1
|
Gan D, Wang X, Wu X, Han S. Higher dietary intake of live microbes is inversely associated with accelerated biological aging. Food Funct 2025; 16:1809-1819. [PMID: 39927928 DOI: 10.1039/d4fo04230a] [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: 02/11/2025]
Abstract
Objective: It remains unclear whether dietary live microbe intake is associated with biological aging. Therefore, the present study aimed to investigate the associations between dietary live microbe intake and biological aging. Methods: Our study included 7719 participants aged 20 years and older from the 2007-2010 cycles of the National Health and Nutrition Examination Survey (NHANES). Participants were categorized into groups using two distinct methods. The first method grouped participants based on the live microbial level of the consumed foods, dividing them into three dietary live microbe intake groups: low, medium, and high. The second method classified participants according to the quantity of live microbe-containing foods they consumed (referred to as MedHi), with three groups: G1 (no MedHi intake), G2 (MedHi intake below the median), and G3 (MedHi intake above the median). Biological age was evaluated using the Klemera-Doubal method biological age (KDM-BA) and phenotypic age (PA). KDM-BA acceleration and PA acceleration was determined if participants' KDM-BA or PA exceeded their chronological age, respectively. Multivariable logistic regression models were conducted to explore the associations of dietary live microbe intake groups and dietary MedHi intake groups with the acceleration of KDM-BA and PA. Results: Compared with participants in the low dietary live microbe intake group, those in the high dietary live microbe intake group had a 20% (95% CI: 2% to 35%) reduced risk of KDM-BA acceleration and a 25% (95% CI: 4% to 42%) reduced risk of PA acceleration. Similarly, participants in the G3 group with higher dietary MedHi intake had a 24% (95% CI: 10% to 35%) lower risk of KDM-BA acceleration and a 29% (95% CI: 17% to 39%) lower risk of PA acceleration compared with those in the G1 group. The stratified analyses showed that the associations of dietary MedHi intake with accelerated KDM-BA and PA were not influenced by age, gender, race, smoking, alcohol consumption, physical activity, race, and history of hypertension, diabetes, and cardiovascular disease. Conclusion: Higher dietary intake of live microbes was inversely associated with accelerated biological aging. A diet rich in live microbes may help slow down the aging process.
Collapse
Affiliation(s)
- Da Gan
- Jiangxi Medicine Academy of Nutrition and Health Management, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Xiaoyan Wang
- Department of Nutrition and Food Hygiene, School of Public Health, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Xuebiao Wu
- Center for Molecular Pathology, Department of Pathophysiology, Gannan Medical University, Ganzhou 341000, China
| | - Shuang Han
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, Zhejiang, China.
| |
Collapse
|
2
|
Odiase P, Ma J, Ranganathan S, Ogunkua O, Turner WB, Marshall D, Ochieng J. The Role of Fetuin-A in Tumor Cell Growth, Prognosis, and Dissemination. Int J Mol Sci 2024; 25:12918. [PMID: 39684629 DOI: 10.3390/ijms252312918] [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: 10/26/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Fetuin-A, also known as alpha-2-Heremans-Schmid-glycoprotein (Ahsg), is a multifunctional molecule with diverse roles in biological processes such as mineralization, tumor growth, and inflammation. This review explores the involvement of Ahsg in various cancers, including liver, breast, prostate, colorectal, brain, osteosarcoma, and lung cancers. In many cancer types, Ahsg promotes tumor growth, invasion, and metastasis through various mechanisms, including cellular adhesion, spreading, chemotaxis, and modulation of cell-growth signaling pathways. Additionally, Ahsg has been implicated in the regulation of inflammatory cytokine production, making it a potential marker of inflammation in cancer. The complex interplay between Ahsg and cancer progression highlights its potential as a diagnostic biomarker and therapeutic target in various cancers. However, further research is needed to fully elucidate the mechanisms of action of Ahsg in cancer and to explore its clinical implications in cancer diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Peace Odiase
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Jonathan Ma
- College of Arts and Science, Vanderbilt University, Nashville, TN 37203, USA
| | | | - Olugbemiga Ogunkua
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Winston B Turner
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Dana Marshall
- Department of Pathology, Meharry Medical College, Nashville, TN 37208, USA
| | - Josiah Ochieng
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
- Department of Biomedical Science, School of Graduate Studies Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
3
|
Feng K, Ye T, Xie X, Liu J, Gong L, Chen Z, Zhang J, Li H, Li Q, Wang Y. ESC-sEVs alleviate non-early-stage osteoarthritis progression by rejuvenating senescent chondrocytes via FOXO1A-autophagy axis but not inducing apoptosis. Pharmacol Res 2024; 209:107474. [PMID: 39433168 DOI: 10.1016/j.phrs.2024.107474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
Osteoarthritis (OA) is a common joint degenerative disease which currently lacks satisfactory disease-modifying treatments. Oxidative stress-mediated senescent chondrocytes accumulation is closely associated with OA progression, which abrogates cartilage metabolism homeostasis by secreting senescence-associated secretory phenotype (SASP) factors. Numerous studies suggested mesenchymal stem cells-derived small extracellular vesicles (MSC-sEVs) have been regarded as promising candidates for OA therapy. However, MSC-sEVs were applied before the occurrence of cartilage degeneration or at early-stage OA, while in clinical practice, most OA patients who present with pain are already in non-early-stage. Recently, embryonic stem cells-derived sEVs (ESC-sEVs) have been reported to possess powerful anti-aging effects. However, whether ESC-sEVs could attenuate non-early-stage OA progression remains unknown. In this study, we demonstrated ESC-sEVs ameliorated senescent phenotype and cartilage destruction in both mechanical stress-induced non-early-stage posttraumatic OA and naturally aged mice. More importantly, we found ESC-sEVs alleviated senescent phenotype by rejuvenating aged chondrocytes but not inducing apoptosis. We also provided evidence that the FOXO1A-autophagy axis played an important role in the anti-aging effects of ESC-sEVs. To promote clinical translation, we confirmed ESC-sEVs reversed senescent phenotype in ex-vivo cultured human end-stage OA cartilage explants. Collectively, our findings reveal that ESC-sEVs-based therapy is of high translational value in non-early-stage OA treatment.
Collapse
Affiliation(s)
- Kai Feng
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Teng Ye
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xuetao Xie
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jiashuo Liu
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Liangzhi Gong
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zhengsheng Chen
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Juntao Zhang
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Haiyan Li
- Chemical and Environmental Engineering, School of Engineering, STEM College, RMIT University, 124 La Trobe St, Melbourne, VIC 3000, Australia
| | - Qing Li
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Yang Wang
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
4
|
Li X, Wang J, Zhang M, Li X, Fan Y, Zhou X, Sun Y, Qiu Z. Biological aging mediates the associations of metabolic score for insulin resistance with all-cause and cardiovascular disease mortality among US adults: A nationwide cohort study. Diabetes Obes Metab 2024; 26:3552-3564. [PMID: 38853301 DOI: 10.1111/dom.15694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/11/2024] [Accepted: 05/20/2024] [Indexed: 06/11/2024]
Abstract
AIM To investigate the associations of metabolic score for insulin resistance (METS-IR) with all-cause and cardiovascular disease (CVD)-specific mortality and the potential mediating role of biological ageing. METHODS A cohort of 19 204 participants from the National Health and Nutrition Examination Survey (NHANES) 1999-2018 was recruited for this study. Cox regression models, restricted cubic splines, and Kaplan-Meier survival curves were used to determine the relationships of METS-IR with all-cause and CVD-specific mortality. Mediation analyses were performed to explore the possible intermediary role of biological ageing markers, including phenotypic age (PhenoAge) and biological age (BioAge). RESULTS During a median follow-up of 9.17 years, we observed 2818 deaths, of which 875 were CVD-specific. Multivariable Cox regression showed that the highest METS-IR level (Q4) was associated with increased all-cause (hazard ratio [HR] 1.38, 95% confidence interval [CI] 1.14-1.67) and CVD mortality (HR 1.52, 95% CI 1.10-2.12) compared with the Q1 level. Restricted cubic splines showed a nonlinear relationship between METS-IR and all-cause mortality. Only METS-IR above the threshold (41.02 μg/L) was positively correlated with all-cause death. METS-IR had a linear positive relationship with CVD mortality. In mediation analyses, we found that PhenoAge mediated 51.32% (p < 0.001) and 41.77% (p < 0.001) of the association between METS-IR and all-cause and CVD-specific mortality, respectively. For BioAge, the mediating proportions of PhenoAge were 21.33% (p < 0.001) and 15.88% (p < 0.001), respectively. CONCLUSIONS This study highlights the detrimental effects of insulin resistance, as measured by METS-IR, on all-cause and CVD mortality. Moreover, it underscores the role of biological ageing in mediating these associations, emphasizing the need for interventions targeting both insulin resistance and ageing processes to mitigate mortality risks in metabolic disorders.
Collapse
Affiliation(s)
- Xiaoxuan Li
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jia Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mengqi Zhang
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangjun Li
- Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuchen Fan
- Department of Medicine, Qingdao University, Qingdao, China
| | - Xinbei Zhou
- Department of Critical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuxin Sun
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenkang Qiu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
5
|
Zhang C, Gordon MD, Joseph KM, Diaz‐Hernandez ME, Drissi H, Illien‐Jünger S. Differential efficacy of two small molecule PHLPP inhibitors to promote nucleus Pulposus cell health. JOR Spine 2024; 7:e1306. [PMID: 38222816 PMCID: PMC10782076 DOI: 10.1002/jsp2.1306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/31/2023] [Accepted: 11/04/2023] [Indexed: 01/16/2024] Open
Abstract
Background Intervertebral disc (IVD) degeneration is associated with chronic back pain. We previously demonstrated that the phosphatase pleckstrin homology domain and leucine-rich repeat protein phosphatase (PHLPP) 1 was positively correlated with IVD degeneration and its deficiency decelerated IVD degeneration in both mouse IVDs and human nucleus pulposus (NP) cells. Small molecule PHLPP inhibitors may offer a translatable method to alleviate IVD degeneration. In this study, we tested the effectiveness of the two PHLPP inhibitors NSC117079 and NSC45586 in promoting a healthy NP phenotype. Methods Tail IVDs of 5-month-old wildtype mice were collected and treated with NSC117079 or NSC45586 under low serum conditions ex vivo. Hematoxylin & eosin staining was performed to examine IVD structure and NP cell morphology. The expression of KRT19 was analyzed through immunohistochemistry. Cell apoptosis was assessed by TUNEL assay. Human NP cells were obtained from patients with IVD degeneration. The gene expression of KRT19, ACAN, SOX9, and MMP13 was analyzed via real time qPCR, and AKT phosphorylation and the protein expression of FOXO1 was analyzed via immunoblot. Results In a mouse IVD organ culture model, NSC45586, but not NSC117079, preserved vacuolated notochordal cell morphology and KRT19 expression while suppressing cell apoptosis, counteracting the degenerative changes induced by serum deprivation, especially in males. Likewise, in degenerated human NP cells, NSC45586 increased cell viability and the expression of KRT19, ACAN, and SOX9 and reducing the expression of MMP13, while NSC117079 treatment only increased KRT19 expression. Mechanistically, NSC45586 treatment increased FOXO1 protein expression in NP cells, and inhibiting FOXO1 offset NSC45586-induced regenerative potential, especially in males. Conclusions Our study indicates that NSC45586 was effective in promoting NP cell health, especially in males, suggesting that PHLPP plays a key role in NP cell homeostasis and that NSC45586 might be a potential drug candidate in treating IVD degeneration.
Collapse
Affiliation(s)
- Changli Zhang
- Department of OrthopaedicsEmory University School of MedicineAtlantaGeorgiaUSA
| | - Madeleine D. Gordon
- Department of OrthopaedicsEmory University School of MedicineAtlantaGeorgiaUSA
| | - Katherine M. Joseph
- Department of OrthopaedicsEmory University School of MedicineAtlantaGeorgiaUSA
| | | | - Hicham Drissi
- Department of OrthopaedicsEmory University School of MedicineAtlantaGeorgiaUSA
- Atlanta VA Health Care SystemDecaturGeorgiaUSA
| | - Svenja Illien‐Jünger
- Department of OrthopaedicsEmory University School of MedicineAtlantaGeorgiaUSA
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| |
Collapse
|
6
|
Lim YJ, Kim HS, Bae S, So KA, Kim TJ, Lee JH. Pan-EGFR Inhibitor Dacomitinib Resensitizes Paclitaxel and Induces Apoptosis via Elevating Intracellular ROS Levels in Ovarian Cancer SKOV3-TR Cells. Molecules 2024; 29:274. [PMID: 38202856 PMCID: PMC10780346 DOI: 10.3390/molecules29010274] [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: 10/27/2023] [Revised: 12/19/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024] Open
Abstract
Paclitaxel is still used as a standard first-line treatment for ovarian cancer. Although paclitaxel is effective for many types of cancer, the emergence of chemoresistant cells represents a major challenge in chemotherapy. Our study aimed to analyze the cellular mechanism of dacomitinib, a pan-epidermal growth factor receptor (EGFR) inhibitor, which resensitized paclitaxel and induced cell cytotoxicity in paclitaxel-resistant ovarian cancer SKOV3-TR cells. We investigated the significant reduction in cell viability cotreated with dacomitinib and paclitaxel by WST-1 assay and flow cytometry analysis. Dacomitinib inhibited EGFR family proteins, including EGFR and HER2, as well as its downstream signaling proteins, including AKT, STAT3, ERK, and p38. In addition, dacomitinib inhibited the phosphorylation of Bad, and combination treatment with paclitaxel effectively suppressed the expression of Mcl-1. A 2'-7'-dichlorodihydrofluorescein diacetate (DCFH-DA) assay revealed a substantial elevation in cellular reactive oxygen species (ROS) levels in SKOV3-TR cells cotreated with dacomitinib and paclitaxel, which subsequently mediated cell cytotoxicity. Additionally, we confirmed that dacomitinib inhibits chemoresistance in paclitaxel-resistant ovarian cancer HeyA8-MDR cells. Collectively, our research indicated that dacomitinib effectively resensitized paclitaxel in SKOV3-TR cells by inhibiting EGFR signaling and elevating intracellular ROS levels.
Collapse
Affiliation(s)
- Ye Jin Lim
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea; (Y.J.L.); (H.S.K.); (S.B.)
| | - Hee Su Kim
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea; (Y.J.L.); (H.S.K.); (S.B.)
| | - Seunghee Bae
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea; (Y.J.L.); (H.S.K.); (S.B.)
| | - Kyeong A So
- Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul 05030, Republic of Korea; (K.A.S.); (T.J.K.)
| | - Tae Jin Kim
- Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul 05030, Republic of Korea; (K.A.S.); (T.J.K.)
| | - Jae Ho Lee
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea; (Y.J.L.); (H.S.K.); (S.B.)
| |
Collapse
|
7
|
Shimokawa I. Mechanisms underlying retardation of aging by dietary energy restriction. Pathol Int 2023; 73:579-592. [PMID: 37975408 PMCID: PMC11551835 DOI: 10.1111/pin.13387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
Moderate restriction of dietary energy intake, referred to here as dietary restriction (DR), delays aging and extends lifespan in experimental animals compared with a diet of ad libitum feeding (AL) control animals. Basic knowledge of the mechanisms underlying the effects of DR could be applicable to extending the healthspan in humans. This review highlights the importance of forkhead box O (FoxO) transcription factors downstream of the growth hormone-insulin-like growth factor 1 signaling in the effects of DR. Our lifespan studies in mice with heterozygous Foxo1 or Foxo3 gene knockout indicated differential roles of FoxO1 and FoxO3 in the tumor-inhibiting and life-extending effects of DR. Subsequent studies suggested a critical role of FoxO3 in metabolic and mitochondrial bioenergetic adaptation to DR. Our studies also verified hypothalamic neuropeptide Y (Npy) as a vital neuropeptide showing pleiotropic and sexually dimorphic effects for extending the healthspan in the context of nutritional availability. Npy was necessary for DR to exert its effects in male and female mice; meanwhile, under AL conditions, the loss of Npy prevented obesity and insulin resistance only in female mice. Overnutrition disrupts FoxO- and Npy-associated metabolic and mitochondrial bioenergetic adaptive processes, causing the acceleration of aging and related diseases.
Collapse
Affiliation(s)
- Isao Shimokawa
- Department of Pathology INagasaki University School of Medicine and Graduate School of Biomedical SciencesNagasakiJapan
- SAGL, LLCFukuokaJapan
| |
Collapse
|
8
|
Pintor S, Lopez A, Flores D, Lozoya B, Soti B, Pokhrel R, Negrete J, Persans MW, Gilkerson R, Gunn B, Keniry M. FOXO1 promotes the expression of canonical WNT target genes in examined basal-like breast and glioblastoma multiforme cancer cells. FEBS Open Bio 2023; 13:2108-2123. [PMID: 37584250 PMCID: PMC10626282 DOI: 10.1002/2211-5463.13696] [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: 12/09/2021] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/17/2023] Open
Abstract
Basal-like breast cancer (BBC) and glioblastoma multiforme (GBM) are aggressive cancers associated with poor prognosis. BBC and GBM have stem cell-like gene expression signatures, which are in part driven by forkhead box O (FOXO) transcription factors. To gain further insight into the impact of FOXO1 in BBC, we treated BT549 cells with AS1842856 and performed RNA sequencing. AS1842856 binds to unphosphorylated FOXO1 and inhibits its ability to directly bind to DNA. Gene Set Enrichment Analysis indicated that a set of WNT pathway target genes, including lymphoid enhancer-binding factor 1 (LEF1) and transcription factor 7 (TCF7), were robustly induced after AS1842856 treatment. These same genes were also induced in GBM cell lines U87MG, LN18, LN229, A172, and DBTRG upon AS1842856 treatment. By contrast, follow-up RNA interference (RNAi) targeting of FOXO1 led to reduced LEF1 and TCF7 gene expression in BT549 and U87MG cells. In agreement with RNAi experiments, CRISPR Cas9-mediated FOXO1 disruption reduced the expression of canonical WNT genes LEF1 and TCF7 in U87MG cells. The loss of TCF7 gene expression in FOXO1 disruption mutants was restored by exogenous expression of the DNA-binding-deficient FOXO1-H215R. Therefore, FOXO1 induces TCF7 in a DNA-binding-independent manner, similar to other published FOXO1-activated genes such as TCF4 and hes family bHLH transcription factor 1. Our work demonstrates that FOXO1 promotes canonical WNT gene expression in examined BBC and GBM cells, similar to results found in Drosophila melanogaster, T-cell development, and murine acute myeloid leukemia models.
Collapse
Affiliation(s)
- Shania Pintor
- Department of BiologyThe University of Texas Rio Grande ValleyEdinburgTXUSA
| | - Alma Lopez
- Department of BiologyThe University of Texas Rio Grande ValleyEdinburgTXUSA
| | - David Flores
- Department of BiologyThe University of Texas Rio Grande ValleyEdinburgTXUSA
| | - Brianda Lozoya
- Department of BiologyThe University of Texas Rio Grande ValleyEdinburgTXUSA
| | - Bipul Soti
- Department of BiologyThe University of Texas Rio Grande ValleyEdinburgTXUSA
| | - Rishi Pokhrel
- Department of BiologyThe University of Texas Rio Grande ValleyEdinburgTXUSA
| | - Joaquin Negrete
- Department of BiologyThe University of Texas Rio Grande ValleyEdinburgTXUSA
| | - Michael W. Persans
- Department of BiologyThe University of Texas Rio Grande ValleyEdinburgTXUSA
| | - Robert Gilkerson
- Department of BiologyThe University of Texas Rio Grande ValleyEdinburgTXUSA
- Medical Laboratory SciencesThe University of Texas Rio Grande ValleyEdinburgTXUSA
| | - Bonnie Gunn
- Department of BiologyThe University of Texas Rio Grande ValleyEdinburgTXUSA
| | - Megan Keniry
- Department of BiologyThe University of Texas Rio Grande ValleyEdinburgTXUSA
| |
Collapse
|
9
|
Sadaf, Hazazi A, Alkhalil SS, Alsaiari AA, Gharib AF, Alhuthali HM, Rana S, Aloliqi AA, Eisa AA, Hasan MR, Dev K. Role of Fork-Head Box Genes in Breast Cancer: From Drug Resistance to Therapeutic Targets. Biomedicines 2023; 11:2159. [PMID: 37626655 PMCID: PMC10452497 DOI: 10.3390/biomedicines11082159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/17/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer has been acknowledged as one of the most notorious cancers, responsible for millions of deaths around the globe. Understanding the various factors, genetic mutations, comprehensive pathways, etc., that are involved in the development of breast cancer and how these affect the development of the disease is very important for improving and revitalizing the treatment of this global health issue. The forkhead-box gene family, comprising 19 subfamilies, is known to have a significant impact on the growth and progression of this cancer. The article looks into the various forkhead genes and how they play a role in different types of cancer. It also covers their impact on cancer drug resistance, interaction with microRNAs, explores their potential as targets for drug therapies, and their association with stem cells.
Collapse
Affiliation(s)
- Sadaf
- Department of Biotechnology, Jamia Millia Islamia, New Delhi 110025, India;
| | - Ali Hazazi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh 11481, Saudi Arabia;
| | - Samia S. Alkhalil
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Alquwayiyah 11961, Saudi Arabia;
| | - Ahad Amer Alsaiari
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia; (A.A.A.); (A.F.G.); (H.M.A.)
| | - Amal F. Gharib
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia; (A.A.A.); (A.F.G.); (H.M.A.)
| | - Hayaa M. Alhuthali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia; (A.A.A.); (A.F.G.); (H.M.A.)
| | - Shanika Rana
- School of Biosciences, Apeejay Stya University, Gurugram 122003, India;
| | - Abdulaziz A. Aloliqi
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia;
| | - Alaa Abdulaziz Eisa
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Taibah University, Medina 30002, Saudi Arabia;
| | - Mohammad Raghibul Hasan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Alquwayiyah 11961, Saudi Arabia;
| | - Kapil Dev
- Department of Biotechnology, Jamia Millia Islamia, New Delhi 110025, India;
| |
Collapse
|
10
|
Sun X, Li Q, Tang Y, Hu W, Chen G, An H, Huang D, Tong T, Zhang Y. Epigenetic activation of secretory phenotypes in senescence by the FOXQ1-SIRT4-GDH signaling. Cell Death Dis 2023; 14:481. [PMID: 37516739 PMCID: PMC10387070 DOI: 10.1038/s41419-023-06002-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/31/2023]
Abstract
Although metabolic reprogramming is characterized as a hallmark of aging, implications of the crucial glutamate dehydrogenase (GDH) in human senescence remain poorly understood. Here, we report that GDH activity is significantly increased in aged mice and senescent human diploid fibroblasts. This enzymatic potentiation is associated with de-repression of GDH from its functionally suppressive ADP-ribosylation modification catalyzed by NAD-dependent ADP-ribosyltransferase/deacetylase SIRT4. A series of transcription analyses led to the identification of FOXQ1, a forkhead family transcription factor (TF), responsible for the maintenance of SIRT4 expression levels in juvenile cells. However, this metabolically balanced FOXQ1-SIRT4-GDH axis, is shifted in senescence with gradually decreasing expressions of FOXQ1 and SIRT4 and elevated GDH activity. Importantly, pharmaceutical inhibition of GDH suppresses the aberrantly activated transcription of IL-6 and IL-8, two major players in senescence-associated secretory phenotype (SASP), and this action is mechanistically associated with erasure of the repressive H3K9me3 (trimethylation of lysine 9 on histone H3) marks at IL-6 and IL-8 promoters, owing to the requirement of α-ketoglutaric acid (α-KG) from GDH-mediated glutamate dehydrogenase reaction as a cofactor for histone demethylation. In supplement with the phenotypic evidence from FOXQ1/SIRT4/GDH manipulations, these data support the integration of metabolism alterations and epigenetic regulation in driving senescence progression and highlight the FOXQ1-SIRT4-GDH axis as a novel druggable target for improving human longevity.
Collapse
Affiliation(s)
- Xinpei Sun
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Qian Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, 100081, Beijing, China
| | - Yunyi Tang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Wanjin Hu
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Gengyao Chen
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Hongguang An
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Daoyuan Huang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Tanjun Tong
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yu Zhang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
11
|
El-Tanani M, Nsairat H, Aljabali AA, Serrano-Aroca Á, Mishra V, Mishra Y, Naikoo GA, Alshaer W, Tambuwala MM. Role of mammalian target of rapamycin (mTOR) signalling in oncogenesis. Life Sci 2023; 323:121662. [PMID: 37028545 DOI: 10.1016/j.lfs.2023.121662] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/07/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023]
Abstract
The signalling system known as mammalian target of rapamycin (mTOR) is believed to be required for several biological activities involving cell proliferation. The serine-threonine kinase identified as mTOR recognises PI3K-AKT stress signals. It is well established in the scientific literature that the deregulation of the mTOR pathway plays a crucial role in cancer growth and advancement. This review focuses on the normal functions of mTOR as well as its abnormal roles in cancer development.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; Institute of Cancer Therapeutics, University of Bradford, Bradford, West Yorkshire BD7 1DP, United Kingdom.
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan.
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001, Valencia, Spain.
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Gowhar A Naikoo
- Department of Mathematics and Sciences, College of Arts and Applied Sciences, Dhofar University, Salalah, PC 211, Oman.
| | - Walhan Alshaer
- Cell Therapy Center, the University of Jordan, Amman 11942, Jordan
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, United Kingdom.
| |
Collapse
|
12
|
Transcription of Autophagy Associated Gene Expression as Possible Predictors of a Colorectal Cancer Prognosis. Biomedicines 2023; 11:biomedicines11020418. [PMID: 36830954 PMCID: PMC9952998 DOI: 10.3390/biomedicines11020418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
(1) Background: Autophagy plays a dual role in oncogenesis-it contributes to the growth of the tumor and can inhibit its development. The aim of this study was to assess changes in the transcriptional activity of LAMP-2, BECN1, PINK1, and FOXO1 genes involved in the autophagy process in histopathologically confirmed adenocarcinoma sections of colorectal cancer: (2) Methods: A gene expression profile analysis was performed using HG-U133A and the RT-qPCR reaction. The transcriptional activity of genes was compared in sections of colorectal cancer in the four clinical stages (CSI-CSIV) concerning the control group; (3) Results: In CSI, the transcriptional activity of the PINK1 gene is highest; in CS II, the LAMP-2 gene is highest, while FOXO1 increases gradually from CSI reaching a maximum in CSIII. There is no BECN1 gene expression in colorectal cancer cells; (4) Conclusions: The observed differences in the mRNA concentration profile of autophagy-related genes in colon cancer specimens may indicate the role of autophagy in the pathogenesis of this cancer. Genes involved in autophagy may be diagnostic tools for colorectal cancer screening and personalized therapy in the future.
Collapse
|
13
|
Frenay J, Bellaye PS, Oudot A, Helbling A, Petitot C, Ferrand C, Collin B, Dias AMM. IL-1RAP, a Key Therapeutic Target in Cancer. Int J Mol Sci 2022; 23:ijms232314918. [PMID: 36499246 PMCID: PMC9735758 DOI: 10.3390/ijms232314918] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer is a major cause of death worldwide and especially in high- and upper-middle-income countries. Despite recent progress in cancer therapies, such as chimeric antigen receptor T (CAR-T) cells or antibody-drug conjugate (ADC), new targets expressed by the tumor cells need to be identified in order to selectively drive these innovative therapies to tumors. In this context, IL-1RAP recently showed great potential to become one of these new targets for cancer therapy. IL-1RAP is highly involved in the inflammation process through the interleukins 1, 33, and 36 (IL-1, IL-33, IL-36) signaling pathways. Inflammation is now recognized as a hallmark of carcinogenesis, suggesting that IL-1RAP could play a role in cancer development and progression. Furthermore, IL-1RAP was found overexpressed on tumor cells from several hematological and solid cancers, thus confirming its potential involvement in carcinogenesis. This review will first describe the structure and genetics of IL-1RAP as well as its role in tumor development. Finally, a focus will be made on the therapies based on IL-1RAP targeting, which are now under preclinical or clinical development.
Collapse
Affiliation(s)
- Jame Frenay
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Pierre-Simon Bellaye
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Alexandra Oudot
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Alex Helbling
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Camille Petitot
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Christophe Ferrand
- INSERM UMR1098, EFS BFC, Université de Bourgogne Franche-Comté, 25000 Besançon, France
- CanCell Therapeutics, 25000 Besançon, France
| | - Bertrand Collin
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR CNRS 6302, 21000 Dijon, France
| | - Alexandre M M Dias
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| |
Collapse
|
14
|
Yin X, Liu Q, Liu F, Tian X, Yan T, Han J, Jiang S. Emerging Roles of Non-proteolytic Ubiquitination in Tumorigenesis. Front Cell Dev Biol 2022; 10:944460. [PMID: 35874839 PMCID: PMC9298949 DOI: 10.3389/fcell.2022.944460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/15/2022] [Indexed: 12/13/2022] Open
Abstract
Ubiquitination is a critical type of protein post-translational modification playing an essential role in many cellular processes. To date, more than eight types of ubiquitination exist, all of which are involved in distinct cellular processes based on their structural differences. Studies have indicated that activation of the ubiquitination pathway is tightly connected with inflammation-related diseases as well as cancer, especially in the non-proteolytic canonical pathway, highlighting the vital roles of ubiquitination in metabolic programming. Studies relating degradable ubiquitination through lys48 or lys11-linked pathways to cellular signaling have been well-characterized. However, emerging evidence shows that non-degradable ubiquitination (linked to lys6, lys27, lys29, lys33, lys63, and Met1) remains to be defined. In this review, we summarize the non-proteolytic ubiquitination involved in tumorigenesis and related signaling pathways, with the aim of providing a reference for future exploration of ubiquitination and the potential targets for cancer therapies.
Collapse
Affiliation(s)
- Xiu Yin
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Qingbin Liu
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Fen Liu
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Xinchen Tian
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining Medical University, Jining, China.,Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tinghao Yan
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining Medical University, Jining, China.,Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Han
- Department of Thyroid and Breast Surgery, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Shulong Jiang
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining Medical University, Jining, China
| |
Collapse
|
15
|
Zhang L, Liang B, Xu H, Gong Y, Hu W, Jin Z, Wu X, Chen X, Li M, Shi L, Shi Y, Wang Y, Yang L. Cinobufagin induces FOXO1-regulated apoptosis, proliferation, migration, and invasion by inhibiting G9a in non-small-cell lung cancer A549 cells. JOURNAL OF ETHNOPHARMACOLOGY 2022; 291:115095. [PMID: 35176466 DOI: 10.1016/j.jep.2022.115095] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/23/2022] [Accepted: 02/06/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bufonis (VB), an animal drug called Chansu in China, is the product of the secretion of Bufo gargarizans Cantor or B. melanostictus Schneider. As a traditional Chinese medicine (TCM) for a long time, it has been widely used in the treatment of heart failure, ulcer, pain, and various cancers. Cinobufaginn (CNB), the cardiotonic steroid or bufalene lactone extracted from VB, has the effects of detoxification, detumescence, and analgesia. AIM OF THE STUDY The present study aimed to define the effects of CNB on non-small-cell lung cancer (NSCLC) and identify the potential molecular mechanisms. MATERIALS AND METHODS A549 cells were treated with cinobufagin and cell viability, apoptosis, migration, and invasion were then evaluated using Cell Counting Kit-8 (CCK8) assays, flow cytometry, and Transwell assays, respectively. Moreover, the levels of proliferating cell nuclear antigen (PCNA), cytokeratin8 (CK8), poly ADP-ribose polymerase (PARP), Caspase3, Caspase8, B-cell lymphoma/lewkmia-2(Bcl-2), Bcl2-Associated X(Bax), forkhead box O1 (FOXO1), and euchromatic histone-lysine N-methyltransferase2 (G9a, EHMT2) in A549 cells were evaluated using qRT-PCR and/or Western blot analysis (WB), Co-IP, immunofluorescence, and immunohistochemistry. An in vivo imaging system, TUNEL, Immunofluorescence, and immunohistochemistry were also used to detect proliferating cell nuclear antigen(PCNA), Ki67, E-Cadherin(E-Cad), FOXO1, and G9a in mouse xenograft model experiments. RESULTS CNB suppressed cell proliferation, migration, and invasion but promoted apoptosis in A549 cells in a dose- and time-dependent manner, while cinobufagin had no cytotoxic effect on BEAS-2B cells. In vivo, cinobufagin inhibited the proliferation, migration, and invasion of A549 cells and promoted their apoptosis. The occurrence of the above phenomena was accompanied by an increase in FOXO1 expression and a decrease in G9a expression. In A549 cells, CNB did not reverse the changes in the proliferation, migration, invasion, and apoptosis of A549 cells after FOXO1 was successfully silenced. CONCLUSION Our study provides the first evidence that cinobufagin suppresses the malignant biological behaviours of NSCLC cells in vivo and in vitro and suggests that mechanistically, this effect may be achieved by inhibiting the expression of the histone methyltransferase G9a and activating the tumour suppressor gene FOXO1. Taken together, our findings provide important insights into the molecular mechanism underlying cinobufagin's anticancer activity, and suggest that cinobufagin could be a candidate for targeted cancer therapy.
Collapse
Affiliation(s)
- Lingling Zhang
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bing Liang
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Xu
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanju Gong
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wangming Hu
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhong Jin
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Wu
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiongbin Chen
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Min Li
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liangqin Shi
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yaping Shi
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Wang
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Lan Yang
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
16
|
Quque M, Villette C, Criscuolo F, Sueur C, Bertile F, Heintz D. Eusociality is linked to caste-specific differences in metabolism, immune system, and somatic maintenance-related processes in an ant species. Cell Mol Life Sci 2021; 79:29. [PMID: 34971425 PMCID: PMC11073003 DOI: 10.1007/s00018-021-04024-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/08/2021] [Accepted: 11/05/2021] [Indexed: 01/08/2023]
Abstract
The social organization of many primate, bird and rodent species and the role of individuals within that organization are associated with specific individual physiological traits. However, this association is perhaps most pronounced in eusocial insects (e.g., termites, ants). In such species, genetically close individuals show significant differences in behavior, physiology, and life expectancy. Studies addressing the metabolic changes according to the social role are still lacking. We aimed at understanding how sociality could influence essential molecular processes in a eusocial insect, the black garden ant (Lasius niger) where queens can live up to ten times longer than workers. Using mass spectrometry-based analysis, we explored the whole metabolome of queens, nest-workers and foraging workers. A former proteomics study done in the same species allowed us to compare the findings of both approaches. Confirming the former results at the proteome level, we showed that queens had fewer metabolites related to immunity. Contrary to our predictions, we did not find any metabolite linked to reproduction in queens. Among the workers, foragers had a metabolic signature reflecting a more stressful environment and a more highly stimulated immune system. We also found that nest-workers had more digestion-related metabolites. Hence, we showed that specific metabolic signatures match specific social roles. Besides, we identified metabolites differently expressed among behavioral castes and involved in nutrient sensing and longevity pathways (e.g., sirtuins, FOXO). The links between such molecular pathways and aging being found in an increasing number of taxa, our results confirm and strengthen their potential universality.
Collapse
Affiliation(s)
- Martin Quque
- Université de Strasbourg, CNRS, IPHC UMR 7178, 23 rue du Loess, F-67000, Strasbourg, France.
| | - Claire Villette
- Plant Imaging and Mass Spectrometry (PIMS), Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, 12 rue du Général Zimmer, F-67000, Strasbourg, France
| | - François Criscuolo
- Université de Strasbourg, CNRS, IPHC UMR 7178, 23 rue du Loess, F-67000, Strasbourg, France
| | - Cédric Sueur
- Université de Strasbourg, CNRS, IPHC UMR 7178, 23 rue du Loess, F-67000, Strasbourg, France
- Institut Universitaire de France, 75005, Paris, France
| | - Fabrice Bertile
- Université de Strasbourg, CNRS, IPHC UMR 7178, 23 rue du Loess, F-67000, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI, FR2048, Strasbourg, France
| | - Dimitri Heintz
- Plant Imaging and Mass Spectrometry (PIMS), Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, 12 rue du Général Zimmer, F-67000, Strasbourg, France
| |
Collapse
|
17
|
He Y, Sun MM, Zhang GG, Yang J, Chen KS, Xu WW, Li B. Targeting PI3K/Akt signal transduction for cancer therapy. Signal Transduct Target Ther 2021; 6:425. [PMID: 34916492 PMCID: PMC8677728 DOI: 10.1038/s41392-021-00828-5] [Citation(s) in RCA: 729] [Impact Index Per Article: 182.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 11/02/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt pathway plays a crucial role in various cellular processes and is aberrantly activated in cancers, contributing to the occurrence and progression of tumors. Examining the upstream and downstream nodes of this pathway could allow full elucidation of its function. Based on accumulating evidence, strategies targeting major components of the pathway might provide new insights for cancer drug discovery. Researchers have explored the use of some inhibitors targeting this pathway to block survival pathways. However, because oncogenic PI3K pathway activation occurs through various mechanisms, the clinical efficacies of these inhibitors are limited. Moreover, pathway activation is accompanied by the development of therapeutic resistance. Therefore, strategies involving pathway inhibitors and other cancer treatments in combination might solve the therapeutic dilemma. In this review, we discuss the roles of the PI3K/Akt pathway in various cancer phenotypes, review the current statuses of different PI3K/Akt inhibitors, and introduce combination therapies consisting of signaling inhibitors and conventional cancer therapies. The information presented herein suggests that cascading inhibitors of the PI3K/Akt signaling pathway, either alone or in combination with other therapies, are the most effective treatment strategy for cancer.
Collapse
Affiliation(s)
- Yan He
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Miao Miao Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Tumor Pathology, Zhengzhou, China
| | - Guo Geng Zhang
- MOE Key Laboratory of Tumor Molecular Biology and Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jing Yang
- MOE Key Laboratory of Tumor Molecular Biology and Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Kui Sheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Tumor Pathology, Zhengzhou, China.
| | - Wen Wen Xu
- MOE Key Laboratory of Tumor Molecular Biology and Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Bin Li
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China.
| |
Collapse
|
18
|
Kitamoto T, Kuo T, Okabe A, Kaneda A, Accili D. An integrative transcriptional logic model of hepatic insulin resistance. Proc Natl Acad Sci U S A 2021; 118:e2102222118. [PMID: 34732569 PMCID: PMC8609333 DOI: 10.1073/pnas.2102222118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2021] [Indexed: 12/27/2022] Open
Abstract
Abnormalities of lipid/lipoprotein and glucose metabolism are hallmarks of hepatic insulin resistance in type 2 diabetes. The former antedate the latter, but the latter become progressively refractory to treatment and contribute to therapeutic failures. It's unclear whether the two processes share a common pathogenesis and what underlies their progressive nature. In this study, we investigated the hypothesis that genes in the lipid/lipoprotein pathway and those in the glucose metabolic pathway are governed by different transcriptional regulatory logics that affect their response to physiologic (fasting/refeeding) as well as pathophysiologic cues (insulin resistance and hyperglycemia). To this end, we obtained genomic and transcriptomic maps of the key insulin-regulated transcription factor, FoxO1, and integrated them with those of CREB, PPAR-α, and glucocorticoid receptor. We found that glucose metabolic genes are primarily regulated by promoter and intergenic enhancers in a fasting-dependent manner, while lipid genes are regulated through fasting-dependent intron enhancers and fasting-independent enhancerless introns. Glucose genes also showed a remarkable transcriptional resiliency (i.e., the ability to compensate following constitutive FoxO1 ablation through an enrichment of active marks at shared PPAR-α/FoxO1 regulatory elements). Unexpectedly, insulin resistance and hyperglycemia were associated with a "spreading" of FoxO1 binding to enhancers and the emergence of unique target sites. We surmise that this unusual pattern correlates with the progressively intractable nature of hepatic insulin resistance. This transcriptional logic provides an integrated model to interpret the combined lipid and glucose abnormalities of type 2 diabetes.
Collapse
Affiliation(s)
- Takumi Kitamoto
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032;
- Naomi Berrie Diabetes Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Taiyi Kuo
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
- Naomi Berrie Diabetes Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Atsushi Okabe
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
| | - Domenico Accili
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
- Naomi Berrie Diabetes Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| |
Collapse
|
19
|
Wang K, Li Y, Qiang T, Chen J, Wang X. Role of epigenetic regulation in myocardial ischemia/reperfusion injury. Pharmacol Res 2021; 170:105743. [PMID: 34182132 DOI: 10.1016/j.phrs.2021.105743] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/09/2021] [Accepted: 06/23/2021] [Indexed: 12/30/2022]
Abstract
Nowadays acute myocardial infarction (AMI) is a serious cardiovascular disease threatening the human life and health worldwide. The most effective treatment is to quickly restore coronary blood flow through revascularization. However, timely revascularization may lead to reperfusion injury, thereby reducing the clinical benefits of revascularization. At present, no effective treatment is available for myocardial ischemia/reperfusion injury. Emerging evidence indicates that epigenetic regulation is closely related to the pathogenesis of myocardial ischemia/reperfusion injury, indicating that epigenetics may serve as a novel therapeutic target to ameliorate or prevent ischemia/reperfusion injury. This review aimed to briefly summarize the role of histone modification, DNA methylation, noncoding RNAs, and N6-methyladenosine (m6A) methylation in myocardial ischemia/reperfusion injury, with a view to providing new methods and ideas for the research and treatment of myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Keyan Wang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China
| | - Yiping Li
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China
| | - Tingting Qiang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China
| | - Jie Chen
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China
| | - Xiaolong Wang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China.
| |
Collapse
|
20
|
Build-UPS and break-downs: metabolism impacts on proteostasis and aging. Cell Death Differ 2021; 28:505-521. [PMID: 33398091 PMCID: PMC7862225 DOI: 10.1038/s41418-020-00682-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/30/2022] Open
Abstract
Perturbation of metabolism elicits cellular stress which profoundly modulates the cellular proteome and thus protein homeostasis (proteostasis). Consequently, changes in the cellular proteome due to metabolic shift require adaptive mechanisms by molecular protein quality control. The mechanisms vitally controlling proteostasis embrace the entire life cycle of a protein involving translational control at the ribosome, chaperone-assisted native folding, and subcellular sorting as well as proteolysis by the proteasome or autophagy. While metabolic imbalance and proteostasis decline have been recognized as hallmarks of aging and age-associated diseases, both processes are largely considered independently. Here, we delineate how proteome stability is governed by insulin/IGF1 signaling (IIS), mechanistic target of Rapamycin (TOR), 5′ adenosine monophosphate-activated protein kinase (AMPK), and NAD-dependent deacetylases (Sir2-like proteins known as sirtuins). This comprehensive overview is emphasizing the regulatory interconnection between central metabolic pathways and proteostasis, indicating the relevance of shared signaling nodes as targets for future therapeutic interventions. ![]()
Collapse
|
21
|
Liu YR, Cheng YQ, Wang SB, Su YR, Liu Y, Li CY, Jin L, Wan Q, Sang X, Wang ZC. Therapeutic effects and perspective of stem cell extracellular vesicles in aging and cancer. J Cell Physiol 2020; 236:4783-4796. [PMID: 33368322 DOI: 10.1002/jcp.30212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/02/2020] [Accepted: 11/28/2020] [Indexed: 12/16/2022]
Abstract
Senescent cells can secrete a plethora of cytokines which induce senescent phenotype of neighboring cells and was called senescence-associated secretory phenotype. Previously, it was believed that cancer was caused by the infinite division and uncontrolled proliferation of cells. Based on this, anticancer treatments were all aimed at killing cancer cells. Cancer is now considered an age-related disease. Cancer cells are not exogenous, but one of the worst results of injuries which initially induce cell senescence. Therefore, reversing cell senescence can fundamentally prevent and treat cancer. Though current anticancer treatments induce the cancer cells apoptosis, they induce senescence of normal cells at the same time, thus promoting the occurrence and development of cancer and forming a vicious circle. Extracellular vesicles (EVs) are nano-sized vesicles which partially mirror their parent cells. In the tumor microenvironment, EVs of senescent cells can change the expression profile of cancer cells, contributing to their resistance to chemotherapy. There is growing evidence indicates that stem cell EVs exert effective antiaging and anticancer actions by transferring functional microRNAs and proteins. This review will summarize the therapeutic role of stem cell EVs in reversing aging and cancer, which suggests the broad clinical application perspective.
Collapse
Affiliation(s)
- Yu-Run Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ya-Qi Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shou-Bi Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ya-Ru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ying Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chao-Yang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lin Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Qi Wan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xuan Sang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Chong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
22
|
Goswami S, Kareem O, Goyal RK, Mumtaz SM, Tonk RK, Gupta R, Pottoo FH. Role of Forkhead Transcription Factors of the O Class (FoxO) in Development and Progression of Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:709-721. [PMID: 33001019 DOI: 10.2174/1871527319666201001105553] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 07/20/2020] [Accepted: 08/31/2020] [Indexed: 11/22/2022]
Abstract
In the Central Nervous System (CNS), a specific loss of focal neurons leads to mental and neurological disorders like dementia, Alzheimer's Disease (AD), Huntington's disease, Parkinson's disease, etc. AD is a neurological degenerative disorder, which is progressive and irreversible in nature and is the widely recognized reason for dementia in the geriatric populace. It affects 10% of people above the age of 65 and is the fourth driving reason for death in the United States. Numerous evidence suggests that the neuronal compartment is not the only genesis of AD, but transcription factors also hold significant importance in the occurrence and advancement of the disease. It is the need of the time to find the novel molecular targets and new techniques for treating or slowing down the progression of neurological disorders, especially AD. In this article, we summarised a conceivable association between transcriptional factors and their defensive measures against neurodegeneration and AD. The mammalian forkhead transcription factors of the class O (FoxO) illustrate one of the potential objectives for the development of new methodologies against AD and other neurocognitive disorders. The presence of FoxO is easily noticeable in the "cognitive centers" of the brain, specifically in the amygdala, hippocampus, and the nucleus accumbens. FoxO proteins are the prominent and necessary factors in memory formation and cognitive functions. FoxO also assumes a pertinent role in the protection of multiple cells in the brain by controlling the involving mechanism of autophagy and apoptosis and also modulates the process of phosphorylation of the targeted protein, thus FoxO must be a putative target in the mitigation of AD. This review features the role of FoxO as an important biomarker and potential new targets for the treatment of AD.
Collapse
Affiliation(s)
- Shikha Goswami
- Delhi Pharmaceutical Sciences and Research University, Mehrauli- Badarpur Rd, Sector 3, PushpVihar, New Delhi, India
| | - Ozaifa Kareem
- Department of Pharmaceutical Sciences, Faculty of Applied Sciences and Technology, University of Kashmir, Srinagar, JK, India
| | - Ramesh K Goyal
- Delhi Pharmaceutical Sciences and Research University, Mehrauli- Badarpur Rd, Sector 3, PushpVihar, New Delhi, India
| | - Sayed M Mumtaz
- Delhi Pharmaceutical Sciences and Research University, Mehrauli- Badarpur Rd, Sector 3, PushpVihar, New Delhi, India
| | - Rajiv K Tonk
- Delhi Pharmaceutical Sciences and Research University, Mehrauli- Badarpur Rd, Sector 3, PushpVihar, New Delhi, India
| | - Rahul Gupta
- Delhi Pharmaceutical Sciences and Research University, Mehrauli- Badarpur Rd, Sector 3, PushpVihar, New Delhi, India
| | - Faheem H Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University P.O.BOX 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
23
|
Comparison of miRNA Expression Profiles between HIV-1 and HIV-2 Infected Monocyte-Derived Macrophages (MDMs) and Peripheral Blood Mononuclear Cells (PBMCs). Int J Mol Sci 2020; 21:ijms21186970. [PMID: 32971935 PMCID: PMC7556008 DOI: 10.3390/ijms21186970] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
During the progression of HIV-1 infection, macrophage tropic HIV-1 that use the CCR5 co-receptor undergoes a change in co-receptor use to CXCR4 that is predominately T cell tropic. This change in co-receptor preference makes the virus able to infect T cells. HIV-2 is known to infect MDMs and T cells and is dual tropic. The aim of this study was to elucidate the differential expression profiles of host miRNAs and their role in cells infected with HIV-1/HIV-2. To achieve this goal, a comparative global miRNA expression profile was determined in human PBMCs and MDMs infected with HIV-1/HIV-2. Differentially expressed miRNAs were identified in HIV-1/HIV-2 infected PBMCs and MDMs using the next-generation sequencing (NGS) technique. A comparative global miRNA expression profile in infected MDMs and PBMCs with HIV-1 and HIV-2 identified differential expression of several host miRNAs. These differentially expressed miRNAs are likely to be involved in many signaling pathways, like the p53 signaling pathway, PI3K-Akt signaling pathways, MAPK signaling pathways, FoxO signaling pathway, and viral carcinogenesis. Thus, a comparative study of the differential expression of host miRNAs in MDMs and T cell in response to HIV-1 and HIV-2 infection will help us to identify unique biomarkers that can differentiate HIV-1 and HIV-2 infection.
Collapse
|
24
|
Molecular characterization and expression analysis of foxo3l in response to exogenous hormones in black rockfish (Sebastes schlegelii). Gene 2020; 753:144777. [PMID: 32428695 DOI: 10.1016/j.gene.2020.144777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/07/2020] [Accepted: 05/14/2020] [Indexed: 11/22/2022]
Abstract
As a crucial member of the Forkhead Box family, class O (FoxO) plays an essential role in growth, cell differentiation, metabolism, immunization, and apoptosis. Meanwhile, FoxO3 is the primary regulator and effective inhibitor of primordial follicle activation. In this study, seven foxo genes were identified in black rockfish (Sebastes schlegelii), including two foxo1 genes (foxo1a, foxo1b), two foxo3 genes (foxo3, foxo3l), one foxo4 gene, and two foxo6 genes (foxo6a, foxo6b). foxo3l was derived from teleost-specific whole-genome duplication events. Evaluation of tissue expression pattern revealed that foxo3l displayed sexually dimorphic expression with a high level in the ovary and spatial expression only in the cytoplasm of follicle cells and oocytes. When the ovaries were stimulated by estrogen and gonadotropin, foxo3l expression was remarkably reduced, and the effect of androgen was completely different. We considered that foxo3l lost its ability to inhibit follicular precocity because of mass ovulation by hormone stimulation, resulting in its decreased expression. Such evidence indicated that foxo3l is an important regulator of reproduction-related functions in black rockfish. This study provides new insights into foxo3l genes for further functional research in teleost.
Collapse
|
25
|
Cheung KCP, Fanti S, Mauro C, Wang G, Nair AS, Fu H, Angeletti S, Spoto S, Fogolari M, Romano F, Aksentijevic D, Liu W, Li B, Cheng L, Jiang L, Vuononvirta J, Poobalasingam TR, Smith DM, Ciccozzi M, Solito E, Marelli-Berg FM. Preservation of microvascular barrier function requires CD31 receptor-induced metabolic reprogramming. Nat Commun 2020; 11:3595. [PMID: 32681081 PMCID: PMC7367815 DOI: 10.1038/s41467-020-17329-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/09/2020] [Indexed: 12/19/2022] Open
Abstract
Endothelial barrier (EB) breaching is a frequent event during inflammation, and it is followed by the rapid recovery of microvascular integrity. The molecular mechanisms of EB recovery are poorly understood. Triggering of MHC molecules by migrating T-cells is a minimal signal capable of inducing endothelial contraction and transient microvascular leakage. Using this model, we show that EB recovery requires a CD31 receptor-induced, robust glycolytic response sustaining junction re-annealing. Mechanistically, this response involves src-homology phosphatase activation leading to Akt-mediated nuclear exclusion of FoxO1 and concomitant β-catenin translocation to the nucleus, collectively leading to cMyc transcription. CD31 signals also sustain mitochondrial respiration, however this pathway does not contribute to junction remodeling. We further show that pathologic microvascular leakage in CD31-deficient mice can be corrected by enhancing the glycolytic flux via pharmacological Akt or AMPK activation, thus providing a molecular platform for the therapeutic control of EB response.
Collapse
Affiliation(s)
- Kenneth C P Cheung
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- School of Life Sciences, Centre for Cell & Developmental Biology and Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Silvia Fanti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Claudio Mauro
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Mindelson Way, Birmingham, B152WB, UK
| | - Guosu Wang
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Anitha S Nair
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Hongmei Fu
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Silvia Angeletti
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Rome, Italy
| | - Silvia Spoto
- Internal Medicine Department, University campus Bio-Medico of Rome, Rome, Italy
| | - Marta Fogolari
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Rome, Italy
| | - Francesco Romano
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Rome, Italy
| | - Dunja Aksentijevic
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Weiwei Liu
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, 510060, People's Republic of China
| | - Baiying Li
- School of Life Sciences, Centre for Cell & Developmental Biology and Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lixin Cheng
- School of Life Sciences, Centre for Cell & Developmental Biology and Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Liwen Jiang
- School of Life Sciences, Centre for Cell & Developmental Biology and Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Juho Vuononvirta
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Thanushiyan R Poobalasingam
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - David M Smith
- AstraZeneca R&D, Cambridge Science Park, Milton Road, Cambridge, CB4 0WG, UK
| | - Massimo Ciccozzi
- Unit of Medical Statistic and Molecular Epidemiology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Egle Solito
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universita degli studi di Napoli "Federico II", 80131, Naples, Italy
| | - Federica M Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
- Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, Charterhouse Square, London, UK.
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW FOXOs are transcription factors that regulate downstream target genes to counteract to cell stress. Here we review the function and regulation of FOXO transcription factors, the mechanism of FOXO3 activation in the kidney, and the role of FOXO3 in delaying the development of chronic kidney disease (CKD). RECENT FINDINGS Progressive renal hypoxia from vascular dropout and metabolic perturbation is a pathogenic factor for the initiation and development of CKD. Hypoxia and low levels of α-ketoglutarate generated from the TCA cycle inhibit prolyl hydroxylase domain (PHD)-mediated prolyl hydroxylation of FoxO3, thus reducing FoxO3 protein degradation via the ubiquitin proteasomal pathway, similar to HIF stabilization under hypoxic conditions. FoxO3 accumulation and nuclear translocation activate two key cellular defense mechanisms, autophagy and antioxidative response in renal tubular cells, to reduce cell injury and promote cell survival. FoxO3 directly activates the expression of Atg proteins, which replenishes core components of the autophagic machinery to allow sustained autophagy in the chronically hypoxic kidney. FoxO3 protects mitochondria by stimulating the expression of superoxide dismutase 2 (SOD2), as tubular deletion of FoxO3 in mice results in reduced SOD2 levels and profound mitochondrial damage. SUMMARY Knowledge gained from animal studies may help understand the function of stress responsive transcription factors that could be targeted to prevent or treat CKD.
Collapse
Affiliation(s)
- Fangming Lin
- Division of Pediatric Nephrology, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
27
|
Yang S, Li L, Yu L, Sun L, Li K, Tong C, Xu W, Cui G, Long M, Li P. Selenium-enriched yeast reduces caecal pathological injuries and intervenes changes of the diversity of caecal microbiota caused by Ochratoxin-A in broilers. Food Chem Toxicol 2020; 137:111139. [DOI: 10.1016/j.fct.2020.111139] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/06/2020] [Accepted: 01/19/2020] [Indexed: 12/11/2022]
|
28
|
Hoxhaj G, Manning BD. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat Rev Cancer 2020; 20:74-88. [PMID: 31686003 PMCID: PMC7314312 DOI: 10.1038/s41568-019-0216-7] [Citation(s) in RCA: 1259] [Impact Index Per Article: 251.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/30/2019] [Indexed: 02/06/2023]
Abstract
The altered metabolic programme of cancer cells facilitates their cell-autonomous proliferation and survival. In normal cells, signal transduction pathways control core cellular functions, including metabolism, to couple the signals from exogenous growth factors, cytokines or hormones to adaptive changes in cell physiology. The ubiquitous, growth factor-regulated phosphoinositide 3-kinase (PI3K)-AKT signalling network has diverse downstream effects on cellular metabolism, through either direct regulation of nutrient transporters and metabolic enzymes or the control of transcription factors that regulate the expression of key components of metabolic pathways. Aberrant activation of this signalling network is one of the most frequent events in human cancer and serves to disconnect the control of cell growth, survival and metabolism from exogenous growth stimuli. Here we discuss our current understanding of the molecular events controlling cellular metabolism downstream of PI3K and AKT and of how these events couple two major hallmarks of cancer: growth factor independence through oncogenic signalling and metabolic reprogramming to support cell survival and proliferation.
Collapse
Affiliation(s)
- Gerta Hoxhaj
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Brendan D Manning
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
29
|
Lian S, Wang J, Zhang L, Xing Q, Hu N, Liu S, Dai X, Zhang F, Hu X, Bao Z, Wang S. Integration of Biochemical, Cellular, and Genetic Indicators for Understanding the Aging Process in a Bivalve Mollusk Chlamys farreri. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2019; 21:718-730. [PMID: 31392593 DOI: 10.1007/s10126-019-09917-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/24/2019] [Indexed: 06/10/2023]
Abstract
The major causal factors for the irreversible decline in physical vitality during organismal aging are postulated to be a chronic state of cellular redox imbalance, metabolic toxicity, and impaired energy homeostasis. We assessed whether the relevant enzyme activity, oxidative stress, and intracellular ATP might be causally involved in the aging of short-lived Chlamys farreri (life span 4~5 years). A total of eight related biochemical and cellular indicators were chosen for the subsequent analysis. All the indicators were measured in seven different tissues from scallops aged one to four years, and our data support that the aging of C. farreri is associated with attenuated tissue enzyme activity as well as a decreased metabolic rate. Through principal component analysis, we developed an integrated vigor index for each tissue for comprehensive age-related fitness evaluation. Remarkably, all tissue-integrated vigor indexes significantly declined with age, and the kidney was observed to be the most representative tissue. Further transcriptional profiling of the enzymatic genes provided additional detail on the molecular responses that may underlie the corresponding biochemical results. Moreover, these critical molecular responses may be attributed to the conserved hierarchical regulators, e.g., FOXO, AMPKs, mTOR, and IGF1R, which were identified as potentially novel markers for chronic fitness decline with age in bivalves. The present study provides a systematic approach that could potentially benefit the global assessment of the aging process in C. farreri and provide detailed evaluation of the biochemical, cellular, and genetic indicators that might be involved. This information may assist in a better understanding of bivalve adaptability and life span.
Collapse
Affiliation(s)
- Shanshan Lian
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Jing Wang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Lingling Zhang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Qiang Xing
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Naina Hu
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Sinuo Liu
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Xiaoting Dai
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Fengmei Zhang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Xiaoli Hu
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Zhenmin Bao
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Shi Wang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
30
|
Chae YC, Kim JY, Park JW, Kim KB, Oh H, Lee KH, Seo SB. FOXO1 degradation via G9a-mediated methylation promotes cell proliferation in colon cancer. Nucleic Acids Res 2019; 47:1692-1705. [PMID: 30535125 PMCID: PMC6393239 DOI: 10.1093/nar/gky1230] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/07/2018] [Accepted: 11/28/2018] [Indexed: 12/15/2022] Open
Abstract
Posttranslational modifications of the Forkhead family transcription factor, FOXO1, have been known to have important regulatory implications in its diverse activities. Several types of modifications of FOXO1, including acetylation, phosphorylation, and ubiquitination, have been reported. However, lysine methylation of FOXO1 has not yet been identified. Here, we reported that FOXO1 is methylated by G9a at K273 residue in vitro and in vivo. Methylation of FOXO1 by G9a increased interaction between FOXO1 and a specific E3 ligase, SKP2, and decreased FOXO1 protein stability. In addition, G9a expression was increased by insulin and resulted in insulin-mediated FOXO1 degradation by K273 methylation. Tissue array analysis indicated that G9a was overexpressed and FOXO1 levels decreased in human colon cancer. Cell proliferation assays revealed that G9a-mediated FOXO1 methylation increased colon cancer cell proliferation. Fluorescence-activated cell sorting (FACS) analysis indicated that apoptosis rates were higher in the presence of FOXO1 than in FOXO1 knock-out cells. Furthermore, we found that G9a protein levels were elevated and FOXO1 protein levels were decreased in human colon cancer patients tissue samples. Here, we report that G9a specific inhibitor, BIX-01294, can regulate cell proliferation and apoptosis by inhibiting G9a-mediated FOXO1 methylation.
Collapse
Affiliation(s)
- Yun-Cheol Chae
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul 156-756, South Korea
| | - Ji-Young Kim
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul 156-756, South Korea
| | - Jin Woo Park
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul 156-756, South Korea
| | - Kee-Beom Kim
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul 156-756, South Korea
| | - Hyein Oh
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul 156-756, South Korea
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Hwasun Hospital and Medical School, Hwasun, Jeollanam-do, South Korea
| | - Sang-Beom Seo
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul 156-756, South Korea
| |
Collapse
|
31
|
Wang B, Wang Y, Wang L, He X, He Y, Bai M, Zhu L, Zheng J, Yuan D, Jin T. The role of FOXO3 polymorphisms in susceptibility to tuberculosis in a Chinese population. Mol Genet Genomic Med 2019; 7:e770. [PMID: 31241240 PMCID: PMC6687658 DOI: 10.1002/mgg3.770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/27/2019] [Accepted: 05/08/2019] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Tuberculosis (TB) is a significant worldwide health problem, and is caused by Mycobacteria tuberculosis. Recent studies have suggested that FOXO3 plays vital roles in the risk of immune-related infectious diseases such as TB. METHODS AND RESULTS The present study aimed to evaluate FOXO3 genetic variants and TB risk. We recruited 510 TB patients and 508 healthy controls in this study. All subjects were genotyped with the Agena MassARRAY platform. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated using logistic regression adjusted for age and gender. Our result revealed that rs3800229 T/G and rs4946935 G/A genotypes significantly increased the risk of TB (OR = 1.34, 95% CI = 1.04-1.74, p = 0.026; OR = 1.34, 95% CI = 1.03-1.73, p = 0.029, respectively). In stratified analysis according to gender and age, we observed that rs3800229 T/G and rs4946935 G/A genotypes were associated with an increase the risk of TB among males and age ≤41 years, respectively (OR = 1.47, 95% CI = 1.06-2.04, p = 0.022 and OR = 1.45, 95% CI = 1.05-2.02, p = 0.025). CONCLUSIONS Our study showed that rs3800229 and rs4946935 in FOXO3 were associated with a risk of TB in the Chinese population.
Collapse
Affiliation(s)
- Bo Wang
- Department of the 4th Internal MedicineXi’an Chest HospitalXi’anShaanxiChina
| | - Yuhe Wang
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
- Department of Clinical LaboratoryAffiliated Hospital of Xizang Minzu UniversityXianyangShaanxiChina
| | - Li Wang
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
- School of Basic Medical SciencesXizang Minzu UniversityXianyangShaanxiChina
| | - Xue He
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
- School of Basic Medical SciencesXizang Minzu UniversityXianyangShaanxiChina
| | - Yongjun He
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
- School of Basic Medical SciencesXizang Minzu UniversityXianyangShaanxiChina
| | - Mei Bai
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
- School of Basic Medical SciencesXizang Minzu UniversityXianyangShaanxiChina
| | - Linhao Zhu
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
- School of Basic Medical SciencesXizang Minzu UniversityXianyangShaanxiChina
| | - Jianwen Zheng
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
- Department of NeurologyAffiliated hospital of Xizang Minzu UniversityXianyangShaanxiChina
| | - Dongya Yuan
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
- School of Basic Medical SciencesXizang Minzu UniversityXianyangShaanxiChina
| | - Tianbo Jin
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
- School of Basic Medical SciencesXizang Minzu UniversityXianyangShaanxiChina
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University)Ministry of EducationXi’anShaanxiChina
| |
Collapse
|
32
|
Lettieri-Barbato D, Ioannilli L, Aquilano K, Ciccarone F, Rosina M, Ciriolo MR. FoxO1 localizes to mitochondria of adipose tissue and is affected by nutrient stress. Metabolism 2019; 95:84-92. [PMID: 30974111 DOI: 10.1016/j.metabol.2019.04.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Mitochondria play pivotal roles in orchestrating signaling pathways in order to guarantee metabolic homeostasis under different stimuli. It has been demonstrated that the mito-nuclear communication is fundamental for facing physiological and/or stress-mediated cellular response through the activation of nuclear transcription factors. Here, we focused on the Forkhead box protein O1 (FoxO1) transcription factor that belongs to the FoxOs family proteins and is considered a "nutrients sensor" modulating the expression of nutrient-stress response genes. METHODS In vitro and in vivo experimental systems, including 3T3-L1 white, X-9 beige and T37i brown adipocytes and different fat depots from C57BL/6 mice were used. The mitochondrial localization of FoxO1 was demonstrated by western blot analysis, confocal microscopy and chromatin immunoprecipitation assay, after sub-cellular compartment isolation. RT-qPCR analysis was used to evaluate the expression of antioxidant and mitochondrial genes after modulation of FoxO1 activity/localization. Treatment with diverse reactive oxygen species (ROS) species/sources were performed and assessed by cytofluorimetric analysis. RESULTS We demonstrated that FoxO1 not exclusively localizes to cytosol and nucleus of adipocytes but also to mitochondria where it binds to mitochondrial DNA. We also proved that mitochondrial FoxO1 is phosphorylated upon normal feeding condition. Mitochondrial FoxO1 responds to starvation leaving mitochondrial compartment by ROS-mediated activation of the mitochondrial phosphatase PTPMT1. Indeed, FoxO1 de-phosphorylation and mito-to-nucleus shuttling was observed under starvation. Moreover, we provided evidence that ROS species/sources are able to differently modulate the mitochondrial localization of FoxO1. CONCLUSION The ability to localize at different cell compartments, including mitochondria, highlights a different layer of regulation of FoxO1 necessary for assuring a fast and efficient nutrient-stress response in white/beige adipose tissue. FoxO1 could be thus endorsed in the list of transcription factors involved in the mito-nuclear communication where ROS can act as upstream signals.
Collapse
Affiliation(s)
- Daniele Lettieri-Barbato
- Dept. of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Laura Ioannilli
- Dept. of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Katia Aquilano
- Dept. of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Fabio Ciccarone
- Dept. of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Marco Rosina
- Dept. of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Maria Rosa Ciriolo
- Dept. of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica, 00133 Rome, Italy; IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy.
| |
Collapse
|
33
|
AMPK: A promising molecular target for combating cisplatin toxicities. Biochem Pharmacol 2019; 163:94-100. [DOI: 10.1016/j.bcp.2019.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/05/2019] [Indexed: 02/07/2023]
|
34
|
Niu Y, Lin Z, Wan A, Chen H, Liang H, Sun L, Wang Y, Li X, Xiong XF, Wei B, Wu X, Wan G. RNA N6-methyladenosine demethylase FTO promotes breast tumor progression through inhibiting BNIP3. Mol Cancer 2019; 18:46. [PMID: 30922314 PMCID: PMC6437932 DOI: 10.1186/s12943-019-1004-4] [Citation(s) in RCA: 414] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/13/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) modification is the most pervasive modification in mRNA, and has been considered as a new layer of epigenetic regulation on mRNA processing, stability and translation. Despite its functional significance in various physiological processes, the role of the m6A modification involved in breast cancer is yet fully understood. METHODS We used the m6A-RNA immunoprecipitation sequencing to identify the potential targets in breast cancer. To determine the underlying mechanism for the axis of FTO-BNIP3, we performed a series of in vitro and in vivo assays in 3 breast cancer cell lines and 36 primary breast tumor tissues and 12 adjunct tissues. RESULTS We showed that FTO, a key m6A demethylase, was up-regulated in human breast cancer. High level of FTO was significantly associated with lower survival rates in patients with breast cancer. FTO promoted breast cancer cell proliferation, colony formation and metastasis in vitro and in vivo. We identified BNIP3, a pro-apoptosis gene, as a downstream target of FTO-mediated m6A modification. Epigenetically, FTO mediated m6A demethylation in the 3'UTR of BNIP3 mRNA and induced its degradation via an YTHDF2 independent mechanism. BNIP3 acts as a tumor suppressor and is negatively correlated with FTO expression in clinical breast cancer patients. BNIP3 dramatically alleviated FTO-dependent tumor growth retardation and metastasis. CONCLUSIONS Our findings demonstrate the functional significance of the m6A modification in breast cancer, and suggest that FTO may serve as a novel potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Yi Niu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Ziyou Lin
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Arabella Wan
- The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Honglei Chen
- The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Heng Liang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Lei Sun
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Yuan Wang
- School of Pharmaceutical Sciences, the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xi Li
- The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Xiao-Feng Xiong
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Bo Wei
- The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China.
| | - Xiaobin Wu
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Guohui Wan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China. .,Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| |
Collapse
|
35
|
Li L, Kang H, Zhang Q, D'Agati VD, Al-Awqati Q, Lin F. FoxO3 activation in hypoxic tubules prevents chronic kidney disease. J Clin Invest 2019; 129:2374-2389. [PMID: 30912765 DOI: 10.1172/jci122256] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Acute kidney injury (AKI) can lead to chronic kidney disease (CKD) if injury is severe and/or repair is incomplete. However, the pathogenesis of CKD following renal ischemic injury is not fully understood. Capillary rarefaction and tubular hypoxia are common findings during the AKI to CKD transition. We investigated the tubular stress response to hypoxia and demonstrated that a stress responsive transcription factor, FoxO3, was regulated by prolyl hydroxylase. Hypoxia inhibited FoxO3 prolyl hydroxylation and FoxO3 degradation, thus leading to FoxO3 accumulation and activation in tubular cells. Hypoxia-activated Hif-1α contributed to FoxO3 activation and functioned to protect kidneys, as tubular deletion of Hif-1α decreased hypoxia-induced FoxO3 activation, and resulted in more severe tubular injury and interstitial fibrosis following ischemic injury. Strikingly, tubular deletion of FoxO3 during the AKI to CKD transition aggravated renal structural and functional damage leading to a more profound CKD phenotype. We showed that tubular deletion of FoxO3 resulted in decreased autophagic response and increased oxidative injury, which may explain renal protection by FoxO3. Our study indicates that in the hypoxic kidney, stress responsive transcription factors can be activated for adaptions to counteract hypoxic insults, thus attenuating CKD development.
Collapse
Affiliation(s)
- Ling Li
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Huimin Kang
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA.,Department of Pediatrics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Qing Zhang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Qais Al-Awqati
- Department of Internal Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Fangming Lin
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Work in the past decade has revealed key functions of the evolutionary conserved transcription factors Forkhead box O (FOXO) in the maintenance of homeostatic hematopoiesis. Here the diverse array of FOXO functions in normal and diseased hematopoietic stem and progenitor cells is reviewed and the main findings in the past decade are highlighted. Future work should reveal FOXO-regulated networks whose alterations contribute to hematological disorders. RECENT FINDINGS Recent studies have identified unanticipated FOXO functions in hematopoiesis including in hematopoietic stem and progenitor cells (HSPC), erythroid cells, and immune cells. These findings suggest FOXO3 is critical for the regulation of mitochondrial and metabolic processes in hematopoietic stem cells, the balanced lineage determination, the T and B homeostasis, and terminal erythroblast maturation and red blood cell production. In aggregate these findings highlight the context-dependent function of FOXO in hematopoietic cells. Recent findings also question the nature of FOXO's contribution to heme malignancies as well as the mechanisms underlying FOXO's regulation in HSPC. SUMMARY FOXO are safeguards of homeostatic hematopoiesis. FOXO networks and their regulators and coactivators in HSPC are greatly complex and less well described. Identifications and characterizations of these FOXO networks in disease are likely to uncover disease-promoting mechanisms.
Collapse
|
37
|
Gurska LM, Ames K, Gritsman K. Signaling Pathways in Leukemic Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1143:1-39. [PMID: 31338813 PMCID: PMC7249489 DOI: 10.1007/978-981-13-7342-8_1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hematopoietic stem cells (HSCs) and leukemic stem cells (LSCs) utilize many of the same signaling pathways for their maintenance and survival. In this review, we will focus on several signaling pathways whose roles have been extensively studied in both HSCs and LSCs. Our main focus will be on the PI3K/AKT/mTOR pathway and several of its regulators and downstream effectors. We will also discuss several other signaling pathways of particular importance in LSCs, including the WNT/β-catenin pathway, the NOTCH pathway, and the TGFβ pathway. For each of these pathways, we will emphasize differences in how these pathways operate in LSCs, compared to their function in HSCs, to highlight opportunities for the specific therapeutic targeting of LSCs. We will also highlight areas of crosstalk between multiple signaling pathways that may affect LSC function.
Collapse
Affiliation(s)
- Lindsay M Gurska
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kristina Ames
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kira Gritsman
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA.
- Department of Medical Oncology, Montefiore Hospital, Bronx, New York, USA.
| |
Collapse
|
38
|
Bardaweel SK, Gul M, Alzweiri M, Ishaqat A, ALSalamat HA, Bashatwah RM. Reactive Oxygen Species: the Dual Role in Physiological and Pathological Conditions of the Human Body. Eurasian J Med 2018; 50:193-201. [PMID: 30515042 DOI: 10.5152/eurasianjmed.2018.17397] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Reactive oxygen species (ROS) are well-known for playing a dual role as destructive and constructive species. Indeed, ROS are engaged in many redox-governing activities of the cells for the preservation of cellular homeostasis. However, its overproduction has been reported to result in oxidative stress, which is considered as a deleterious process, and is involved in the damage of cell structures that causes various diseased states. This review provides a concise view on some of the current research published in this topic for an improved understanding of the key roles of ROS in diverse conditions of health and disease. Previous research demonstrated that ROS perform as potential signaling molecules to control several normal physiological functions at the cellular level. Additionally, there is a growing body of evidence supporting the role of ROS in various pathological states. The binary nature of ROS with their profitable and injurious characteristics indicates the complexities of their specific roles at a biological compartment and the difficulties in establishing convenient intervention procedures to treat ROS-related diseases.
Collapse
Affiliation(s)
- Sanaa K Bardaweel
- Department of Pharmaceutical Sciences, University of Jordan School of Pharmacy, Amman, Jordan
| | - Mustafa Gul
- Department of Physiology, Atatürk University School of Medicine, Erzurum, Turkey
| | - Muhammad Alzweiri
- Department of Pharmaceutical Sciences, University of Jordan School of Pharmacy, Amman, Jordan
| | - Aman Ishaqat
- Department of Pharmaceutical Sciences, University of Jordan School of Pharmacy, Amman, Jordan
| | - Husam A ALSalamat
- Department of Pharmaceutical Sciences, University of Jordan School of Pharmacy, Amman, Jordan
| | - Rasha M Bashatwah
- Department of Pharmaceutical Sciences, University of Jordan School of Pharmacy, Amman, Jordan
| |
Collapse
|
39
|
Arjunan P, Meghil MM, Pi W, Xu J, Lang L, El-Awady A, Sullivan W, Rajendran M, Rabelo MS, Wang T, Tawfik OK, Kunde-Ramamoorthy G, Singh N, Muthusamy T, Susin C, Teng Y, Arce RM, Cutler CW. Oral Pathobiont Activates Anti-Apoptotic Pathway, Promoting both Immune Suppression and Oncogenic Cell Proliferation. Sci Rep 2018; 8:16607. [PMID: 30413788 PMCID: PMC6226501 DOI: 10.1038/s41598-018-35126-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/30/2018] [Indexed: 12/25/2022] Open
Abstract
Chronic periodontitis (CP) is a microbial dysbiotic disease linked to increased risk of oral squamous cell carcinomas (OSCCs). To address the underlying mechanisms, mouse and human cell infection models and human biopsy samples were employed. We show that the ‘keystone’ pathogen Porphyromonas gingivalis, disrupts immune surveillance by generating myeloid-derived dendritic suppressor cells (MDDSCs) from monocytes. MDDSCs inhibit CTLs and induce FOXP3 + Tregs through an anti-apoptotic pathway. This pathway, involving pAKT1, pFOXO1, FOXP3, IDO1 and BIM, is activated in humans with CP and in mice orally infected with Mfa1 expressing P. gingivalis strains. Mechanistically, activation of this pathway, demonstrating FOXP3 as a direct FOXO1-target gene, was demonstrated by ChIP-assay in human CP gingiva. Expression of oncogenic but not tumor suppressor markers is consistent with tumor cell proliferation demonstrated in OSCC-P. gingivalis cocultures. Importantly, FimA + P. gingivalis strain MFI invades OSCCs, inducing inflammatory/angiogenic/oncogenic proteins stimulating OSCCs proliferation through CXCR4. Inhibition of CXCR4 abolished Pg-MFI-induced OSCCs proliferation and reduced expression of oncogenic proteins SDF-1/CXCR4, plus pAKT1-pFOXO1. Conclusively, P. gingivalis, through Mfa1 and FimA fimbriae, promotes immunosuppression and oncogenic cell proliferation, respectively, through a two-hit receptor-ligand process involving DC-SIGN+hi/CXCR4+hi, activating a pAKT+hipFOXO1+hiBIM−lowFOXP3+hi and IDO+hi- driven pathway, likely to impact the prognosis of oral cancers in patients with periodontitis.
Collapse
Affiliation(s)
- Pachiappan Arjunan
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America.
| | - Mohamed M Meghil
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America.,Department of Oral Biology, Augusta University, Augusta, Georgia, United States of America
| | - Wenhu Pi
- Department of Radiation Oncology, Indiana University, Indianapolis, Indiana, United States of America
| | - Jinxian Xu
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Liwei Lang
- Department of Oral Biology, Augusta University, Augusta, Georgia, United States of America
| | - Ahmed El-Awady
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - William Sullivan
- Department of Energy, Joint Genome Institute, California, United States of America
| | - Mythilypriya Rajendran
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Mariana Sousa Rabelo
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America.,Department of Periodontics, University of São Paulo, Sao Paulo, Brazil
| | - Tong Wang
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Omnia K Tawfik
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | | | - Nagendra Singh
- Department of Biochemistry & Molecular Biology, Cancer Research Center, Augusta University, Augusta, Georgia, United States of America
| | - Thangaraju Muthusamy
- Department of Biochemistry & Molecular Biology, Cancer Research Center, Augusta University, Augusta, Georgia, United States of America
| | - Cristiano Susin
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Yong Teng
- Department of Oral Biology, Augusta University, Augusta, Georgia, United States of America
| | - Roger M Arce
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Christopher W Cutler
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America.
| |
Collapse
|
40
|
Bach DH, Long NP, Luu TTT, Anh NH, Kwon SW, Lee SK. The Dominant Role of Forkhead Box Proteins in Cancer. Int J Mol Sci 2018; 19:E3279. [PMID: 30360388 PMCID: PMC6213973 DOI: 10.3390/ijms19103279] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 12/16/2022] Open
Abstract
Forkhead box (FOX) proteins are multifaceted transcription factors that are significantly implicated in cancer, with various critical roles in biological processes. Herein, we provide an overview of several key members of the FOXA, FOXC, FOXM1, FOXO and FOXP subfamilies. Important pathophysiological processes of FOX transcription factors at multiple levels in a context-dependent manner are discussed. We also specifically summarize some major aspects of FOX transcription factors in association with cancer research such as drug resistance, tumor growth, genomic alterations or drivers of initiation. Finally, we suggest that targeting FOX proteins may be a potential therapeutic strategy to combat cancer.
Collapse
Affiliation(s)
- Duc-Hiep Bach
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | | | | | - Nguyen Hoang Anh
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Sang Kook Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
41
|
Bednářová A, Tomčala A, Mochanová M, Kodrík D, Krishnan N. Disruption of Adipokinetic Hormone Mediated Energy Homeostasis Has Subtle Effects on Physiology, Behavior and Lipid Status During Aging in Drosophila. Front Physiol 2018; 9:949. [PMID: 30079029 PMCID: PMC6062650 DOI: 10.3389/fphys.2018.00949] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/28/2018] [Indexed: 12/17/2022] Open
Abstract
The impact of disruption of adipokinetic hormone (AKH) signaling was studied during aging in Drosophila in a sexually dimorphic manner. A mutant (Akh1) producing a non-functional AKH peptide was compared with isogenized wild-type controls (w1118), and Akh-rescue line where AKH was ectopically expressed in the mutant background (EE-Akh). Longevity, fecundity, and locomotor activity rhythms remained unaffected by lack of AKH signaling. While the strength of rhythms declined in general with age across all fly lines tested this was more so in case of Akh1 flies. Negative geotaxis was significantly impaired in Akh1 flies. Only young Akh1 flies of both sexes and old Akh1 females showed significantly higher body weight compared to age-matched iso-control flies (except in case of EE-Akh). Expression of genes involved in energy homeostasis and aging indicated that dTOR and Akt expression were elevated in Akh1 flies compared to other genotypes, whereas AMPK and dFoxO expression levels were significantly reduced. Multivariate analysis of the distribution of lipid species revealed a significant accumulation of specific diglyceride (DG) and triglyceride (TG) lipid species, irrespective of sex, attributable in part due to lack of AKH. Moreover, irrespective of lack of AKH, older flies of all genotypes accumulated TGs. Taken together, the results strongly suggest that disruption of AKH has very subtle effects on physiology, behavior and lipid status during aging.
Collapse
Affiliation(s)
- Andrea Bednářová
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, Mississippi State University, Starkville, MS, United States.,Biology Centre, Institute of Entomology, Academy of Sciences, České Budějovice, Czechia
| | - Aleš Tomčala
- Biology Centre, Institute of Parasitology, Academy of Sciences, České Budějovice, Czechia
| | - Michaela Mochanová
- Biology Centre, Institute of Entomology, Academy of Sciences, České Budějovice, Czechia.,Faculty of Science, University of South Bohemia, České Budějovice, Czechia
| | - Dalibor Kodrík
- Biology Centre, Institute of Entomology, Academy of Sciences, České Budějovice, Czechia.,Faculty of Science, University of South Bohemia, České Budějovice, Czechia
| | - Natraj Krishnan
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, Mississippi State University, Starkville, MS, United States
| |
Collapse
|
42
|
Role of Forkhead Box O (FOXO) transcription factor in aging and diseases. Gene 2018; 648:97-105. [PMID: 29428128 DOI: 10.1016/j.gene.2018.01.051] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 12/26/2017] [Accepted: 01/14/2018] [Indexed: 12/21/2022]
Abstract
Fork head box O (FOXO) transcription factor is a key player in an evolutionarily conserved pathway. The mammalian FOXO family consists of FOXO1, 3, 4 and 6, are highly similar in their structure, function and regulation. To maintain optimum body function, the organisms have developed complex mechanisms for homeostasis. Importantly, it is well known that when these mechanisms dysregulate it results in the development of age-related disease. FOXO proteins are involved in a diverse cellular function and also have clinical significance including cell cycle arrest, cell differentiation, tumour suppression, DNA repair, longevity, diabetic complications, immunity, wound healing, regulation of metabolism and thus treatment of several types of diseases. By the combinations of post-translational modifications FOXO's serve as a 'molecular code' to sense external stimuli and recruit it as to specific regions of the genome and provide an integrated cellular response to changing physiological conditions. Akt/Protein kinase B a signaling pathway as a main regulator of FOXO to perform a diverse function in organisms. The present review summarizes the molecular and clinical aspects of FOXO transcription factor. And also elaborate the interaction of FOXO with the nucleosome remodelling complex to target genes, which is essential to cellular homeostasis.
Collapse
|
43
|
Abstract
Stem cells self-renew and differentiate to generate all tissues and cells in the body. Stem cell health promotes adaptive responses to tissue damage or loss and is essential for tissue regeneration with age. In the past decade, the evolutionarily conserved transcription factors FOXO with known functions in promoting healthy aging have emerged as key regulators of stem cell homeostasis in various tissues, including blood, neural, and muscle stem cells. Aberrant FOXO functions have been implicated in a variety of disorders including neurodegenerative, blood, cancer, and diabetes some of which are fostered by abnormal stem cell function. As discussed in this chapter, at least in some stem cells FOXO regulatory mechanisms and applied functions follow a complex set of rules distinct from that operating in progenitor cell populations and in cultured cell lines. Elucidating the exact nature of FOXO properties in stem cells will be critical for identifying and targeting aberrant FOXO-mediated mechanisms that promote stem cell-derived disease specifically with age.
Collapse
Affiliation(s)
- Raymond Liang
- Icahn School of Medicine at Mount Sinai, New York, NY, United States; Developmental and Stem Cell Biology, Multidisciplinary Training Area, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Saghi Ghaffari
- Icahn School of Medicine at Mount Sinai, New York, NY, United States; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
44
|
Delpoux A, Michelini RH, Verma S, Lai CY, Omilusik KD, Utzschneider DT, Redwood AJ, Goldrath AW, Benedict CA, Hedrick SM. Continuous activity of Foxo1 is required to prevent anergy and maintain the memory state of CD8 + T cells. J Exp Med 2017; 215:575-594. [PMID: 29282254 PMCID: PMC5789410 DOI: 10.1084/jem.20170697] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 10/18/2017] [Accepted: 12/06/2017] [Indexed: 12/11/2022] Open
Abstract
Delpoux et al. show, in a model of latent infection, how FOXO1 is required to prevent apoptosis, the acquisition of an anergy phenotype, and to be constantly expressed for maintaining the differentiation state of CD8+ T cells. Upon infection with an intracellular pathogen, cytotoxic CD8+ T cells develop diverse differentiation states characterized by function, localization, longevity, and the capacity for self-renewal. The program of differentiation is determined, in part, by FOXO1, a transcription factor known to integrate extrinsic input in order to specify survival, DNA repair, self-renewal, and proliferation. At issue is whether the state of T cell differentiation is specified by initial conditions of activation or is actively maintained. To study the spectrum of T cell differentiation, we have analyzed an infection with mouse cytomegalovirus, a persistent-latent virus that elicits different cytotoxic T cell responses characterized as acute resolving or inflationary. Our results show that FOXO1 is continuously required for all the phenotypic characteristics of memory-effector T cells such that with acute inactivation of the gene encoding FOXO1, T cells revert to a short-lived effector phenotype, exhibit reduced viability, and manifest characteristics of anergy.
Collapse
Affiliation(s)
- Arnaud Delpoux
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA.,Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | - Rodrigo Hess Michelini
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA.,Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | - Shilpi Verma
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Chen-Yen Lai
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA.,Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | - Kyla D Omilusik
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA
| | - Daniel T Utzschneider
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA.,Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | - Alec J Redwood
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - Ananda W Goldrath
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA
| | - Chris A Benedict
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Stephen M Hedrick
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA .,Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| |
Collapse
|
45
|
Kan H, Huang Y, Li X, Liu D, Chen J, Shu M. Zinc finger protein ZBTB20 is an independent prognostic marker and promotes tumor growth of human hepatocellular carcinoma by repressing FoxO1. Oncotarget 2017; 7:14336-49. [PMID: 26893361 PMCID: PMC4924719 DOI: 10.18632/oncotarget.7425] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 01/29/2016] [Indexed: 12/17/2022] Open
Abstract
Zinc finger and BTB domain-containing 20 (ZBTB20) is a new BTB/POZ-domain gene and a member of the POK family of transcriptional repressors. Notably, the role of ZBTB20 and its underlying mechanisms involved in hepatocarcinogenesis are poorly investigated. In this study, the expression of ZBTB20 was significantly overexpressed in hepatocellular carcinoma (HCC) tissues. The positive expression of ZBTB20 was associated with large tumor size, high Edmondson-Steiner grading and advanced tumor-node-metastasis (TNM) tumor stage. Additionally, HCC patients with positive expression of ZBTB20 had a poorer 5-year survival. Multivariate analyses revealed that ZBTB20 overexpression was an independent prognostic factor for HCC. Gain- and loss-of-function experiments demonstrated that ZBTB20 promoted HCC cell viability, proliferation, tumorigenicity, and cell cycle progression. Mechanistically, Cyclin D1 and Cyclin E were increased, while p21 and p27 were decreased by ZBTB20 in HCC cells. FoxO1 was inversely correlated with ZBTB20 protein expression in the same cohort of HCC specimens. We further revealed that FoxO1 was transcriptionally repressed by ZBTB20 in HCC. Moreover, restoration of FoxO1 expression partially abrogated ZBTB20-induced HCC cell proliferation and growth entry in vitro and in vivo. Collectively, these results indicate that ZBTB20 may serve as a prognostic marker and promotes tumor growth of HCC via transcriptionally repressing FoxO1.
Collapse
Affiliation(s)
- Heping Kan
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuqi Huang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xianghong Li
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Dingli Liu
- Department of Infectious Disease, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jianjia Chen
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Miaojiang Shu
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
46
|
Guo H, Ding E, Bai Y, Zhang H, Shen H, Wang J, Song X, Cai W, Guo J, Zhu B. Association of genetic variations in FOXO3 gene with susceptibility to noise induced hearing loss in a Chinese population. PLoS One 2017; 12:e0189186. [PMID: 29220389 PMCID: PMC5722378 DOI: 10.1371/journal.pone.0189186] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 11/21/2017] [Indexed: 11/18/2022] Open
Abstract
Noise induced hearing loss (NIHL), a multifactorial disease involving both genetic and environmental factors, is one of the most important occupational health hazards. Nonetheless, the influence of FOXO3 variants on NIHL risk have not been illuminated. This research was conducted to explore the effects of FOXO3 polymorphisms on individual susceptibility to NIHL. A total of 2689 industrial workers from one textile factory of east China were recruited to participate in the current research. Venous blood was collected, questionnaire and pure-tone audiometry (PTA) was conducted by specialist physicians. Then, we performed genotyping of three selected SNPs (rs2802292, rs10457180, and rs12206094) in FOXO3 gene in 566 NIHL patients and 566 controls. Subsequently, the main effects of genotype and its interactions were evaluated. Our results revealed that individuals with the G allele of rs2802292, G allele of rs10457180, T allele of rs12206094 (OR = 1.43, 1.43, and 1.31 respectively) and the haplotype GAC and others (TGT/GGT/GGC/GAT) (rs2802292-rs10457180-rs12206094) (OR = 1.49 and 2.09 respectively) are associated with an increased risk of NIHL in a Chinese population. Stratified analysis showed that an increased NIHL risk was found in the subjects who exposed to noise >16 years with rs2802292 GG/GT and rs10457180 AG/GG genotype with an OR of 1.62 and 1.66 respectively. Multifactor dimensionality reduction analysis indicated that rs10457180, rs2802292, and rs12206094 have interactions and are related to increased NIHL risk (OR = 1.53). The genetic polymorphism rs2802292, rs10457180, and rs12206094 within FOXO3 gene are associated with an increased risk of NIHL in a Chinese population and have potential to be biomarkers for noise exposed workers.
Collapse
Affiliation(s)
- Haoran Guo
- School of Public Health, Southeast University, Nanjing, Jiangsu Province, China
- Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, Jiangsu Province, China
| | - Enmin Ding
- Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, Jiangsu Province, China
| | - Ying Bai
- Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, Jiangsu Province, China
| | - Hengdong Zhang
- Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, Jiangsu Province, China
| | - Huanxi Shen
- Kunshan Centers for Disease Prevention and Control, Kunshan, Jiangsu Province, China
| | - Jun Wang
- School of Public Health, Southeast University, Nanjing, Jiangsu Province, China
| | - Xianping Song
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Wenyan Cai
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jiadi Guo
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Baoli Zhu
- School of Public Health, Southeast University, Nanjing, Jiangsu Province, China
- Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, Jiangsu Province, China
- * E-mail:
| |
Collapse
|
47
|
Doherty A, Kernogitski Y, Kulminski AM, Pedro de Magalhães J. Identification of polymorphisms in cancer patients that differentially affect survival with age. Aging (Albany NY) 2017; 9:2117-2136. [PMID: 29064820 PMCID: PMC5680559 DOI: 10.18632/aging.101305] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 10/06/2017] [Indexed: 01/08/2023]
Abstract
The World Health Organization predicts that the proportion of the world's population over 60 will almost double from 12% to 22% between 2015 and 2050. Ageing is the biggest risk factor for cancer, which is a leading cause of deaths worldwide. Unfortunately, research describing how genetic variants affect cancer progression commonly neglects to account for the ageing process. Herein is the first systematic analysis that combines a large longitudinal data set with a targeted candidate gene approach to examine the effect of genetic variation on survival as a function of age in cancer patients. Survival was significantly decreased in individuals with heterozygote or rare homozygote (i.e. variant) genotypes compared to those with a common homozygote genotype (i.e. wild type) for two single nucleotide polymorphisms (rs11574358 and rs4147918), one gene (SIRT3) and one pathway (FoxO signalling) in an age-dependent manner. All identified genes and pathways have previously been associated with ageing and cancer. These observations demonstrate that there are ageing-related genetic elements that differentially affect mortality in cancer patients in an age-dependent manner. Understanding the genetic determinants affecting prognosis differently with age will be invaluable to develop age-specific prognostic biomarkers and personalized therapies that may improve clinical outcomes for older individuals.
Collapse
Affiliation(s)
- Aoife Doherty
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, United Kingdom
| | - Yelena Kernogitski
- Biodemography of Aging Research Unit (BARU), Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit (BARU), Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, United Kingdom
| |
Collapse
|
48
|
Jiang J, Huang Z, Chen X, Luo R, Cai H, Wang H, Zhang H, Sun T, Zhang Y. Trifluoperazine Activates FOXO1-Related Signals to Inhibit Tumor Growth in Hepatocellular Carcinoma. DNA Cell Biol 2017; 36:813-821. [PMID: 28876084 DOI: 10.1089/dna.2017.3790] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Schizophrenic patients tend to have reduced incidence of some cancers due to the treatment of antipsychotic drugs with antitumor effects, such as chlorpromazine and trifluoperazine (TFP). Forkhead Box O1 (FOXO1) as tumor suppressor in many malignancies is often inactivated by nuclear export, which could be inhibited by TFP. However, the antitumor efficiency of TFP and related role of FOXO1 in hepatocellular carcinoma (HCC) are unclear. Thus, two HCC cell lines SMMC-7721 and Bel-7402 were treated with different concentrations of TFP and the IC50 was determined. We found that TFP could inhibit the vitality of two cell lines and induce cell cycle arrest at G0/G1. Meanwhile, the apoptosis was also increased and the ability of migration or invasion was found to be impaired by TFP. Interestingly, TFP reversed the cytoplasmic localization of FOXO1 to nuclear and increased its expression in nuclear, and increased the ratio of Bax/Bcl-2. However, knockdown of FOXO1 significantly abrogated the TFP-induced
apoptosis by decreasing the Bcl-2 expression [corrected]. Furthermore, we found that TFP in vivo could effectively restrict the angiogenesis and tumor growth with reduced expression of VEGF, Bcl-2, and PCNA, and increased the nuclear localization of FOXO1, which indicated its antitumor role in HCC.
Collapse
Affiliation(s)
- Jingwen Jiang
- 1 Department of Medical Oncology, Hainan Province Hospital of Traditional Chinese Medicine , Haikou, China
| | - Zhongxi Huang
- 2 Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical Sciences, Southern Medical University , Guangzhou, China
| | - Xuewu Chen
- 1 Department of Medical Oncology, Hainan Province Hospital of Traditional Chinese Medicine , Haikou, China
| | - Rongcheng Luo
- 3 TCM-Integrated Cancer Center, Southern Medical University , Guangzhou, China
| | - Hongbin Cai
- 4 Department of Medical Oncology, TCM-Integrated Cancer Center of Southern Medical University , Guangzhou, China
| | - Hairu Wang
- 5 Department of Surgical Oncology, Haikou People's Hospital , Haikou, China
| | - Hui Zhang
- 1 Department of Medical Oncology, Hainan Province Hospital of Traditional Chinese Medicine , Haikou, China
| | - Tao Sun
- 6 College of Traditional Chinese Medicine, Hainan Medical University , Haikou, Hainan, China
| | - Yunfang Zhang
- 1 Department of Medical Oncology, Hainan Province Hospital of Traditional Chinese Medicine , Haikou, China
| |
Collapse
|
49
|
Guastafierro T, Bacalini MG, Marcoccia A, Gentilini D, Pisoni S, Di Blasio AM, Corsi A, Franceschi C, Raimondo D, Spanò A, Garagnani P, Bondanini F. Genome-wide DNA methylation analysis in blood cells from patients with Werner syndrome. Clin Epigenetics 2017; 9:92. [PMID: 28861129 PMCID: PMC5577832 DOI: 10.1186/s13148-017-0389-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/15/2017] [Indexed: 12/15/2022] Open
Abstract
Background Werner syndrome is a progeroid disorder characterized by premature age-related phenotypes. Although it is well established that autosomal recessive mutations in the WRN gene is responsible for Werner syndrome, the molecular alterations that lead to disease phenotype remain still unidentified. Results To address whether epigenetic changes can be associated with Werner syndrome phenotype, we analysed genome-wide DNA methylation profile using the Infinium MethylationEPIC BeadChip in the whole blood from three patients affected by Werner syndrome compared with three age- and sex-matched healthy controls. Hypermethylated probes were enriched in glycosphingolipid biosynthesis, FoxO signalling and insulin signalling pathways, while hypomethylated probes were enriched in PI3K-Akt signalling and focal adhesion pathways. Twenty-two out of 47 of the differentially methylated genes belonging to the enriched pathways resulted differentially expressed in a publicly available dataset on Werner syndrome fibroblasts. Interestingly, differentially methylated regions identified CERS1 and CERS3, two members of the ceramide synthase family. Moreover, we found differentially methylated probes within ITGA9 and ADAM12 genes, whose methylation is altered in systemic sclerosis, and within the PRDM8 gene, whose methylation is affected in dyskeratosis congenita and Down syndrome. Conclusions DNA methylation changes in the peripheral blood from Werner syndrome patients provide new insight in the pathogenesis of the disease, highlighting in some cases a functional correlation of gene expression and methylation status. Electronic supplementary material The online version of this article (doi:10.1186/s13148-017-0389-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- T Guastafierro
- UOC of Clinical Biochemistry, Sandro Pertini Hospital, Rome, Italy.,CRIIS (Interdisciplinary, Interdepartmental and Specialistic Reference Center for Early Diagnosis of Scleroderma, Treatment of Sclerodermic Ulcers and Videocapillaroscopy), Sandro Pertini Hospital, Rome, Italy
| | - M G Bacalini
- IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - A Marcoccia
- CRIIS (Interdisciplinary, Interdepartmental and Specialistic Reference Center for Early Diagnosis of Scleroderma, Treatment of Sclerodermic Ulcers and Videocapillaroscopy), Sandro Pertini Hospital, Rome, Italy.,UOSD Ischemic Microangiopathy and Sclerodermic Ulcers, Sandro Pertini Hospital, Rome, Italy
| | - D Gentilini
- Centre for Biomedical Research and Technologies, Italian Auxologic Institute, IRCCS, Milan, Italy
| | - S Pisoni
- Centre for Biomedical Research and Technologies, Italian Auxologic Institute, IRCCS, Milan, Italy
| | - A M Di Blasio
- Centre for Biomedical Research and Technologies, Italian Auxologic Institute, IRCCS, Milan, Italy
| | - A Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - C Franceschi
- IRCCS Institute of Neurological Sciences, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,Interdepartmental Center "L. Galvani", University of Bologna, Bologna, Italy
| | - D Raimondo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - A Spanò
- UOC of Clinical Biochemistry, Sandro Pertini Hospital, Rome, Italy
| | - P Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,Interdepartmental Center "L. Galvani", University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), St. Orsola-Malpighi University Hospital, Bologna, Italy.,Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute at Huddinge University Hospital, S-141 86 Stockholm, Sweden.,CNR Institute for Molecular Genetics, Unit of Bologna, Bologna, Italy.,Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute, Bologna, Italy
| | - F Bondanini
- CRIIS (Interdisciplinary, Interdepartmental and Specialistic Reference Center for Early Diagnosis of Scleroderma, Treatment of Sclerodermic Ulcers and Videocapillaroscopy), Sandro Pertini Hospital, Rome, Italy.,UOC of Clinical Pathology, Saint' Eugenio Hospital, Rome, Italy
| |
Collapse
|
50
|
Wang L, Ni Z, Liu Y, Ji S, Jin F, Jiang K, Ma J, Ren C, Zhang H, Hu Z, Zha X. Hyperactivated mTORC1 downregulation of FOXO3a/PDGFRα/AKT cascade restrains tuberous sclerosis complex-associated tumor development. Oncotarget 2017; 8:54858-54872. [PMID: 28903387 PMCID: PMC5589626 DOI: 10.18632/oncotarget.18963] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/18/2017] [Indexed: 12/14/2022] Open
Abstract
Hyperactivation of mammalian target of rapamycin complex 1 (mTORC1), caused by loss-of-function mutations in either the TSC1 or TSC2 gene, leads to the development of tuberous sclerosis complex (TSC), a benign tumor syndrome with multiple affected organs. mTORC1-mediated inhibition of AKT constrains the tumor progression of TSC, but the exact mechanisms remain unclear. Herein we showed that loss of TSC1 or TSC2 downregulation of platelet-derived growth factor receptor α (PDGFRα) expression was mediated by mTORC1. Moreover, mTORC1 inhibited PDGFRα expression via suppression of forkhead box O3a (FOXO3a)-mediated PDGFRα gene transcription. In addition, ectopic expression of PDGFRα promoted AKT activation and enhanced proliferation and tumorigenic capacity of Tsc1- or Tsc2-null mouse embryonic fibroblasts (MEFs), and vice versa. Most importantly, rapamycin in combination with AG1295, a PDGFR inhibitor, significantly inhibited growth of TSC1/TSC2 complex-deficient cells in vitro and in vivo. Therefore, downregulated FOXO3a/PDGFRα/AKT pathway exerts a protective effect against hyperactivated mTORC1-induced tumorigenesis caused by loss of TSC1/TSC2 complex, and the combination of rapamycin and AG1295 may be a new effective strategy for TSC-associated tumors treatment.
Collapse
Affiliation(s)
- Li Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Zhaofei Ni
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Yujie Liu
- The First Clinical Medical School, Anhui Medical University, Hefei, China
| | - Shuang Ji
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Fuquan Jin
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Keguo Jiang
- Department of Nephrology, The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Junfang Ma
- Department of Neurology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Cuiping Ren
- Department of Parasitology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Hongbing Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaojun Zha
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|