycemia in obese diabetic mice. FAM3C expression is also reduced in the liver under diabetic condition. FAM3C is a new hepatokine that activates HSF1-CaM-Akt pathway and represses mTOR-SREBP1-FAS pathway to suppress hepatic gluconeogenesis and lipogenesis. In contrast, hepatic expression of FAM3B, also called PANDER, is increased under obese state. FAM3B promotes hepatic lipogenesis and gluconeogenesis by repressing Akt and AMPK activities, and activating lipogenic pathway. Under obese state, the imbalance among hepatic FAM3A, FAM3B and FAM3C signaling networks plays important roles in the pathogenesis of NAFLD and type 2 diabetes. This review briefly discussed the latest research progress on the roles and mechanisms of FAM3A, FAM3B and FAM3C in the regulation of hepatic glucose and lipid metabolism.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Weili Yang
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Junpei Wang
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Yuhong Meng
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Jichun Yang
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
7
|
FAM3B/PANDER inhibits cell death and increases prostate tumor growth by modulating the expression of Bcl-2 and Bcl-X L cell survival genes. BMC Cancer 2018; 18:90. [PMID: 29357840 PMCID: PMC5778767 DOI: 10.1186/s12885-017-3950-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 12/20/2017] [Indexed: 02/06/2023] Open
Abstract
Background FAM3B/PANDER is a novel cytokine-like protein that induces apoptosis in insulin-secreting beta-cells. Since in silico data revealed that FAM3B can be expressed in prostate tumors, we evaluated the putative role of this cytokine in prostate tumor progression. Methods FAM3B expression was analyzed by quantitative PCR in tumor tissue clinical samples and prostate tumor cell lines. Culture growth and viability of DU145 cell line were evaluated after treatment with either exogenous FAM3B protein obtained from conditioned media (CM) of 293 T cells overexpressing FAM3B or a recombinant FAM3B protein produced in a bacterial host. DU145 cells overexpressing FAM3B protein were produced by lentiviral-mediated transduction of full-length FAM3B cDNA. Cell viability and apoptosis were analyzed in DU145/FAM3B cells after treatment with several cell death inducers, such as TNF-alpha, staurosporine, etoposide, camptothecin, and serum starvation conditions. Anchorage-independent growth in soft agarose assay was used to evaluate in vitro tumorigenicity. In vivo tumorigenicity and invasiveness were evaluated by tumor xenograft growth in nude mice. Results We observed an increase in FAM3B expression in prostate tumor samples when compared to normal tissues. DU145 cell viability and survival increased after exogenous treatment with recombinant FAM3B protein or FAM3B-secreted protein. Overexpression of FAM3B in DU145 cells promoted inhibition of DNA fragmentation and phosphatidylserine externalization in a time and dose-dependent fashion, upon apoptosis triggered by TNF-alpha. These events were accompanied by increased gene expression of anti-apoptotic Bcl-2 and Bcl-XL, decreased expression of pro-apoptotic Bax and diminished caspase-3, −8 and −9 proteolytic activities. Furthermore, inhibition of Bcl-2 anti-apoptotic family proteins with small molecules antagonists decreases protective effects of FAM3B in DU145 cells. When compared to the respective controls, cells overexpressing FAM3B displayed a decreased anchorage- independent growth in vitro and increased tumor growth in xenografted nude mice. The immunohistochemistry analysis of tumor xenografts revealed a similar anti-apoptotic phenotype displayed by FAM3B-overexpressing tumor cells. Conclusions Taken together, by activating pro-survival mechanisms FAM3B overexpression contributes to increased resistance to cell death and tumor growth in nude mice, highlighting a putative role for this cytokine in prostate cancer progression. Electronic supplementary material The online version of this article (10.1186/s12885-017-3950-9) contains supplementary material, which is available to authorized users.
Collapse
|
8
|
Chen Z, Wang J, Yang W, Chen J, Meng Y, Feng B, Chi Y, Geng B, Zhou Y, Cui Q, Yang J. FAM3C activates HSF1 to suppress hepatic gluconeogenesis and attenuate hyperglycemia of type 1 diabetic mice. Oncotarget 2017; 8:106038-106049. [PMID: 29285313 PMCID: PMC5739700 DOI: 10.18632/oncotarget.22524] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/30/2017] [Indexed: 12/17/2022] Open
Abstract
FAM3C, a member of FAM3 gene family, has been shown to improve insulin resistance and hyperglycemia in obese mice. This study further determined whether FAM3C functions as a hepatokine to suppress hepatic gluconeogenesis of type 1 diabetic mice. In STZ-induced type 1 diabetic mouse liver, the FAM3C-HSF1-CaM signaling axis was repressed. Hepatic FAM3C overexpression activated HSF1-CaM-Akt pathway to repress gluconeogenic gene expression and ameliorate hyperglycemia of type 1 diabetic mice. Moreover, hepatic HSF1 overexpression also activated CaM-Akt pathway to repress gluconeogenic gene expression and improve hyperglycemia of type 1 diabetic mice. Hepatic FAM3C and HSF1 overexpression had little effect on serum insulin levels in type 1 diabetic mice. In cultured hepatocytes, conditioned medium of Ad-FAM3C-infected cells induced Akt phosphorylation. Moreover, Akt activation and gluconeogenesis repression induced by FAM3C overexpression were reversed by the treatment with anti-FAM3C antibodies. Treatment with recombinant FAM3C protein induced Akt activation in a HSF1- and CaM-dependent manner in cultured hepatocytes. Furthermore, recombinant FAM3C protein repressed gluconeogenic gene expression and gluconeogenesis by inactivating FOXO1 in a HSF1-dependent manner in cultured hepatocytes. In conclusion, FAM3C is a new hepatokine that suppresses hepatic gluconeogenic gene expression and gluconeogenesis independent of insulin by activating HSF1-CaM-Akt pathway.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Junpei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Weili Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Ji Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Yuhong Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Biaoqi Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Yujing Chi
- Institute of Clinical Molecular Biology & Central Laboratory, Peking University People's Hospital, Beijing 100044, China
| | - Bin Geng
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China
| | - Yong Zhou
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Qinghua Cui
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China.,Department of Biomedical Informatics, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education, Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
9
|
Kral M, Klimek C, Kutay B, Timelthaler G, Lendl T, Neuditschko B, Gerner C, Sibilia M, Csiszar A. Covalent dimerization of interleukin-like epithelial-to-mesenchymal transition (EMT) inducer (ILEI) facilitates EMT, invasion, and late aspects of metastasis. FEBS J 2017; 284:3484-3505. [DOI: 10.1111/febs.14207] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/10/2017] [Accepted: 08/22/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Maria Kral
- Institute of Cancer Research; Department of Medicine I; Medical University of Vienna; Austria
| | - Christoph Klimek
- Institute of Cancer Research; Department of Medicine I; Medical University of Vienna; Austria
| | - Betül Kutay
- Research Institute of Molecular Pathology; Vienna Austria
| | - Gerald Timelthaler
- Institute of Cancer Research; Department of Medicine I; Medical University of Vienna; Austria
| | - Thomas Lendl
- Research Institute of Molecular Pathology; Vienna Austria
| | - Benjamin Neuditschko
- Department of Analytical Chemistry; Faculty of Chemistry; University of Vienna; Austria
| | - Christopher Gerner
- Department of Analytical Chemistry; Faculty of Chemistry; University of Vienna; Austria
| | - Maria Sibilia
- Institute of Cancer Research; Department of Medicine I; Medical University of Vienna; Austria
| | - Agnes Csiszar
- Institute of Cancer Research; Department of Medicine I; Medical University of Vienna; Austria
| |
Collapse
|
10
|
Chen Z, Ding L, Yang W, Wang J, Chen L, Chang Y, Geng B, Cui Q, Guan Y, Yang J. Hepatic Activation of the FAM3C-HSF1-CaM Pathway Attenuates Hyperglycemia of Obese Diabetic Mice. Diabetes 2017; 66:1185-1197. [PMID: 28246289 DOI: 10.2337/db16-0993] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 02/18/2017] [Indexed: 11/13/2022]
Abstract
FAM3C is a member of the family with sequence similarity 3 (FAM3) gene family, and this study determined its role and mechanism in regulation of hepatic glucose/lipid metabolism. In obese diabetic mice, FAM3C expression was reduced in the liver, and hepatic FAM3C restoration improved insulin resistance, hyperglycemia, and fatty liver. FAM3C overexpression increased the expression of heat shock factor 1 (HSF1), calmodulin (CaM), and phosphorylated protein kinase B (Akt) and reduced that of gluconeogenic and lipogenic genes in diabetic mouse livers with the suppression of gluconeogenesis and lipid deposition. In cultured hepatocytes, FAM3C overexpression upregulated HSF1 expression, which elevated CaM protein level by inducing CALM1 transcription to activate Akt in a Ca2+- and insulin-independent manner. Furthermore, FAM3C overexpression promoted nuclear exclusion of FOXO1 and repressed gluconeogenic gene expression and gluconeogenesis in a CaM-dependent manner in hepatocytes. Hepatic HSF1 overexpression activated the CaM-Akt pathway to repress gluconeogenic and lipogenic gene expression and improve hyperglycemia and fatty liver in obese diabetic mice. In conclusion, the FAM3C-HSF1-CaM-Akt pathway plays important roles in regulating glucose and lipid metabolism in hepatocytes independent of insulin and calcium. Restoring hepatic FAM3C expression is beneficial for the management of type 2 diabetes and fatty liver.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Liwei Ding
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Weili Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Junpei Wang
- Department of Biomedical Informatics, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Liming Chen
- Department of Biophysics and Molecular Physiology, Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science & Technology School of Life Science & Technology, Wuhan, China
| | - Yongsheng Chang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Geng
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qinghua Cui
- Department of Biomedical Informatics, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| |
Collapse
|
11
|
Athanason MG, Ratliff WA, Chaput D, MarElia CB, Kuehl MN, Stevens SM, Burkhardt BR. Quantitative proteomic profiling reveals hepatic lipogenesis and liver X receptor activation in the PANDER transgenic model. Mol Cell Endocrinol 2016; 436:41-9. [PMID: 27394190 PMCID: PMC5789791 DOI: 10.1016/j.mce.2016.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/06/2016] [Accepted: 07/05/2016] [Indexed: 12/21/2022]
Abstract
PANcreatic-DERived factor (PANDER) is a member of a superfamily of FAM3 proteins modulating glycemic levels by metabolic regulation of the liver and pancreas. The precise PANDER-induced hepatic signaling mechanism is still being elucidated and has been very complex due to the pleiotropic nature of this novel hormone. Our PANDER transgenic (PANTG) mouse displays a selective hepatic insulin resistant (SHIR) phenotype whereby insulin signaling is blunted yet lipogenesis is increased, a phenomena observed in type 2 diabetes. To examine the complex PANDER-induced mechanism of SHIR, we utilized quantitative mass spectrometry-based proteomic analysis using Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) to reveal the global hepatic proteome differences within the PANTG under the metabolic states of fasting, fed and insulin-stimulated conditions. Proteomic analysis identified lipid metabolism as one of the top cellular functions differentially altered in all metabolic states. Differentially expressed proteins within the PANTG having a lipid metabolic role included ACC, ACLY, CD36, CYP7A1, FASN and SCD1. Central to the differentially expressed proteins involved in lipid metabolism was the predicted activation of the liver X receptor (LXR) pathway. Western analysis validated the increased hepatic expression of LXRα along with LXR-directed targets such as FASN and CYP7A1 within the PANTG liver. Furthermore, recombinant PANDER was capable of inducing LXR promoter activity in-vitro as determined by luciferase reporter assays. Taken together, PANDER strongly impacts hepatic lipid metabolism across metabolic states and may induce a SHIR phenotype via the LXR pathway.
Collapse
Affiliation(s)
- Mark G Athanason
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Whitney A Ratliff
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Dale Chaput
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Catherine B MarElia
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Melanie N Kuehl
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Stanley M Stevens
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Brant R Burkhardt
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA.
| |
Collapse
|
12
|
FAM3A attenuates ER stress-induced mitochondrial dysfunction and apoptosis via CHOP-Wnt pathway. Neurochem Int 2016; 94:82-9. [DOI: 10.1016/j.neuint.2016.02.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 02/18/2016] [Accepted: 02/19/2016] [Indexed: 12/13/2022]
|
13
|
Mo X, Yang C, Wang X, Burkhardt BR, Li Y, Xia H, Cao X. F3MB(PANDER) decreases mice hepatic triglyceride and is associated with decreased DGAT1 expression. PLoS One 2015; 10:e0117156. [PMID: 25679806 PMCID: PMC4334525 DOI: 10.1371/journal.pone.0117156] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/18/2014] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Pancreatic-derived factor (PANDER, also named as FAM3B) is secreted by pancreatic α and β cells. Increasing evidence suggests that it may serve a hormonal function related to glycemic and lipid metabolism. In this study, we investigated the effects of PANDER overexpression on hepatic and adipose triglyceride metabolism in high-fat diet-fed male C57BL/6 mice. METHODS PANDER overexpression was achieved by tail-vein injection of recombinant Ad-PANDER and Ad-GFP injected mice served as a control. The TG metabolism in both groups were compared. RESULTS Adenoviral-mediated overexpression of PANDER did not affect body weight, food consumption, or liver enzymes. The triglyceride (TG) content of both liver and adipose tissue was significantly decreased in Ad-PANDER mice (liver: 6.16±1.89 mg/g vs. control 14.95±2.27 mg/g, P<0.05; adipose: 39.31±1.99 mg/100mg vs. 47.22±2.21 mg/100mg, P<0.05). The free fatty acid (FFA) content of adipose tissue in Ad-PANDER mice was also decreased (1.38±0.18 mg/g vs. 2.77±0.31 mg/g, P<0.01). The investigation of key enzymes of triglyceride hydrolysis and FFA oxidation in liver and adipose tissue showed that p-HSL/HSL was significantly increased and that DGAT1 gene and protein expression were significantly reduced in the liver of PANDER-overexpressing mice. PKA phosphorylation was also significantly increased in the livers of Ad-PANDER mice. No differences in ATGL, CPT1, ACOX1, or DGAT2 expression were observed. CONCLUSION Overexpression of PANDER is associated with observable decreases in TG, increases in PKA phosphorylation, and decreased DGAT1 expression, suggesting a possible interrelationship. The mechanisms by which this occurs remain to be elucidated.
Collapse
Affiliation(s)
- Xiaoqing Mo
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, PR.China
| | - Chijiao Yang
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, PR.China
| | - Xuelan Wang
- Department of Pharmacology, School of Medicine, Sun Yat-sen University, No.74 Zhongshan 2nd Road, Guangzhou 510080, PR.China
| | - Brant R. Burkhardt
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 E. Fowler Avenue, BSF 206, Tampa, FL33620-5550, United States of America
| | - Yangbin Li
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, PR.China
| | - Haipeng Xia
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, PR.China
| | - Xiaopei Cao
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, PR.China
- * E-mail:
| |
Collapse
|
14
|
Wang C, Chi Y, Li J, Miao Y, Li S, Su W, Jia S, Chen Z, Du S, Zhang X, Zhou Y, Wu W, Zhu M, Wang Z, Yang H, Xu G, Wang S, Yang J, Guan Y. FAM3A activates PI3K p110α/Akt signaling to ameliorate hepatic gluconeogenesis and lipogenesis. Hepatology 2014; 59:1779-90. [PMID: 24806753 DOI: 10.1002/hep.26945] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 11/18/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED FAM3A belongs to a novel cytokine-like gene family, and its physiological role remains largely unknown. In our study, we found a marked reduction of FAM3A expression in the livers of db/db and high-fat diet (HFD)-induced diabetic mice. Hepatic overexpression of FAM3A markedly attenuated hyperglycemia, insulin resistance, and fatty liver with increased Akt (pAkt) signaling and repressed gluconeogenesis and lipogenesis in the livers of those mice. In contrast, small interfering RNA (siRNA)-mediated knockdown of hepatic FAM3A resulted in hyperglycemia with reduced pAkt levels and increased gluconeogenesis and lipogenesis in the livers of C57BL/6 mice. In vitro study revealed that FAM3A was mainly localized in the mitochondria, where it increases adenosine triphosphate (ATP) production and secretion in cultured hepatocytes. FAM3A activated Akt through the p110α catalytic subunit of PI3K in an insulin-independent manner. Blockade of P2 ATP receptors or downstream phospholipase C (PLC) and IP3R and removal of medium calcium all significantly reduced FAM3A-induced increase in cytosolic free Ca(2+) levels and attenuated FAM3A-mediated PI3K/Akt activation. Moreover, FAM3A-induced Akt activation was completely abolished by the inhibition of calmodulin (CaM). CONCLUSION FAM3A plays crucial roles in the regulation of glucose and lipid metabolism in the liver, where it activates the PI3K-Akt signaling pathway by way of a Ca(2+) /CaM-dependent mechanism. Up-regulating hepatic FAM3A expression may represent an attractive means for the treatment of insulin resistance, type 2 diabetes, and nonalcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- Chunjiong Wang
- Department of Physiology and Pathophysiology, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Wang C, Chen Z, Li S, Zhang Y, Jia S, Li J, Chi Y, Miao Y, Guan Y, Yang J. Hepatic overexpression of ATP synthase β subunit activates PI3K/Akt pathway to ameliorate hyperglycemia of diabetic mice. Diabetes 2014; 63:947-59. [PMID: 24296716 DOI: 10.2337/db13-1096] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
ATP synthase β subunit (ATPSβ) had been previously shown to play an important role in controlling ATP synthesis in pancreatic β-cells. This study aimed to investigate the role of ATPSβ in regulation of hepatic ATP content and glucose metabolism in diabetic mice. ATPSβ expression and ATP content were both reduced in the livers of type 1 and type 2 diabetic mice. Hepatic overexpression of ATPSβ elevated cellular ATP content and ameliorated hyperglycemia of streptozocin-induced diabetic mice and db/db mice. ATPSβ overexpression increased phosphorylated Akt (pAkt) levels and reduced PEPCK and G6pase expression levels in the livers. Consistently, ATPSβ overexpression repressed hepatic glucose production in db/db mice. In cultured hepatocytes, ATPSβ overexpression increased intracellular and extracellular ATP content, elevated the cytosolic free calcium level, and activated Akt independent of insulin. The ATPSβ-induced increase in cytosolic free calcium and pAkt levels was attenuated by inhibition of P2 receptors. Notably, inhibition of calmodulin (CaM) completely abolished ATPSβ-induced Akt activation in liver cells. Inhibition of P2 receptors or CaM blocked ATPSβ-induced nuclear exclusion of forkhead box O1 in liver cells. In conclusion, a decrease in hepatic ATPSβ expression in the liver, leading to the attenuation of ATP-P2 receptor-CaM-Akt pathway, may play an important role in the progression of diabetes.
Collapse
Affiliation(s)
- Chunjiong Wang
- Department of Physiology and Pathophysiology, Key Laboratory of Molecular Cardiovascular Science, Peking (Beijing) University Health Science Center, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Yang J, Guan Y. Family with sequence similarity 3 gene family and nonalcoholic fatty liver disease. J Gastroenterol Hepatol 2013; 28 Suppl 1:105-11. [PMID: 23855304 DOI: 10.1111/jgh.12033] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/20/2013] [Indexed: 01/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) comprises a disease spectrum ranging from simple steatosis (fatty liver) and nonalcoholic steatohepatitis to fibrosis and cirrhosis. NAFLD has become the leading cause of chronic liver diseases as well as liver-related morbidity and mortality worldwide. NAFLD is also associated with increased risk of cardiovascular diseases, hyperlipidemia, and type 2 diabetes. Insulin resistance in adipose tissues and the liver plays crucial roles in the progression of NAFLD. The family with sequence similarity 3 (FAM3) gene family is a cytokine-like gene family with four members designated FAM3A, FAM3B, FAM3C, and FAM3D, respectively. Increasing evidence suggests that the FAM3 gene family members are involved in the pathogenesis of NAFLD. In particular, FAM3B, also called pancreatic-derived factor, is an important regulator of glucose and lipid metabolism. In obesity status, increased expression and secretion of FAM3B in pancreatic islets and liver may induce lipid accumulation in the liver via the induction of hepatic insulin resistance and lipogenesis. FAM3A and FAM3D may also participate in the regulation of lipid and energy metabolism. In this brief review, we discussed the latest findings regarding the role of FAM3 gene family members, in particular FAM3B, in the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- Jichun Yang
- Department of Physiology and Pathophysiology, Peking University Diabetes Center, Beijing, China
| | | |
Collapse
|
17
|
FAM3A is a target gene of peroxisome proliferator-activated receptor gamma. Biochim Biophys Acta Gen Subj 2013; 1830:4160-70. [PMID: 23562554 DOI: 10.1016/j.bbagen.2013.03.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 03/05/2013] [Accepted: 03/27/2013] [Indexed: 11/23/2022]
Abstract
BACKGROUND To date, the biological function of FAM3A, the first member of FAM3 gene family, remains unknown. We aimed to investigate whether the expression of FAM3A in liver cells is regulated by peroxisome proliferator-activated receptors (PPARs). METHODS AND RESULTS The transcriptional activity of human and mouse FAM3A gene promoters was determined by luciferase reporter assay system. PPARγ agonist rosiglitazone induced FAM3A expression in primary cultured mouse hepatocytes and human HepG2 cells. PPARγ antagonism blocked rosiglitazone-induced FAM3A expression, whereas PPARγ overexpression stimulated FAM3A expression in HepG2 cells. In contrast, PPARα agonist fenofibrate or PPARβ agonist GW0742 failed to affect FAM3A expression in HepG2 cells. The transcriptional activities of human and mouse FAM3A promoters were markedly stimulated by PPARγ activation, but not by PPARα and PPARβ activation. Chromatin immunoprecipitation (ChIP) assay revealed a direct binding of PPARγ to the putative peroxisome proliferator response element (PPRE) located at -1258/-1246 in the human FAM3A promoter. Site-directed mutagenesis of this PPRE-like motif abolished PPARγ's stimulatory effect on the transcriptional activity of human FAM3A promoter. In vivo, oral rosiglitazone treatment upregulated FAM3A expression in the livers of C57BL/6 mice and db/db mice. Moreover, upregulation of FAM3A by PPARγ activation was correlated with increased level of phosphorylated Akt (pAkt) in liver cells. CONCLUSIONS FAM3A as a novel target gene of PPARγ. Upregulation of FAM3A by PPARγ activation is correlated with increased pAkt level in liver cells. GENERAL SIGNIFICANCE Upregulation of FAM3A might contribute to PPARγ's metabolic effects in the liver.
Collapse
|
18
|
Mou H, Li Z, Yao P, Zhuo S, Luan W, Deng B, Qian L, Yang M, Mei H, Le Y. Knockdown of FAM3B triggers cell apoptosis through p53-dependent pathway. Int J Biochem Cell Biol 2013; 45:684-91. [DOI: 10.1016/j.biocel.2012.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 11/28/2012] [Accepted: 12/03/2012] [Indexed: 01/08/2023]
|
19
|
Johansson P, Bernström J, Gorman T, Oster L, Bäckström S, Schweikart F, Xu B, Xue Y, Schiavone LH. FAM3B PANDER and FAM3C ILEI represent a distinct class of signaling molecules with a non-cytokine-like fold. Structure 2013; 21:306-13. [PMID: 23333428 DOI: 10.1016/j.str.2012.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 12/13/2012] [Accepted: 12/14/2012] [Indexed: 01/06/2023]
Abstract
The FAM3 superfamily is predicted to contain classical four-helix bundle cytokines, featuring a typical up-up-down-down fold. Two members of FAM3 have been extensively studied. FAM3B PANDER has been shown to regulate glucose homeostasis and β cell function, whereas the homologous FAM3C ILEI has been shown to be involved in epithelial-mesenchymal transition and cancer. Here, we present a three-dimensional structure of a FAM3 protein, murine PANDER. Contrary to previous suggestions, PANDER exhibits a globular β-β-α fold. The structure is composed of two antiparallel β sheets lined by three short helices packing to form a highly conserved water-filled cavity. The fold shares no relation to the predicted four-helix cytokines but is conserved throughout the FAM3 superfamily. The available biological data and the unexpected new fold indicate that FAM3 PANDER and ILEI could represent a new structural class of signaling molecules, with a different mode of action compared to the traditional four-helix bundle cytokines.
Collapse
Affiliation(s)
- Patrik Johansson
- Structure and Biophysics, Discovery Sciences, AstraZeneca, Mölndal 431-83, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Wang C, Burkhardt BR, Guan Y, Yang J. Role of pancreatic-derived factor in type 2 diabetes: evidence from pancreatic β cells and liver. Nutr Rev 2012; 70:100-6. [PMID: 22300596 DOI: 10.1111/j.1753-4887.2011.00457.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pancreatic-derived factor (PANDER) is a cytokine-like protein that is highly expressed in pancreatic islets. In vitro, PANDER pretreatment or viral-mediated overexpression promotes apoptosis of islet β cells. Under conditions of insulin resistance, chronic hyperglycemia potently activates PANDER expression and stimulates the cosecretion of insulin and PANDER in β cells. PANDER binds to the liver cell membrane and induces insulin resistance, resulting in increased gluconeogenesis. Recently, PANDER was found to be expressed in rodent and human liver, and its expression is increased in the liver of diabetic mice and rats. Hepatic overexpression of PANDER promotes lipogenesis in the liver and induces insulin resistance in C57BL/6 mice, whereas the inactivation of hepatic PANDER markedly reduces steatosis, insulin resistance, and hyperglycemia in db/db mice. PANDER deficiency protects mice from high-fat-diet-induced hyperglycemia by decreasing gluconeogenesis in the liver. In summary, PANDER plays an important role in the progression of type 2 diabetes by negatively regulating islet β-cell function and insulin sensitivity in the liver.
Collapse
Affiliation(s)
- Chunjiong Wang
- Department of Physiology and Pathophysiology, Key Laboratory of Molecular Cardiovascular Science, Peking University Health Science Center, Beijing, China
| | | | | | | |
Collapse
|
21
|
Li J, Chi Y, Wang C, Wu J, Yang H, Zhang D, Zhu Y, Wang N, Yang J, Guan Y. Pancreatic-derived factor promotes lipogenesis in the mouse liver: role of the Forkhead box 1 signaling pathway. Hepatology 2011; 53:1906-16. [PMID: 21412813 DOI: 10.1002/hep.24295] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2010] [Accepted: 03/07/2011] [Indexed: 12/29/2022]
Abstract
UNLABELLED Pancreatic-derived factor (PANDER) is a pancreatic islet-specific cytokine that cosecretes with insulin and is important for β cell function. Here, we show that PANDER is constitutively expressed in hepatocytes, and its expression is significantly increased in steatotic livers of diabetic insulin-resistant db/db mice and mice fed a high-fat diet. Overexpression of PANDER in the livers of C57Bl/6 mice promoted lipogenesis, with increased Forkhead box 1 (FOXO1) expression, whereas small interfering RNA-mediated knockdown of hepatic PANDER significantly attenuated steatosis, with reduced FOXO1 expression in db/db mice. Hepatic PANDER silencing also attenuated insulin resistance and hyperglycemia in db/db mice. In cultured hepatocytes, PANDER overexpression induced lipid deposition, increased FOXO1 expression, and suppressed insulin-stimulated Akt activation and FOXO1 inactivation. Moreover, FOXO1 overexpression increased PANDER expression in cultured hepatocytes and mouse livers. CONCLUSION PANDER promotes lipogenesis and compromises insulin signaling in the liver by increasing FOXO1 activity. PANDER may represent a potential therapeutic target for the treatment of fatty liver and insulin resistance.
Collapse
Affiliation(s)
- Jing Li
- Department of Physiology and Pathophysiology, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Zhang D, Yang H, Kong X, Wang K, Mao X, Yan X, Wang Y, Liu S, Zhang X, Li J, Chen L, Wu J, Wei M, Yang J, Guan Y. Proteomics analysis reveals diabetic kidney as a ketogenic organ in type 2 diabetes. Am J Physiol Endocrinol Metab 2011; 300:E287-95. [PMID: 20959534 DOI: 10.1152/ajpendo.00308.2010] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Diabetic nephropathy (DN) is the leading cause of end-stage renal disease. To date, the molecular mechanisms of DN remain largely unclear. The present study aimed to identify and characterize novel proteins involved in the development of DN by a proteomic approach. Proteomic analysis revealed that 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) synthase 2 (HMGCS2), the key enzyme in ketogenesis, was increased fourfold in the kidneys of type 2 diabetic db/db mice. Consistently, the activity of HMGCS2 in kidneys and 24-h urinary excretion of the ketone body β-hydroxybutyrate (β-HB) were significantly increased in db/db mice. Immunohistochemistry, immunofluorescence, and real-time PCR studies further demonstrated that HMGCS2 was highly expressed in renal glomeruli of db/db mice, with weak expression in the kidneys of control mice. Because filtered ketone bodies are mainly reabsorbed in the proximal tubules, we used RPTC cells, a rat proximal tubule cell line, to examine the effect of the increased level of ketone bodies. Treating cultured RPTC cells with 1 mM β-HB significantly induced transforming growth factor-β1 expression, with a marked increase in collagen I expression. β-HB treatment also resulted in a marked increase in vimentin protein expression and a significant reduction in E-cadherin protein levels, suggesting an enhanced epithelial-to-mesenchymal transition in RPTCs. Collectively, these findings demonstrate that diabetic kidneys exhibit excess ketogenic activity resulting from increased HMGCS2 expression. Enhanced ketone body production in the diabetic kidney may represent a novel mechanism involved in the pathogenesis of DN.
Collapse
Affiliation(s)
- Dongjuan Zhang
- Dept. of Physiology and Pathophysiology, Peking University Health Science Ctr., Haidian District, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Wilson CG, Schupp M, Burkhardt BR, Wu J, Young RA, Wolf BA. Liver-specific overexpression of pancreatic-derived factor (PANDER) induces fasting hyperglycemia in mice. Endocrinology 2010; 151:5174-84. [PMID: 20844005 PMCID: PMC2954722 DOI: 10.1210/en.2010-0379] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The pancreas-derived hormones, insulin and glucagon, are the two main regulators of glucose homeostasis. However, their actions can be modulated by the presence of other circulating factors including cytokines. Pancreatic-derived factor (PANDER) is a novel cytokine-like molecule secreted from the endocrine pancreas, but its biological function is currently unknown. To address this, we employed adenoviral gene delivery to develop a novel murine model of PANDER overexpression, which we used to study PANDER's effect on glucose homeostasis. Although serum metabolites in fed mice were unaffected by PANDER overexpression, fasting glucose, insulin, and corticosterone levels were significantly elevated. Additionally, PANDER-overexpressing mice displayed elevated glucose and insulin levels during a glucose tolerance test, indicating that glucose tolerance was impaired. However, there were no defects in glucose-stimulated insulin secretion or peripheral insulin sensitivity. Elevated transcription of hepatic gluconeogenic genes, PEPCK and G6Pase accompanied the fasting hyperglycemia observed in PANDER-overexpressing animals. Similarly, treatment of primary hepatocytes with PANDER-expressing adenovirus or PANDER-enriched conditioned medium elevated gluconeogenic gene expression and glucose output. PANDER treatment also resulted in higher levels of Ser133-phosphorylated cAMP-response element-binding protein in hepatocytes stimulated with 8-bromo-cAMP and dexamethasone and higher levels of intracellular cAMP upon stimulation with forskolin. In summary, we provide the first report that identifies PANDER as a regulator of hepatic glucose metabolism, where it serves as a novel factor that amplifies hepatic cAMP and cAMP-response element-binding protein signaling to induce gluconeogenic gene expression and glucose output.
Collapse
Affiliation(s)
- Camella G Wilson
- University of Pennsylvania School of Medicine, 803B Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, Pennsylvania 19104-4399, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Robert-Cooperman CE, Carnegie JR, Wilson CG, Yang J, Cook JR, Wu J, Young RA, Wolf BA, Burkhardt BR. Targeted disruption of pancreatic-derived factor (PANDER, FAM3B) impairs pancreatic beta-cell function. Diabetes 2010; 59:2209-18. [PMID: 20566664 PMCID: PMC2927943 DOI: 10.2337/db09-1552] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Accepted: 06/14/2010] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Pancreatic-derived factor (PANDER, FAM3B) is a pancreatic islet-specific cytokine-like protein that is secreted from beta-cells upon glucose stimulation. The biological function of PANDER is unknown, and to address this we generated and characterized a PANDER knockout mouse. RESEARCH DESIGN AND METHODS To generate the PANDER knockout mouse, the PANDER gene was disrupted and its expression was inhibited by homologous recombination via replacement of the first two exons, secretion signal peptide and transcriptional start site, with the neomycin gene. PANDER(-/-) mice were then phenotyped by a number of in vitro and in vivo tests to evaluate potential effects on glucose regulation, insulin sensitivity, and beta-cell morphology and function. RESULTS Glucose tolerance tests demonstrated significantly higher blood glucose levels in PANDER(-/-) versus wild-type male mice. To identify the mechanism of the glucose intolerance, insulin sensitivity and pancreatic beta-cell function were examined. Hyperinsulinemic-euglycemic clamps and insulin tolerance testing showed similar insulin sensitivity for both the PANDER(-/-) and wild-type mice. The in vivo insulin response following intraperitoneal glucose injection surprisingly produced significantly higher insulin levels in the PANDER(-/-) mice, whereas insulin release was blunted with arginine administration. Islet perifusion and calcium imaging studies showed abnormal responses of the PANDER(-/-) islets to glucose stimulation. In contrast, neither islet architecture nor insulin content was impacted by the loss of PANDER. Interestingly, the elevated insulin levels identified in vivo were attributed to decreased hepatic insulin clearance in the PANDER(-/-) islets. Taken together, these results demonstrated decreased pancreatic beta-cell function in the PANDER(-/-) mouse. CONCLUSIONS These results support a potential role of PANDER in the pancreatic beta-cell for regulation or facilitation of insulin secretion.
Collapse
Affiliation(s)
- Claudia E. Robert-Cooperman
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Jason R. Carnegie
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Camella G. Wilson
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Jichun Yang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Peking University Diabetes Center, Beijing, China
| | - Joshua R. Cook
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Jianmei Wu
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Robert A. Young
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Bryan A. Wolf
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Brant R. Burkhardt
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| |
Collapse
|
25
|
Wang C, Guan Y, Yang J. Cytokines in the Progression of Pancreatic β-Cell Dysfunction. Int J Endocrinol 2010; 2010:515136. [PMID: 21113299 PMCID: PMC2989452 DOI: 10.1155/2010/515136] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 08/05/2010] [Accepted: 10/07/2010] [Indexed: 12/29/2022] Open
Abstract
The dysfunction of pancreatic β-cell and the reduction in β-cell mass are the decisive events in the progression of type 2 diabetes. There is increasing evidence that cytokines play important roles in the procedure of β-cell failure. Cytokines, such as IL-1β, IFN-γ, TNF-α, leptin, resistin, adiponectin, and visfatin, have been shown to diversely regulate pancreatic β-cell function. Recently, islet-derived cytokine PANcreatic DERived factor (PANDER or FAM3B) has also been demonstrated to be a regulator of islet β-cell function. The change in cytokine profile in islet and plasma is associated with pancreatic β-cell dysfunction and apoptosis. In this paper, we summarize and discuss the recent studies on the effects of certain important cytokines on pancreatic β-cell function. The imbalance in deleterious and protective cytokines plays pivotal roles in the development and progression of pancreatic β-cell dysfunction under insulin-resistant conditions.
Collapse
Affiliation(s)
- Chunjiong Wang
- Department of Physiology and Pathophysiology, Peking University Diabetes Center, Peking University Health Science Center, Beijing 100191, China
| | - Youfei Guan
- Department of Physiology and Pathophysiology, Peking University Diabetes Center, Peking University Health Science Center, Beijing 100191, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, Peking University Diabetes Center, Peking University Health Science Center, Beijing 100191, China
- *Jichun Yang:
| |
Collapse
|
26
|
PANDER binds to the liver cell membrane and inhibits insulin signaling in HepG2 cells. FEBS Lett 2009; 583:3009-15. [PMID: 19683528 DOI: 10.1016/j.febslet.2009.08.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Revised: 08/03/2009] [Accepted: 08/06/2009] [Indexed: 11/23/2022]
Abstract
PANDER is a cytokine co-secreted with insulin from islet beta-cells. To date, the physiological function of PANDER remains largely unknown. Here we show that PANDER binds to the liver membrane by (125)I-PANDER saturation and competitive binding assays. In HepG2 cells, pre-treatment with PANDER ranging from 4 pM to 4 nM for 8h resulted in a maximal inhibition of insulin-stimulated activation of insulin receptor and insulin receptor substrate 1 by 52% and 63%, respectively. Moreover, PANDER treatment also reduced insulin-stimulated PI3K and pAkt levels by 55% and 48%, respectively. In summary, we have identified the liver as a novel target for PANDER, and PANDER may be involved in the progression of diabetes by regulating hepatic insulin signaling pathways.
Collapse
|
27
|
Huang HL, Wu BY, Yang SB, Shao Y, You WD, Wang WH, Wang MW. Down-regulation of PANDER mRNA in gastric cancer. Shijie Huaren Xiaohua Zazhi 2008; 16:1513-1518. [DOI: 10.11569/wcjd.v16.i14.1513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of PANDER gene in paired gastric cancer tissues and metastatic lymphatic tissues, and to evaluate its relationship with the clinicopathologic characteristics and disease prognosis.
METHODS: Reverse transcription-polymerase chain reaction (RT-PCR) was used to examine the expression of PANDER mRNA in 22 fresh surgical samples of primary gastric cancer tissues and their paired surrounding normal gastric mucosa. A gastric cancer tissue microarray containing 1020 duplicate matched normal mucosa, malignant tissues and metastatic lymphatic tissues from 208 gastric cancer patients was constructed. In situ hybridization analysis was performed on the tissue microarray and the correlation between PANDER mRNA expression and clinicopathologic factors was analyzed.
RESULTS: RT-PCR showed that the expression of PANDER mRNA decreased in 72.7% (16/22) of gastric cancer samples. Similar results were obtained by in situ hybridization analysis (60.9%, 112/184). The expression of PANDER mRNA showed an ordinally decreasing trend in the matched normal mucosa, tumor tissues and metastatic lymphatic tissues (χ2 = 81.135, P = 0.00). There was no difference between early gastric cancer and progressive gastric cancer (χ2 = 5.362, P = 0.147). In tumor tissues, lower expression of PANDER mRNA was associated with deeper invasion (Z = -2.52, P = 0.012). There was no correlation between PANDER mRNA expression and disease prognosis.
CONCLUSION: Down-regulation of PANDER gene expression occurs at the early stage of gastric cancer, which may be involved in the genesis and development of gastric cancer.
Collapse
|
28
|
Narayan SB, Rakheja D, Pastor JV, Rosenblatt K, Greene SR, Yang J, Wolf BA, Bennett MJ. Over-expression of CLN3P, the Batten disease protein, inhibits PANDER-induced apoptosis in neuroblastoma cells: further evidence that CLN3P has anti-apoptotic properties. Mol Genet Metab 2006; 88:178-83. [PMID: 16515873 DOI: 10.1016/j.ymgme.2006.01.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 01/18/2006] [Accepted: 01/19/2006] [Indexed: 11/17/2022]
Abstract
Juvenile neuronal ceroid-lipofuscinosis (JNCL) or Batten/Spielmeyer-Vogt-Sjogren disease (OMIM #204200) is one of a group of nine clinically related inherited neurodegenerative disorders (CLN1-9). JNCL results from mutations in CLN3 on chromosome 16p12.1. The neuronal loss in Batten disease has been shown to be due to a combination of apoptosis and autophagy suggesting that CLN3P, the defective protein, may have an anti-neuronal death function. PANDER (PANcreatic-DERived factor) is a novel cytokine that was recently cloned from pancreatic islet cells. PANDER is specifically expressed in the pancreatic islets, small intestine, testis, prostate, and neurons of the central nervous system, and has been demonstrated to induce apoptosis. In this study, we over-expressed CLN3P in SH-SY5Y neuroblastoma cells and monitored the effects on PANDER-induced apoptosis. CLN3P significantly increased the survival rate of the SH-SY5Y cells in this system. This study provides additional evidence that the function of CLN3P is related to preventing neuronal apoptosis.
Collapse
Affiliation(s)
- Srinivas B Narayan
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Yang J, Robert CE, Burkhardt BR, Young RA, Wu J, Gao Z, Wolf BA. Mechanisms of glucose-induced secretion of pancreatic-derived factor (PANDER or FAM3B) in pancreatic beta-cells. Diabetes 2005; 54:3217-28. [PMID: 16249448 DOI: 10.2337/diabetes.54.11.3217] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Pancreatic-derived factor (PANDER) is an islet-specific cytokine present in both pancreatic alpha- and beta-cells, which, in vitro, induces beta-cell apoptosis of primary islet and cell lines. In this study, we investigated whether PANDER is secreted by pancreatic alpha- and beta-cells and whether PANDER secretion is regulated by glucose and other insulin secretagogues. In mouse-derived insulin-secreting beta-TC3 cells, PANDER secretion in the presence of stimulatory concentrations of glucose was 2.8 +/- 0.4-fold higher (P < 0.05) than without glucose. Insulin secretion was similarly increased by glucose in the same cells. The total concentration of secreted PANDER in the medium was approximately 6-10 ng/ml (0.3-0.5 nmol/l) after a 24-h culture with glucose. L-Glucose failed to stimulate PANDER secretion in beta-TC3 cells. KCl stimulated PANDER secretion 2.1 +/- 0.1-fold compared with control without glucose. An L-type Ca2+ channel inhibitor, nifedipine, completely blocked both glucose- or KCl-induced insulin and PANDER secretion. In rat-derived INS-1 cells, glucose (20 mmol/l) stimulated PANDER secretion 4.4 +/- 0.9-fold, while leucine plus glutamine stimulated 4.4 +/- 0.7-fold compared with control without glucose. In mouse islets overexpressing PANDER, glucose (20 mmol/l) stimulated PANDER secretion 3.2 +/- 0.5-fold (P < 0.05) compared with basal (3 mmol/l glucose). PANDER was also secreted by alpha-TC3 cells but was not stimulated by glucose. Mutations of cysteine 229 or of cysteines 91 and 229 to serine, which may form one disulfide bond, and truncation of the COOH-terminus or NH2-terminus of PANDER all resulted in failure of PANDER secretion, even though these mutant or truncated PANDERs were highly expressed within the cells. In conclusion, we found that 1) PANDER is secreted from both pancreatic alpha- and beta-cells, 2) glucose stimulates PANDER secretion dose dependently in beta-cell lines and primary islets but not in alpha-cells, 3) PANDER is likely cosecreted with insulin via the same regulatory mechanisms, and 4) structure and conformation is vital for PANDER secretion.
Collapse
Affiliation(s)
- Jichun Yang
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104-4399, USA
| | | | | | | | | | | | | |
Collapse
|