1
|
Moro M, Balestrero FC, Colombo G, Torretta S, Clemente N, Ciccone V, Del Grosso E, Donnini S, Travelli C, Condorelli F, Sangaletti S, Genazzani AA, Grolla AA. Extracellular nicotinamide phosphoribosyltransferase (eNAMPT) drives abnormal pericyte-rich vasculature in triple-negative breast cancer. Angiogenesis 2024; 28:4. [PMID: 39636369 DOI: 10.1007/s10456-024-09956-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/11/2024] [Indexed: 12/07/2024]
Abstract
Tumour angiogenesis supports malignant cells with oxygen and nutrients to promote invasion and metastasis. A number of cytokines released in situ participate in the recruitment of endothelial cells and pericytes to trigger the formation of novel blood vessels, which are often abnormal, leaky, and disorganized. Nicotinamide phosphoribosyltransferase is a key intracellular enzyme involved in NAD metabolism and is up regulated in many cancers to meet bioenergetic demands. Yet, the same protein is also secreted extracellularly (eNAMPT), where it acts as a pro-inflammatory cytokine. High plasma eNAMPT levels have been reported in breast cancer patients and correlate with aggressiveness and prognosis. We now report that in a triple-negative breast cancer model, enriching the tumour microenvironment with eNAMPT leads to abundant angiogenesis and increased metastatization. Atypically, the eNAMPT-mediated pro-angiogenic effect is mainly directed to NG2+ pericytes. Indeed, eNAMPT acts as chemoattractant for pericytes and coordinates vessel-like tube formation, in synergism with the classical factor PDGF-BB. Stimulation of pericytes by eNAMPT leads to a pro-inflammatory activation, characterized by the overexpression of key chemokines (CXCL8, CXCL1, CCL2) and VCAM1, via NF-κB signalling. All these effects were ablated by the use of C269, an anti-eNAMPT neutralizing antibody, suggesting that this might represent a novel anti-angiogenic pharmacological approach for triple-negative breast cancer.
Collapse
Affiliation(s)
- Marianna Moro
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | | | - Giorgia Colombo
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
- Division of Hematology/Oncology Department of Medicine, Weill Cornell Medicine, Cornell University, New York, USA
| | - Simone Torretta
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Nausicaa Clemente
- Department of Health Science, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
| | - Valerio Ciccone
- Department of Life Science, Università di Siena, Siena, Italy
| | - Erika Del Grosso
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Sandra Donnini
- Department of Life Science, Università di Siena, Siena, Italy
| | - Cristina Travelli
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
- Department of Drug Sciences, Università degli Studi di Pavia, Pavia, Italy
| | - Fabrizio Condorelli
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Sabina Sangaletti
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy.
- Department of Drug Science and Technology, Università di Torino, Turin, Italy.
| | - Ambra A Grolla
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
2
|
Ok CY, Kwon RJ, Jang HO, Bae MK, Bae SK. Visfatin Enhances RANKL-Induced Osteoclastogenesis In Vitro: Synergistic Interactions and Its Role as a Mediator in Osteoclast Differentiation and Activation. Biomolecules 2024; 14:1500. [PMID: 39766208 PMCID: PMC11673010 DOI: 10.3390/biom14121500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Visfatin, an adipokine secreted by various cell types, plays multifaceted pathophysiological roles in inflammatory conditions, including obesity, which is closely associated with osteoclastogenesis, a key process underlying bone loss and increased osteoporosis (OP) risk. However, the role of visfatin in osteoclastogenesis remains controversial. This study was conducted to investigate the effects of visfatin on receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast differentiation from precursor cells in vitro. Our results demonstrated that although visfatin exhibited a modest osteoclast-inductive effect relative to that of RANKL, co-stimulation of bone marrow-derived macrophages (BMDMs) with visfatin and RANKL led to significantly enhanced osteoclast differentiation and activation compared to individual stimulation. Neutralization of visfatin activity using blocking antibodies before differentiation markedly suppressed RANKL-induced osteoclastogenesis, as evidenced by a near-complete absence of tartrate-resistant acid phosphatase-positive multinucleated osteoclasts, decreased levels of nuclear factor of activated T cells cytoplasmic 1 and osteoclast-specific proteins, inhibition of nuclear factor-κB and mitogen-activated protein kinase signaling pathways, and a decrease in resorption pit formation. Our findings underscore the critical role of visfatin in RANKL-induced osteoclastogenesis in vitro and highlight the RANKL/visfatin signaling axis as a potential therapeutic target for destructive bone loss-related diseases.
Collapse
Affiliation(s)
- Chang Youp Ok
- Department of Dental Pharmacology, School of Dentistry, Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; (C.Y.O.); (H.-O.J.)
- Periodontal Disease Signaling Network Research Center, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ryuk Jun Kwon
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea;
| | - Hye-Ock Jang
- Department of Dental Pharmacology, School of Dentistry, Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; (C.Y.O.); (H.-O.J.)
| | - Moon-Kyoung Bae
- Department of Oral Physiology, School of Dentistry, Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Soo-Kyung Bae
- Department of Dental Pharmacology, School of Dentistry, Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; (C.Y.O.); (H.-O.J.)
- Periodontal Disease Signaling Network Research Center, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
3
|
Kakkat S, Suman P, Turbat- Herrera EA, Singh S, Chakroborty D, Sarkar C. Exploring the multifaceted role of obesity in breast cancer progression. Front Cell Dev Biol 2024; 12:1408844. [PMID: 39040042 PMCID: PMC11260727 DOI: 10.3389/fcell.2024.1408844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Obesity is a multifaceted metabolic disorder characterized by excessive accumulation of adipose tissue. It is a well-established risk factor for the development and progression of breast cancer. Adipose tissue, which was once regarded solely as a passive energy storage depot, is now acknowledged as an active endocrine organ producing a plethora of bioactive molecules known as adipokines that contribute to the elevation of proinflammatory cytokines and estrogen production due to enhanced aromatase activity. In the context of breast cancer, the crosstalk between adipocytes and cancer cells within the adipose microenvironment exerts profound effects on tumor initiation, progression, and therapeutic resistance. Moreover, adipocytes can engage in direct interactions with breast cancer cells through physical contact and paracrine signaling, thereby facilitating cancer cell survival and invasion. This review endeavors to summarize the current understanding of the intricate interplay between adipocyte-associated factors and breast cancer progression. Furthermore, by discussing the different aspects of breast cancer that can be adversely affected by obesity, this review aims to shed light on potential avenues for new and novel therapeutic interventions.
Collapse
Affiliation(s)
- Sooraj Kakkat
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
| | - Prabhat Suman
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
| | - Elba A. Turbat- Herrera
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
| | - Seema Singh
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, United States
| | - Debanjan Chakroborty
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, United States
| | - Chandrani Sarkar
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
4
|
Semerena E, Nencioni A, Masternak K. Extracellular nicotinamide phosphoribosyltransferase: role in disease pathophysiology and as a biomarker. Front Immunol 2023; 14:1268756. [PMID: 37915565 PMCID: PMC10616597 DOI: 10.3389/fimmu.2023.1268756] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/03/2023] [Indexed: 11/03/2023] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) plays a central role in mammalian cell metabolism by contributing to nicotinamide adenine dinucleotide biosynthesis. However, NAMPT activity is not limited to the intracellular compartment, as once secreted, the protein accomplishes diverse functions in the extracellular space. Extracellular NAMPT (eNAMPT, also called visfatin or pre-B-cell colony enhancing factor) has been shown to possess adipocytokine, pro-inflammatory, and pro-angiogenic activities. Numerous studies have reported the association between elevated levels of circulating eNAMPT and various inflammatory and metabolic disorders such as obesity, diabetes, atherosclerosis, arthritis, inflammatory bowel disease, lung injury and cancer. In this review, we summarize the current state of knowledge on eNAMPT biology, proposed roles in disease pathogenesis, and its potential as a disease biomarker. We also briefly discuss the emerging therapeutic approaches for eNAMPT inhibition.
Collapse
Affiliation(s)
- Elise Semerena
- Light Chain Bioscience - Novimmune SA, Plan-les-Ouates, Switzerland
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | | |
Collapse
|
5
|
Travelli C, Colombo G, Aliotta M, Fagiani F, Fava N, De Sanctis R, Grolla AA, Garcia JGN, Clemente N, Portararo P, Costanza M, Condorelli F, Colombo MP, Sangaletti S, Genazzani AA. Extracellular nicotinamide phosphoribosyltransferase (eNAMPT) neutralization counteracts T cell immune evasion in breast cancer. J Immunother Cancer 2023; 11:e007010. [PMID: 37880182 PMCID: PMC10603332 DOI: 10.1136/jitc-2023-007010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Nicotinamide phosphoribosyltransferase (NAMPT) is a key intracellular enzyme that participates in nicotinamide adenine dinucleotide (NAD) homeostasis as well as a released cytokine (eNAMPT) that is elevated in inflammatory conditions and in cancer. In patients with breast cancer, circulating eNAMPT is elevated and its plasma levels correlate with prognosis and staging. In light of this, we investigated the contribution of eNAMPT in triple negative mammary carcinoma progression by investigating the effect of its neutralization via a specific neutralizing monoclonal antibody (C269). METHODS We used female BALB/c mice injected with 4T1 clone 5 cells and female C57BL6 injected with EO771 cells, evaluating tumoral size, spleen weight and number of metastases. We injected two times a week the anti-eNAMPT neutralizing antibody and we sacrificed the mice after 28 days. Harvested tumors were analyzed by histopathology, flow cytometry, western blot, immunohistochemistry, immunofluorescence and RNA sequencing to define tumor characteristics (isolating tumor infiltrating lymphocytes and tumoral cells) and to investigate the molecular mechanisms behind the observed phenotype. Moreover, we dissected the functional relationship between T cells and tumoral cells using three-dimensional (3D) co-cultures. RESULTS The neutralization of eNAMPT with C269 led to decreased tumor size and reduced number of lung metastases. RNA sequencing and functional assays showed that eNAMPT controlled T-cell response via the programmed death-ligand 1/programmed cell death protein 1 (PD-L1/PD-1) axis and its neutralization led to a restoration of antitumoral immune responses. In particular, eNAMPT neutralization was able to activate CD8+IFNγ+GrzB+ T cells, reducing the immunosuppressive phenotype of T regulatory cells. CONCLUSIONS These studies indicate for the first time eNAMPT as a novel immunotherapeutic target for triple negative breast cancer.
Collapse
Affiliation(s)
- Cristina Travelli
- Department of Drug Sciences, Università degli Studi di Pavia, Pavia, Italy
| | - Giorgia Colombo
- Department of Pharmaceutical Science, University of Eastern Piedmont, Novara, Italy
| | - Martina Aliotta
- Department of Drug Sciences, Università degli Studi di Pavia, Pavia, Italy
| | - Francesca Fagiani
- Department of Drug Sciences, Università degli Studi di Pavia, Pavia, Italy
| | - Natalia Fava
- Department of Drug Sciences, Università degli Studi di Pavia, Pavia, Italy
| | - Rita De Sanctis
- Department of Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Ambra A Grolla
- Department of Pharmaceutical Science, University of Eastern Piedmont, Novara, Italy
| | - Joe G N Garcia
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Nausicaa Clemente
- Dipartimento di Scienze della Salute, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
| | - Paola Portararo
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Massimo Costanza
- Department of Clinical Neuroscience, Istituto Nazionale Neurologico Carlo Besta, Milan, Italy
| | - Fabrizio Condorelli
- Department of Pharmaceutical Science, University of Eastern Piedmont, Novara, Italy
| | - Mario Paolo Colombo
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Sabina Sangaletti
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Science, University of Eastern Piedmont, Novara, Italy
| |
Collapse
|
6
|
Huang CL, Achudhan D, Liu PI, Lin YY, Liu SC, Guo JH, Liu CL, Wu CY, Wang SW, Tang CH. Visfatin upregulates VEGF-C expression and lymphangiogenesis in esophageal cancer by activating MEK1/2-ERK and NF-κB signaling. Aging (Albany NY) 2023; 15:204762. [PMID: 37286356 DOI: 10.18632/aging.204762] [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: 03/23/2023] [Accepted: 05/17/2023] [Indexed: 06/09/2023]
Abstract
Lymph node metastasis is a recognized prognostic factor in esophageal cancer. Adipokines, including visfatin, and the molecule vascular endothelial growth factor (VEGF)-C, are implicated in lymphangiogenesis, but whether any association exists between esophageal cancer, adipokines and VEGF-C is unknown. We examined the relevance of adipokines and VEGF-C in esophageal squamous cell carcinoma (ESCC) in the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. We found significantly higher levels of visfatin and VEGF-C expression in esophageal cancer tissue than in normal tissue. Immunohistochemistry (IHC) staining identified that higher levels of visfatin and VEGF-C expression were correlated with advanced stage ESCC. Visfatin treatment of ESCC cell lines upregulated VEGF-C expression and VEGF-C-dependent lymphangiogenesis in lymphatic endothelial cells. Visfatin induced increases in VEGF-C expression by activating the mitogen-activated protein kinase kinases1/2-extracellular signal-regulated kinase (MEK1/2-ERK) and Nuclear Factor Kappa B (NF-κB) signaling cascades. Transfecting ESCC cells with MEK1/2-ERK and NF-κB inhibitors (PD98059, FR180204, PDTC, and TPCK) and siRNAs inhibited visfatin-induced increases in VEGF-C expression. It appears that visfatin and VEGF-C are promising therapeutic targets in the inhibition of lymphangiogenesis in esophageal cancer.
Collapse
Affiliation(s)
- Chang-Lun Huang
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Surgery, Division of Thoracic Surgery, Changhua Christian Hospital, Changhua 500, Taiwan
| | - David Achudhan
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Po-I Liu
- Department of General Thoracic Surgery, Asia University Hospital, Taichung 41354, Taiwan
- Department of Physical Therapy, Asia University, Taichung 41354, Taiwan
| | - Yen-You Lin
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Shan-Chi Liu
- Department of Medical Education and Research, China Medical University Beigang Hospital, Yunlin 65152, Taiwan
| | - Jeng-Hung Guo
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Neurosurgery, China Medical University Hospital, Taichung 404327, Taiwan
| | - Chun-Lin Liu
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Neurosurgery, China Medical University Hospital, Taichung 404327, Taiwan
| | - Chih-Ying Wu
- Department of Neurosurgery, China Medical University Hospital, Taichung 404327, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 406040, Taiwan
- Department of Neurosurgery, China Medical University Hsinchu Hospital, Hsinchu 302, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei 252, Taiwan
- College of Pharmacy, Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 406040, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Health Science, Asia University, Taichung 41354, Taiwan
- Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu 302, Taiwan
| |
Collapse
|
7
|
Rajput PK, Varghese JF, Srivastava AK, Kumar U, Yadav UCS. Visfatin-induced upregulation of lipogenesis via EGFR/AKT/GSK3β pathway promotes breast cancer cell growth. Cell Signal 2023; 107:110686. [PMID: 37084841 DOI: 10.1016/j.cellsig.2023.110686] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 03/27/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023]
Abstract
Breast cancer (BC) incidence and associated mortality have increased in tandem with the growth in obesity among the females worldwide. An adipokine, visfatin, has been shown to potentially impact glucose, lipid, and protein metabolism, and promote cancer growth however, the mechanism underlying the effect of visfatin on lipid metabolism dysregulation contributing to BC cell survival, proliferation, and metastasis has not been elucidated. Herein, we have investigated the role of visfatin on the induction of Sterol regulatory element binding protein (SREBP-1) and its upstream and downstream mediators in MCF-7 breast cancer cells. The survival and proliferation was investigated using MTT and Trypan blue assays, cytosolic lipid accumulation was observed using Nile red staining, mRNA and protein expressions were examined using RT-qPCR and western blotting, respectively, and cell cycle analysis was performed using fluorescence-activated cell sorting. Our results indicate that visfatin increased the survival and proliferation of MCF-7 cells in a time- and dose-dependent manner and augmented lipid buildup via activation of SREBP-1 and its associated downstream lipid synthesizing enzymes, at both mRNA and protein levels in MCF-7 cells. Inhibiting SREBP-1 using fatostatin or silencing with siRNA abrogated excessive lipid deposition by suppressing the expression of genes related to lipid synthesis pathway. Further, in-silico study showed high affinity binding of visfatin with epidermal growth factor receptor (EGFR), which was confirmed in an in-vitro study where visfatin increased the phosphorylation of EGFR at tyrosine residue and activated its downstream proteins via phosphorylation of AKT and GSK3β in MCF-7 cells. Inhibition of GSK3β by phosphorylation led to increased activity of SREBP-1 and associated downstream proteins. In summary, SREBP-1 may be a critical player in visfatin-induced lipid synthesis and accumulation in BC cells via activation of EGFR/AKT/GSK3β pathway leading to increased cell survival and proliferation of BC cells.
Collapse
Affiliation(s)
- Pradeep Kumar Rajput
- Metabolic Disorders and Inflammatory Pathologies Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Johnna Francis Varghese
- Metabolic Disorders and Inflammatory Pathologies Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India; Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Amit Kumar Srivastava
- School of Nano Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Umesh Kumar
- School of Nano Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Umesh C S Yadav
- Special Centre for Molecular Medicine and Special Centre for Systems Medicine (Concurrent Faculty), Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
8
|
Wu Y, Li X, Li Q, Cheng C, Zheng L. Adipose tissue-to-breast cancer crosstalk: Comprehensive insights. Biochim Biophys Acta Rev Cancer 2022; 1877:188800. [PMID: 36103907 DOI: 10.1016/j.bbcan.2022.188800] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
The review focuses on mechanistic evidence for the link between obesity and breast cancer. According to the IARC study, there is sufficient evidence that obesity is closely related to a variety of cancers. Among them, breast cancer is particularly disturbed by adipose tissue due to the unique histological structure of the breast. The review introduces the relationship between obesity and breast cancer from two aspects, including factors that promote tumorigenesis or metastasis. We summarize alterations in adipokines and metabolic pathways that contribute to breast cancer development. Breast cancer metastasis is closely related to obesity-induced pro-inflammatory microenvironment, adipose stem cells, and miRNAs. Based on the mechanism by which obesity causes breast cancer, we list possible therapeutic directions, including reducing the risk of breast cancer and inhibiting the progression of breast cancer. We also discussed the risk of autologous breast remodeling and fat transplantation. Finally, the causes of the obesity paradox and the function of enhancing immunity are discussed. Evaluating the balance between obesity-induced inflammation and enhanced immunity warrants further study.
Collapse
Affiliation(s)
- Yuan Wu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Xu Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Qiong Li
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Chienshan Cheng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Lan Zheng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China.
| |
Collapse
|
9
|
Papakonstantinou E, Piperigkou Z, Karamanos NK, Zolota V. Altered Adipokine Expression in Tumor Microenvironment Promotes Development of Triple Negative Breast Cancer. Cancers (Basel) 2022; 14:4139. [PMID: 36077676 PMCID: PMC9454958 DOI: 10.3390/cancers14174139] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Obesity is a remarkably important factor for breast carcinogenesis and aggressiveness. The implication of increased BMI in triple negative breast cancer (TNBC) development is also well established. A malignancy-promoting role of the adipose tissue has been supposed, where the adipocytes that constitute the majority of stromal cells release pro-inflammatory cytokines and growth factors. Alterations in adipokines and their receptors play significant roles in breast cancer initiation, progression, metastasis, and drug response. Classic adipokines, such as leptin, adiponectin, and resistin, have been extensively studied in breast cancer and connected with breast cancer risk and progression. Notably, new molecules are constantly being discovered and the list is continuously growing. Additionally, substantial progress has been made concerning their differential expression in association with clinical and pathological parameters of tumors and the prognostic and predictive value of their dysregulation in breast cancer carcinogenesis. However, evidence regarding the mechanisms by which adipose tissue is involved in the development of TNBC is lacking. In the present article we comment on current data on the suggested involvement of these mediators in breast cancer development and progression, with particular emphasis on TNBC, to draw attention to the design of novel targeted therapies and biomarkers.
Collapse
Affiliation(s)
- Efthymia Papakonstantinou
- Department of Gynecology and Obstetrics, School of Medicine, University of Patras, 26504 Patras, Greece or
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH), Institute of Chemical Engineering Sciences (ICE-HT), 26504 Patras, Greece
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH), Institute of Chemical Engineering Sciences (ICE-HT), 26504 Patras, Greece
| | - Vasiliki Zolota
- Department of Pathology, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
10
|
Pan T, Ding H, Jin L, Zhang S, Wu D, Pan W, Dong M, Ma X, Chen Z. DNMT1-mediated demethylation of lncRNA MEG3 promoter suppressed breast cancer progression by repressing Notch1 signaling pathway. Cell Cycle 2022; 21:2323-2337. [PMID: 35822955 DOI: 10.1080/15384101.2022.2094662] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Breast carcinoma is one of the common causes of cancer-related mortality in women. Maternally expressed gene 3 (MEG3), a lncRNA located at 14q32, can be involved in carcinogenesis. In this study, we discovered that MEG3 was downregulated by CpG hypermethylation within its gene promoter. Functionally, treatment of breast cancer cells with the DNA methylation inhibitor 5-AzadC as well as silencing of DNA methyltransferase-1 (DNMT1) could decrease the abnormal hypermethylation of the MEG3 promoter, reverse MEG3 expression, inhibit cell proliferation and promote cell apoptosis. In addition, we found that MEG3 expression was negatively correlated with DNMT1. Mechanistically, MEG3 knockdown combined with 5-AzadC or sh-DNMT1 treatment restored the expression of Notch1 receptor, leading to the Notch1 pathway activation, and promoted the progression of epithelial mesenchymal transformation (EMT). Finally, the mice tumor model experiments showed that DNMT1 knockdown can increase MEG3 expression and inhibit tumor growth. Collectively, our findings uncovered that DNMT1-mediated MEG3 demethylation leads to MEG3 upregulation, which in turn inhibits the Notch1 pathway and EMT process in breast cancer.
Collapse
Affiliation(s)
- Tingting Pan
- Department of General Surgery, Diagnosis and Therapy Center of Thyroid and Breast, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| | - Haiwen Ding
- Department of Pharmacy, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| | - Le Jin
- Department of Pharmacy, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| | - Shaobo Zhang
- Department of General Surgery, Hefei High-tech Cardiovascular Hospital, Hefei, Anhui, P.R. China
| | - Delin Wu
- Department of General Surgery, Diagnosis and Therapy Center of Thyroid and Breast, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| | - Wanwan Pan
- Anhui Provincial Hospital, Wannan Medical College, Hefei, Anhui, P.R. China
| | - Menghao Dong
- Anhui Provincial Hospital, Wannan Medical College, Hefei, Anhui, P.R. China
| | - Xiaopeng Ma
- Department of General Surgery, Diagnosis and Therapy Center of Thyroid and Breast, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| | - Zhaolin Chen
- Department of Pharmacy, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| |
Collapse
|
11
|
Rajput PK, Sharma JR, Yadav UCS. Cellular and molecular insights into the roles of visfatin in breast cancer cells plasticity programs. Life Sci 2022; 304:120706. [PMID: 35691376 DOI: 10.1016/j.lfs.2022.120706] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 11/15/2022]
Abstract
Obesity has reached a pandemic proportion and is responsible for the augmentation of multimorbidity including certain cancers. With the rise in obesity amongst the female population globally, a concomitant increase in breast cancer (BC) incidence and related mortality has been observed. In the present review, we have elucidated the cellular and molecular insight into the visfatin-mediated cellular plasticity programs such as Epithelial to mesenchymal transition (EMT) and Endothelial to mesenchymal transition (EndoMT), and stemness-associated changes in BC cells. EMT and EndoMT are responsible for inducing metastasis in cancer cells and conferring chemotherapy resistance, immune escape, and infinite growth potential. Visfatin, an obesity-associated adipokine implicated in metabolic syndrome, has emerged as a central player in BC pathogenesis. Several studies have indicated the presence of visfatin in the tumor microenvironment (TME) where it augments EMT and EndoMT of BC cells. Further, Visfatin also modulates the TME by acting on the tumor stroma cells such as adipocytes, infiltrated immune cells, and adipose-associated stem cells that secrete factors such as cytokines, and extracellular vesicles responsible for augmenting cellular plasticity program. Visfatin induced altered metabolism of the cancer cells and molecular determinants such as non-coding RNAs involved in EMT and EndoMT have been discussed. We have also highlighted specific therapeutic targets that can be exploited for the development of effective BC treatment. Taken together, these advanced understandings of cellular and molecular insight into the visfatin-mediated cellular plasticity programs may stimulate the development of better approaches for the prevention and therapy of BC, especially in obese patients.
Collapse
Affiliation(s)
- Pradeep Kumar Rajput
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - Jiten R Sharma
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - Umesh C S Yadav
- Special Center for Molecular medicine, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
12
|
Updated Functional Roles of NAMPT in Carcinogenesis and Therapeutic Niches. Cancers (Basel) 2022; 14:cancers14092059. [PMID: 35565188 PMCID: PMC9103253 DOI: 10.3390/cancers14092059] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/22/2022] [Accepted: 04/18/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The advantages and applications of using the non-invasive way to detect serum biomarkers for assessing cancer diagnosis and prognosis have been explored. Nicotinamide phosphoribosyltransferase (NAMPT), also designated as pre-B-cell colony-enhancing factor (PBEF) or visfatin, is a secreted adipokine known to modulate tumor malignancies. Its significance in predicting cancer patient’s survival outcome further renders the implementation of NAMPT in clinical practice. In this review, recent discoveries of NAMPT in cancer studies were focused and integrated. We aim to provide updates for researchers who are proposing relevant objectives. Abstract Nicotinamide phosphoribosyltransferase (NAMPT) is notable for its regulatory roles in tumor development and progression. Emerging evidence regarding NAMPT somatic mutations in cancer patients, NAMPT expressional signatures in normal tissues and cancers, and the prognostic significance of NAMPT in many cancer types has attracted attention, and NAMPT is considered a potential biomarker of cancer. Recent discoveries have demonstrated the indirect association and direct biological functions of NAMPT in modulating cancer metastasis, proliferation, angiogenesis, cancer stemness, and chemoresistance to anticancer drugs. These findings warrant further investigation of the underlying mechanisms to provide knowledge for developing novel cancer therapeutics. In this review article, we explore recent research developments involving the oncogenic activities of NAMPT by summarizing current knowledge regarding NAMPT somatic mutations, clinical trials, transcriptome data, and clinical information and discoveries related to the NAMPT-induced signaling pathway in modulating hallmarks of cancer. Furthermore, the comprehensive representation of NAMPT RNA expression in a pancancer panel as well as in specific normal cell types at single-cell level are demonstrated. The results suggest potential sites and cell types that could facilitate NAMPT-related tumorigenesis. With this review, we aim to shed light on the regulatory roles of NAMPT in tumor development and progression, and provide information to guide future research directions in this field.
Collapse
|
13
|
Umar MI, Hassan W, Murtaza G, Buabeid M, Arafa E, Irfan HM, Asmawi MZ, Huang X. The Adipokine Component in the Molecular Regulation of Cancer Cell Survival, Proliferation and Metastasis. Pathol Oncol Res 2021; 27:1609828. [PMID: 34588926 PMCID: PMC8473628 DOI: 10.3389/pore.2021.1609828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/09/2021] [Indexed: 12/22/2022]
Abstract
A hormonal imbalance may disrupt the rigorously monitored cellular microenvironment by hampering the natural homeostatic mechanisms. The most common example of such hormonal glitch could be seen in obesity where the uprise in adipokine levels is in virtue of the expanding bulk of adipose tissue. Such aberrant endocrine signaling disrupts the regulation of cellular fate, rendering the cells to live in a tumor supportive microenvironment. Previously, it was believed that the adipokines support cancer proliferation and metastasis with no direct involvement in neoplastic transformations and tumorigenesis. However, the recent studies have reported discrete mechanisms that establish the direct involvement of adipokine signaling in tumorigenesis. Moreover, the individual adipokine profile of the patients has never been considered in the prognosis and staging of the disease. Hence, the present manuscript has focused on the reported extensive mechanisms that culminate the basis of poor prognosis and diminished survival rate in obese cancer patients.
Collapse
Affiliation(s)
| | - Waseem Hassan
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Manal Buabeid
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Elshaimaa Arafa
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | | | - Mohd Zaini Asmawi
- School of Pharmaceutical Sciences, University of Science Malaysia, Pulau Pinang, Malaysia
| | - Xianju Huang
- College of Pharmacy, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
14
|
Wang YY, Chen HD, Lo S, Chen YK, Huang YC, Hu SCS, Hsieh YC, Hung AC, Hou MF, Yuan SSF. Visfatin Enhances Breast Cancer Progression through CXCL1 Induction in Tumor-Associated Macrophages. Cancers (Basel) 2020; 12:cancers12123526. [PMID: 33256011 PMCID: PMC7760195 DOI: 10.3390/cancers12123526] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
Visfatin, an adipocytokine highly expressed in breast tumor tissues, is associated with breast cancer progression. Recent studies showed that adipocytokines mediate tumor development through adipocytokine tumor-stromal interactions in the tumor microenvironment. This study focused on the interaction between one key stromal constituent-tumor-associated macrophages-and visfatin. Pretreatment of THP-1 and peripheral blood mononuclear cells (PBMCs) with recombinant visfatin resulted in M2-polarization determined by CD163 and CD206 expression. Indirect co-culture with visfatin-treated THP-1 (V-THP-1) promoted the viability, migration, tumorsphere formation, EMT, and stemness of breast cancer cells. Cytokine array identified an increased CXCL1 secretion in V-THP-1 conditioned medium and recombinant CXCL1 enhanced cell migration and invasion, which were abrogated by the CXCL1-neutralizing antibody. Additionally, visfatin induced pERK in THP-1 cells and clinical samples confirmed a positive CXCL1/pERK correlation. In an orthotopic mouse model, the tumor bioluminescent signal of luciferase-expressing MDA-MB-231 (Luc-MDA-MB-231) cells co-cultured with V-THP-1 and the expression of proliferation marker Ki67 were significantly higher than that co-cultured with THP-1. Furthermore, tail vein-injected Luc-MDA-MB-231 pretreated with V-PBMCs conditioned medium metastasized to lungs more frequently compared to control, and this was reversed by CXCL1 blocking antibody. In summary, this study demonstrated that visfatin enhanced breast cancer progression via pERK/CXCL1 induction in macrophages.
Collapse
Affiliation(s)
- Yen-Yun Wang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-Y.W.); (Y.-K.C.)
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (H.-D.C.); (A.C.H.)
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Huan-Da Chen
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (H.-D.C.); (A.C.H.)
| | - Steven Lo
- Canniesburn Regional Plastic Surgery and Burns Unit, Glasgow Royal Infirmary, Glasgow G4 0SF, UK;
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Yuk-Kwan Chen
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-Y.W.); (Y.-K.C.)
- Division of Oral Pathology & Maxillofacial Radiology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Oral & Maxillofacial Imaging Center, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Ci Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Stephen Chu-Sung Hu
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Dermatology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 812, Taiwan
| | - Ya-Ching Hsieh
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK;
| | - Amos C. Hung
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (H.-D.C.); (A.C.H.)
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Ming-Feng Hou
- Division of General and Gastroenterological Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Shyng-Shiou F. Yuan
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (H.-D.C.); (A.C.H.)
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Chiao Tung University, Hsinchu 300, Taiwan
- Correspondence: ; Tel.: +886-7-312-1101 (ext. 2557)
| |
Collapse
|
15
|
Adipocytokines visfatin and resistin in breast cancer: Clinical relevance, biological mechanisms, and therapeutic potential. Cancer Lett 2020; 498:229-239. [PMID: 33152400 DOI: 10.1016/j.canlet.2020.10.045] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/11/2020] [Accepted: 10/26/2020] [Indexed: 12/17/2022]
Abstract
Obesity is one of the major modifiable risk factors in breast cancer, with obese adipose tissue showing a pathological role in breast cancer development and malignancy via the release of secretory factors, such as proinflammatory cytokines and adipocytokines. The current article focuses on visfatin and resistin, two such adipocytokines that have emerged over the last two decades as leading breast cancer promoting factors in obesity. The clinical association of circulating visfatin and resistin with breast cancer and their biological mechanisms are reviewed, in addition to their role in the context of tumor-stromal interactions in the breast cancer microenvironment. Recent findings have unraveled several mediators of visfatin and resistin that are involved in the crosstalk between breast cancer cells and adipose tissue in the breast tumor microenvironment, including growth differentiation factor 15 (GDF15), interleukin 6 (IL-6), and toll-like receptor 4 (TLR4). Finally, current therapeutics targeting visfatin and resistin and their respective pathways are discussed, including future therapeutic strategies such as new drug design or neutralizing peptides that target extracellular visfatin or resistin. These hold promise in the development of novel breast cancer therapies and are of increasing relevance as the prevalence of obesity-related breast cancer increases worldwide.
Collapse
|
16
|
Motawi TM, Zakhary NI, Darwish HA, Abdullah H, Tadros SA. Significance of Some Non-Invasive Biomarkers in the Early Diagnosis and Staging of Egyptian Breast Cancer Patients. Asian Pac J Cancer Prev 2020; 21:3279-3284. [PMID: 33247685 PMCID: PMC8033118 DOI: 10.31557/apjcp.2020.21.11.3279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Indexed: 12/24/2022] Open
Abstract
Introduction: Breast cancer is one of the most relevant malignancies among women. Early diagnosis and accurate staging of breast cancer is important for the selection of an appropriate therapeutic strategy and achieving a better outcome. Aim: This study aimed to explore the significance of some non-invasive biomarkers in the early diagnosis and staging of Egyptian breast cancer patients. Subjects and Methods: A total of 135 female patients with physically and pathologically confirmed breast cancer and 40 unrelated controls as well as 40 patients with benign breast mass were enrolled in this study. The malignant breast cancer group was further divided into four groups according to tumor size. Serum levels of carcinoembryonic antigen-related cell adhesion molecule-1 (CEACAM1), resistin and visfatin were determined by enzyme immunoassay. Results: Elevated levels of CEACAM1, resistin and visfatin were observed in breast cancer patients when compared with normal control and benign groups. The cutoff values, sensitivities and specificities of these biomarkers were appropriate for the discrimination of breast cancer from controls. Additionally, the serum levels of visfatin increased positively with tumor size and consequently with breast cancer stages. Conclusion: CEACAM1, resistin and visfatin are valuable in early diagnosis of breast cancer, with visfatin being preferentially used in staging.
Collapse
Affiliation(s)
- Tarek Mk Motawi
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nadia I Zakhary
- Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Hebatallah A Darwish
- Pharmacology, Toxicology and Biochemistry Department, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt (FUE), Cairo, Egypt
| | - Hassan Abdullah
- Department of Surgical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Samer A Tadros
- Department of Biochemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), October, Egypt
| |
Collapse
|
17
|
Liu C, Zhao Q, Yu X. Bone Marrow Adipocytes, Adipocytokines, and Breast Cancer Cells: Novel Implications in Bone Metastasis of Breast Cancer. Front Oncol 2020; 10:561595. [PMID: 33123472 PMCID: PMC7566900 DOI: 10.3389/fonc.2020.561595] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/27/2020] [Indexed: 02/05/2023] Open
Abstract
Accumulating discoveries highlight the importance of interaction between marrow stromal cells and cancer cells for bone metastasis. Bone is the most common metastatic site of breast cancer and bone marrow adipocytes (BMAs) are the most abundant component of the bone marrow microenvironment. BMAs are unique in their origin and location, and recently they are found to serve as an endocrine organ that secretes adipokines, cytokines, chemokines, and growth factors. It is reasonable to speculate that BMAs contribute to the modification of bone metastatic microenvironment and affecting metastatic breast cancer cells in the bone marrow. Indeed, BMAs may participate in bone metastasis of breast cancer through regulation of recruitment, invasion, survival, colonization, proliferation, angiogenesis, and immune modulation by their production of various adipocytokines. In this review, we provide an overview of research progress, focusing on adipocytokines secreted by BMAs and their potential roles for bone metastasis of breast cancer, and investigating the mechanisms mediating the interaction between BMAs and metastatic breast cancer cells. Based on current findings, BMAs may function as a pivotal modulator of bone metastasis of breast cancer, therefore targeting BMAs combined with conventional treatment programs might present a promising therapeutic option.
Collapse
Affiliation(s)
- Chang Liu
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Zhao
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Department of General Practice, West China Hospital, Sichuan University, Chengdu, China
| | - Xijie Yu
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Ezzati-Mobaser S, Malekpour-Dehkordi Z, Nourbakhsh M, Tavakoli-Yaraki M, Ahmadpour F, Golpour P, Nourbakhsh M. The up-regulation of markers of adipose tissue fibrosis by visfatin in pre-adipocytes as well as obese children and adolescents. Cytokine 2020; 134:155193. [PMID: 32707422 DOI: 10.1016/j.cyto.2020.155193] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/25/2020] [Accepted: 07/04/2020] [Indexed: 01/01/2023]
Abstract
Adipocytes are surrounded by a three-dimensional network of extracellular matrix (ECM) proteins. Aberrant ECM accumulation and remodeling leads to adipose tissue fibrosis. Visfatin is one of the adipocytokines that is increased in obesity and is implicated in insulin resistance. The objective of this study was to investigate the effect of visfatin on major components of ECM remodeling. In this study, plasma levels of both endotrophin and visfatin in obese children and adolescents were significantly higher than those in control subjects and they showed a positive correlation with each other. Treatment of 3T3-L1 pre-adipocytes with visfatin caused significant up-regulation of Osteopontin (Opn), Collagen type VI (Col6), matrix metalloproteinases MMP-2 and MMP-9. By using inhibitors of major signaling pathways it was shown that visfatin exerted its effect on Col6a3 gene expression through PI3K, JNK, and NF-кB pathways, while induced Opn gene expression via PI3K, JNK, MAPK/ERK, and NOTCH1. Our conclusion is that, the relationship between visfatin, endotrophin and insulin resistance parameters in obesity as well as increased expression of ECM proteins by visfatin suggests adipose tissue fibrosis as a mechanism for devastating effects of visfatin in obesity.
Collapse
Affiliation(s)
- Samira Ezzati-Mobaser
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Malekpour-Dehkordi
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Nourbakhsh
- Hazrat Aliasghar Children's Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Tavakoli-Yaraki
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ahmadpour
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Pegah Golpour
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran; Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Ok CY, Park S, Jang HO, Takata T, Bae MK, Kim YD, Ryu MH, Bae SK. Visfatin Induces Senescence of Human Dental Pulp Cells. Cells 2020; 9:cells9010193. [PMID: 31940881 PMCID: PMC7017355 DOI: 10.3390/cells9010193] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/17/2022] Open
Abstract
Dental pulp plays an important role in the health of teeth. The aging of teeth is strongly related to the senescence of dental pulp cells. A novel adipokine, visfatin, is closely associated with cellular senescence. However, little is known about the effect of visfatin on the senescence of human dental pulp cells (hDPCs). Here, it was found that in vivo visfatin levels in human dental pulp tissues increase with age and are upregulated in vitro in hDPCs during premature senescence activated by H2O2, suggesting a correlation between visfatin and senescence. In addition, visfatin knockdown by small interfering RNA led to the reduction in hDPC senescence; however, treatment with exogenous visfatin protein induced the senescence of hDPCs along with increased NADPH consumption, which was reversed by FK866, a chemical inhibitor of visfatin. Furthermore, visfatin-induced senescence was associated with both the induction of telomere damage and the upregulation of senescence-associated secretory phenotype (SASP) factors as well as NF-κB activation, which were all inhibited by FK866. Taken together, these results demonstrate, for the first time, that visfatin plays a pivotal role in hDPC senescence in association with telomere dysfunction and the induction of SASP factors.
Collapse
Affiliation(s)
- Chang Youp Ok
- Department of Dental Pharmacology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (C.Y.O.); (S.P.); (H.-O.J.)
- Periodontal Disease Signaling Network Research Center, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (M.-K.B.); (Y.-D.K.)
| | - Sera Park
- Department of Dental Pharmacology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (C.Y.O.); (S.P.); (H.-O.J.)
| | - Hye-Ock Jang
- Department of Dental Pharmacology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (C.Y.O.); (S.P.); (H.-O.J.)
| | | | - Moon-Kyoung Bae
- Periodontal Disease Signaling Network Research Center, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (M.-K.B.); (Y.-D.K.)
- Department of Oral Physiology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Yong-Deok Kim
- Periodontal Disease Signaling Network Research Center, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (M.-K.B.); (Y.-D.K.)
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Mi Heon Ryu
- Department of Oral Pathology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea;
| | - Soo-Kyung Bae
- Department of Dental Pharmacology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (C.Y.O.); (S.P.); (H.-O.J.)
- Periodontal Disease Signaling Network Research Center, School of Dentistry, Pusan National University, Yangsan 50612, Korea; (M.-K.B.); (Y.-D.K.)
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Correspondence: ; Tel.: +82-51-510-8253
| |
Collapse
|
20
|
Mohammadi M, Moradi A, Farhadi J, Akbari A, Pourmandi S, Mehrad-Majd H. Prognostic value of visfatin in various human malignancies: A systematic review and meta-analysis. Cytokine 2020; 127:154964. [PMID: 31901760 DOI: 10.1016/j.cyto.2019.154964] [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: 07/13/2019] [Revised: 11/24/2019] [Accepted: 12/25/2019] [Indexed: 12/16/2022]
Abstract
Although numerous studies have shown that visfatin is linked to several cancers, its prognostic value is still unclear. This first comprehensive meta-analysis was performed to evaluate the prognostic effect of visfatin in cancer patients. A systematic search was conducted for relevant studies in health-related electronic databases up to May 2019. The pooled hazard ratios (HRs) and ORs with 95% confidence intervals (CIs) for total and stratified analyses were calculated to demonstrate the prognostic value of visfatin expression level in cancer patients. Heterogeneity and publication bias were also investigated. A total of 14 eligible studies with 1616 patients were included in the current meta-analysis. Pooling results revealed that, high visfatin expression was significantly associated with poorer overall survival (OS) (HR = 2.43, 95% CI 1.64-3.62, P < 0.001). Elevated visfatin level was also correlated with positive lymph node metastasis (OR = 2.45, 95% CI 1.43-4.17, P ≤ 0.001), positive distance metastasis (OR = 2014, 95% CI 1.25-3.69, P ≤ 0.001), advanced tumor stage (OR = 3.01, 95% CI 1.91-7.72, P ≤ 0.001), and larger tumor size (OR = 1.99, 95% CI 1.49-2.69, P ≤ 0.001). Our meta-results indicates that altered visfatin expression is a potential indicator of poor clinical outcomes in tumor patients, suggesting that high visfatin expression may serve as a potential biomarker of poor prognosis and metastasis in cancers.
Collapse
Affiliation(s)
- Masoumeh Mohammadi
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Moradi
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Farhadi
- Department of Biochemistry and Molecular Biology, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Abolfazl Akbari
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shokoufeh Pourmandi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hassan Mehrad-Majd
- Cancer Molecular Pathology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
21
|
Visfatin Mediates Malignant Behaviors through Adipose-Derived Stem Cells Intermediary in Breast Cancer. Cancers (Basel) 2019; 12:cancers12010029. [PMID: 31861872 PMCID: PMC7016886 DOI: 10.3390/cancers12010029] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 12/31/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) have been implicated in tumor growth and metastasis in breast cancer. ADSCs exhibit tumor tropism, and are of increasing clinical relevance due to the autologous fat grafting for breast reconstruction. Although we have previously shown that a high level of the adipocytokine visfatin in human breast cancer tissues correlated with tumor progression mediated by cAbl and STAT3, the effects of visfatin in the tumor microenvironment are unclear. To understand how visfatin modulates breast cancer within the tumor-stromal environment, we examined determinants of breast cancer progression using a visfatin-primed ADSCs-tumor co-culture model. ADSCs were isolated from tumor-free adipose tissue adjacent to breast tumors. ADSCs were treated with or without visfatin for 48 h and then collected for co-culture with breast cancer cell line MDA-MB-231 for 72 h in a transwell system. We found that the MDA-MB-231 cells co-cultured with visfatin-treated ADSCs (vADSCs) had higher levels of cell viability, anchorage independent growth, migration, invasion, and tumorsphere formation than that co-cultured with untreated ADSCs (uADSCs). Growth differentiation factor 15 (GDF15) upregulation was found in the co-culture conditioned medium, with GDF15 neutralizing antibody blocking the promoting effect on MDA-MB-231 in co-culture. In addition, a GDF15-induced AKT pathway was found in MDA-MB-231 and treatment with PI3K/AKT inhibitor also reversed the promoting effect. In an orthotopic xenograft mouse model, MDA-MB-231 co-injected with vADSCs formed a larger tumor mass than with uADSCs. Positive correlations were noted between visfatin, GDF15, and phosphor-AKT expressions in human breast cancer specimens. In conclusion, visfatin activated GDF15-AKT pathway mediated via ADSCs to facilitate breast cancer progression.
Collapse
|
22
|
Christodoulatos GS, Spyrou N, Kadillari J, Psallida S, Dalamaga M. The Role of Adipokines in Breast Cancer: Current Evidence and Perspectives. Curr Obes Rep 2019; 8:413-433. [PMID: 31637624 DOI: 10.1007/s13679-019-00364-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE The current review shows evidence for the role of adipokines in breast cancer (BC) pathogenesis summarizing the mechanisms underlying the association between adipokines and breast malignancy. Special emphasis is given also on intriguing insights into the relationship between obesity and BC as well as on the role of novel adipokines in BC development. RECENT FINDINGS Recent evidence has underscored the role of the triad of obesity, insulin resistance, and adipokines in postmenopausal BC. Adipokines exert independent and joint effects on activation of major intracellular signal networks implicated in BC cell proliferation, growth, survival, invasion, and metastasis, particularly in the context of obesity, considered a systemic endocrine dysfunction characterized by chronic inflammation. To date, more than 10 adipokines have been linked to BC, and this catalog is continuously increasing. The majority of circulating adipokines, such as leptin, resistin, visfatin, apelin, lipocalin 2, osteopontin, and oncostatin M, is elevated in BC, while some adipokines such as adiponectin and irisin (adipo-myokine) are generally decreased in BC and considered protective against breast carcinogenesis. Further evidence from basic and translational research is necessary to delineate the ontological role of adipokines and their interplay in BC pathogenesis. More large-scale clinical and longitudinal studies are awaited to assess their clinical utility in BC prognosis and follow-up. Finally, novel more effective and safer adipokine-centered therapeutic strategies could pave the way for targeted oncotherapy.
Collapse
Affiliation(s)
- Gerasimos Socrates Christodoulatos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias, Goudi, 11527, Athens, Greece
- Laboratory of Microbiology, KAT Hospital, 2 Nikis, Kifisia, 14561, Athens, Greece
| | - Nikolaos Spyrou
- 251 Airforce General Hospital, 3 Kanellopoulou, 11525, Athens, Greece
| | - Jona Kadillari
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias, Goudi, 11527, Athens, Greece
| | - Sotiria Psallida
- Laboratory of Microbiology, KAT Hospital, 2 Nikis, Kifisia, 14561, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias, Goudi, 11527, Athens, Greece.
| |
Collapse
|
23
|
Neutrophil Maturation and Survival Is Controlled by IFN-Dependent Regulation of NAMPT Signaling. Int J Mol Sci 2019; 20:ijms20225584. [PMID: 31717318 PMCID: PMC6888478 DOI: 10.3390/ijms20225584] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/01/2019] [Accepted: 11/06/2019] [Indexed: 12/16/2022] Open
Abstract
Granulocyte-colony stimulating factor (G-CSF)/nicotinamide phosphoribosyltransferase (NAMPT) signaling has been shown to be crucial for the modulation of neutrophil development and functionality. As this signaling pathway is significantly suppressed by type I interferons (IFNs), we aimed to study how the regulation of neutrophil differentiation and phenotype is altered in IFN-deficient mice during granulopoiesis. The composition of bone marrow granulocyte progenitors and their Nampt expression were assessed in bone marrow of type I IFN receptor knockout (Ifnar1-/-) mice and compared to wild-type animals. The impact of NAMPT inhibition on the proliferation, survival, and differentiation of murine bone marrow progenitors, as well as of murine 32D and human HL-60 neutrophil-like cell lines, was estimated. The progressive increase of Nampt expression during neutrophil progenitor maturation could be observed, and it was more prominent in IFN-deficient animals. Altered composition of bone marrow progenitors in these mice correlated with the dysregulation of apoptosis and altered differentiation of these cells. We observed that NAMPT is vitally important for survival of early progenitors, while at later stages it delays the differentiation of neutrophils, with moderate effect on their survival. This study shows that IFN-deficiency leads to the elevated NAMPT expression in the bone marrow, which in turn modulates neutrophil development and differentiation, even in the absence of tumor-derived stimuli.
Collapse
|
24
|
Hui Z, Liu Z, He A, Chen Y, Zhang P, Lei B, Yao H, Yu Y, Liang R, Li Z, Zhang W. Visfatin promotes the malignancy of human acute myeloid leukemia cells via regulation of IL-17. Eur J Pharmacol 2019; 853:103-110. [PMID: 30876976 DOI: 10.1016/j.ejphar.2019.03.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 03/10/2019] [Accepted: 03/11/2019] [Indexed: 11/29/2022]
Abstract
Understanding the mechanisms of malignancy in acute myeloid leukemia (AML) cell is important for the targeted treatment and drug development. We found that visfatin, a 52-kDa adipokine, can positively regulate the proliferation of AML cells. Targeted inhibition of visfatin via its specific siRNAs or inhibitor can suppress the proliferation of AML cells. Further, knockdown of visfatin can increase the doxorubicin (Dox) and cisplatin (CDDP) sensitivity of AML cells. Among the tested six cytokines, si-visfatin can decrease the expression of interleukin-17 (IL-17). Over expression of IL-17 can reverse si-visfatin suppressed cell proliferation and increased Dox sensitivity. The upregulation of IL-17 was also involved in visfatin induced activation of PI3K/Akt signals in AML cells. The inhibitor of PI3K/Akt can synergistically suppress the proliferation of HL60 cells which were transfected with si-visfatin. Knockdown of visfatin can increase the expression of miR-135a, which can bind to the 3'UTR of IL-17 and decrease its expression. The inhibitor of miR-135a can attenuate si-visfatin suppressed expression of IL-17 and proliferation of AML cells. Collectively, our data suggested that visfatin can increase the malignancy of AML cells via regulation of miR-135a/IL-17/PI3K/Akt signals.
Collapse
Affiliation(s)
- Zengqian Hui
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Shaanxi Provincial Corps Hospital of Chinese People's Armed Police Force, Xi'an, Shaanxi 710054, China
| | - Zhao Liu
- Department of Surgery, Xi'an Chest Hospital, Xi'an TB & Thoracic Tumor Hospital, Xi'an, Shaanxi 710100, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yinxia Chen
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Pengyu Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Bo Lei
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Huan Yao
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yong Yu
- Shaanxi Provincial Corps Hospital of Chinese People's Armed Police Force, Xi'an, Shaanxi 710054, China
| | - Rui Liang
- Shaanxi Provincial Corps Hospital of Chinese People's Armed Police Force, Xi'an, Shaanxi 710054, China
| | - Zhanning Li
- Shaanxi Provincial Corps Hospital of Chinese People's Armed Police Force, Xi'an, Shaanxi 710054, China
| | - Wanggang Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
25
|
Visfatin serum concentration and hepatic mRNA expression in chronic hepatitis C. Clin Exp Hepatol 2019; 5:147-154. [PMID: 31501791 PMCID: PMC6728865 DOI: 10.5114/ceh.2019.85074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 12/26/2018] [Indexed: 12/15/2022] Open
Abstract
Aim of the study Chronic hepatitis C (CHC) is a viral disease with metabolic disturbances involved in its pathogenesis. Adipokines may influence the inflammatory response and contribute to development of metabolic abnormalities in CHC. Visfatin exerts immunomodulatory and insulin-mimetic effects. The aim was to measure visfatin serum concentrations and its mRNA hepatic expression in non-obese CHC patients and to assess the relationships with metabolic and histological parameters. Material and methods In a group of 63 non-obese CHC patients (29 M/34 F) infected with genotype 1b aged 46.6 ±14.6 years, body mass index (BMI) 24.8 ±3.0 kg/m2, serum visfatin levels and its mRNA hepatic expression were examined and the subsequent associations with metabolic and histopathological features were assessed. Results Serum visfatin levels were significantly higher in CHC patients compared to controls (22.7 ±5.7 vs. 17.8 ±1.5 ng/ml, p < 0.001). There was no difference in serum visfatin and its mRNA hepatic expression regardless of sex, BMI, insulin sensitivity and lipids concentrations. There was no mutual correlation between serum visfatin and visfatin mRNA hepatic expression. Hepatic visfatin mRNA levels but not visfatin serum levels were higher in patients with steatosis (1.35 ±0.75 vs. 0.98 ±0.34, p = 0.009). Conclusions Serum visfatin levels may reflect its involvement in chronic inflammatory processes accompanying HCV infection. Increased visfatin mRNA hepatic expression in patients with steatosis seems to be a compensatory mechanism enabling hepatocytes to survive metabolic abnormalities resulting from virus-related lipid droplet deposition prerequisite to HCV replication.
Collapse
|
26
|
Garikapati KK, Ammu VVVRK, Krishnamurthy PT, Chintamaneni PK, Pindiprolu SKSS. Possible role of Thiazolidinedione in the management of Type-II Endometrial Cancer. Med Hypotheses 2019; 126:78-81. [PMID: 31010504 DOI: 10.1016/j.mehy.2019.03.014] [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: 12/30/2018] [Revised: 02/19/2019] [Accepted: 03/21/2019] [Indexed: 10/27/2022]
Abstract
Type-II Endometrial Cancer (EMC) is one of the most common types of gynaecological cancer affecting more than 2.7 million people worldwide. Clinical evidence shows that adipokines levels are abnormally altered in Type-II EMC and reported to be one of the major responsible factor for uncontrolled proliferation and metastasis in Type-II EMC. Reversing the altered adipokine levels, therefore, help to control Type-II EMC proliferation and metastasis. In the present hypothesis we focus on the possible role of Thiazolidinediones in favourably altering the adipokine levels to benefit in the management of Type-II EMC.
Collapse
Affiliation(s)
- Kusuma Kumari Garikapati
- Department of Pharmacology, JSS College of Pharmacy (a Constituent College of JSS Academy of Higher Education and Research), Udhagamandalam, Tamil Nadu 643001, India
| | - V V V Ravi Kiran Ammu
- Department of Pharmacology, JSS College of Pharmacy (a Constituent College of JSS Academy of Higher Education and Research), Udhagamandalam, Tamil Nadu 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (a Constituent College of JSS Academy of Higher Education and Research), Udhagamandalam, Tamil Nadu 643001, India.
| | - Pavan Kumar Chintamaneni
- Department of Pharmacology, JSS College of Pharmacy (a Constituent College of JSS Academy of Higher Education and Research), Udhagamandalam, Tamil Nadu 643001, India
| | - Sai Kiran S S Pindiprolu
- Department of Pharmacology, JSS College of Pharmacy (a Constituent College of JSS Academy of Higher Education and Research), Udhagamandalam, Tamil Nadu 643001, India
| |
Collapse
|
27
|
Lin TC. The role of visfatin in cancer proliferation, angiogenesis, metastasis, drug resistance and clinical prognosis. Cancer Manag Res 2019; 11:3481-3491. [PMID: 31114381 PMCID: PMC6497876 DOI: 10.2147/cmar.s199597] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 03/11/2019] [Indexed: 12/21/2022] Open
Abstract
Visfatin, also known as nicotinamide phosphoribosyltransferase or pre-B-cell colony-enhancing factor (PBEF), is an adipocytokine secreted by adipocytes, macrophages and inflamed endothelial tissue. Related reports have indicated a positive correlation between the visfatin level and obesity and cancer risk. In addition to its original function, visfatin is multifunctional and plays critical roles in the promotion of several processes relevant to cancer progression including cancer cell proliferation, angiogenesis, metastasis and drug resistance. The relative expression of visfatin and the potential visfatin receptor on a pan-cancer scale was determined based on the transcriptome analysis data in The Cancer Genome Atlas. We further show the clinical association of its signaling axis with the survival of cancer patients, which reveals its prognostic power for specific cancer types. This review illustrates visfatin’s biological functions related to cancer progression and demonstrates its clinical significance in predicting outcomes of cancer patients.
Collapse
Affiliation(s)
- Tsung-Chieh Lin
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
| |
Collapse
|
28
|
Wang Z, Gao S, Sun C, Li J, Gao W, Yu L. Clinical significance of serum adiponectin and visfatin levels in endometrial cancer. Int J Gynaecol Obstet 2019; 145:34-39. [PMID: 30702161 DOI: 10.1002/ijgo.12772] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 08/09/2018] [Accepted: 01/01/2019] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To investigate serum adiponectin and visfatin levels, and their ratio, in patients with endometrial cancer. METHODS A retrospective case-control study of 53 patients with endometrial cancer admitted to Dalian Municipal Women and Children's Medical Center, China, between May 1, 2009, and January 31, 2013. Ninety-eight healthy women who underwent physical examination at the same time served as the control group. Serum adiponectin and visfatin levels were measured by ELISA. RESULTS Serum adiponectin level in the endometrial cancer group was significantly lower than in the control group (2.09 ± 1.24 μg/mL vs 7.59 ± 2.29 μg/mL; P<0.001). Serum adiponectin was positively correlated with visfatin level (Spearman correlation coefficient 0.472). The visfatin:adiponectin ratio in the endometrial cancer group was significantly higher than the control group (0.28 ± 0.10 vs 0.11 ± 0.09; P=0.047). Multivariate logistic regression showed that decreased serum adiponectin (odds ratio 0.998, 95% CI 0.996-0.999; P=0.045) and increased visfatin (1.010, 1.003-1.017; P=0.042) levels were independent risk factors for the onset of endometrial cancer. CONCLUSION Decreased serum adiponectin or increased visfatin levels are independent risk factors for endometrial cancer. The visfatin:adiponectin ratio has a certain reference value for the diagnosis of endometrial cancer.
Collapse
Affiliation(s)
- Zhongmin Wang
- Female Pelvic Floor Urinary Reconstructive Center, Dalian Municipal Women and Children's Medical Center, Dalian, China
| | - Shan Gao
- Female Pelvic Floor Urinary Reconstructive Center, Dalian Municipal Women and Children's Medical Center, Dalian, China
| | - Caixia Sun
- Female Pelvic Floor Urinary Reconstructive Center, Dalian Municipal Women and Children's Medical Center, Dalian, China
| | - Jinyan Li
- Female Pelvic Floor Urinary Reconstructive Center, Dalian Municipal Women and Children's Medical Center, Dalian, China
| | - Wenhui Gao
- Female Pelvic Floor Urinary Reconstructive Center, Dalian Municipal Women and Children's Medical Center, Dalian, China
| | - Liping Yu
- Female Pelvic Floor Urinary Reconstructive Center, Dalian Municipal Women and Children's Medical Center, Dalian, China
| |
Collapse
|
29
|
Spyrou N, Avgerinos KI, Mantzoros CS, Dalamaga M. Classic and Novel Adipocytokines at the Intersection of Obesity and Cancer: Diagnostic and Therapeutic Strategies. Curr Obes Rep 2018; 7:260-275. [PMID: 30145771 DOI: 10.1007/s13679-018-0318-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW In this review, we investigate the role of classic and novel adipocytokines in cancer pathogenesis synopsizing the mechanisms underlying the association between adipocytokines and malignancy. Special emphasis is given on novel adipocytokines as new evidence is emerging regarding their entanglement in neoplastic development. RECENT FINDINGS Recent data have emphasized the role of the triad of overweight/obesity, insulin resistance and adipocytokines in cancer. In the setting of obesity, classic and novel adipocytokines present independent and joint effects on activation of major intracellular signaling pathways implicated in cell proliferation, expansion, survival, adhesion, invasion, and metastasis. Until now, more than 15 adipocytokines have been associated with cancer, and this list continues to expand. While the plethora of circulating pro-inflammatory adipocytokines, such as leptin, resistin, extracellular nicotinamide phosphoribosyl transferase, and chemerin are elevated in malignancies, some adipocytokines such as adiponectin and omentin-1 are generally decreased in cancers and are considered protective against carcinogenesis. Elucidating the intertwining of inflammation, cellular bioenergetics, and adiposopathy is significant for the development of preventive, diagnostic, and therapeutic strategies against cancer. Novel more effective and safe adipocytokine-centered therapeutic interventions may pave the way for targeted oncotherapy.
Collapse
Affiliation(s)
- Nikolaos Spyrou
- 251 Airforce General Hospital, Kanellopoulou 3, 11525, Athens, Greece
| | | | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
- Section of Endocrinology, VA Boston Healthcare System, Boston, MA, USA
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece.
| |
Collapse
|
30
|
Maillard V, Elis S, Desmarchais A, Hivelin C, Lardic L, Lomet D, Uzbekova S, Monget P, Dupont J. Visfatin and resistin in gonadotroph cells: expression, regulation of LH secretion and signalling pathways. Reprod Fertil Dev 2018; 29:2479-2495. [PMID: 28672116 DOI: 10.1071/rd16301] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 05/21/2017] [Indexed: 12/15/2022] Open
Abstract
Visfatin and resistin appear to interfere with reproduction in the gonads, but their potential action at the hypothalamic-pituitary level is not yet known. The aim of the present study was to investigate the mRNA and protein expression of these adipokines in murine gonadotroph cells and to analyse the effects of different concentrations of recombinant mouse visfatin and resistin (0.01, 0.1, 1 and 10ngmL-1) on LH secretion and signalling pathways in LβT2 cells and/or in primary female mouse pituitary cells. Both visfatin and resistin mRNA and protein were found in vivo in gonadotroph cells. In contrast with resistin, the primary tissue source of visfatin in the mouse was the skeletal muscle, and not adipose tissue. Visfatin and resistin both decreased LH secretion from LβT2 cells after 24h exposure of cells (P<0.03). These results were confirmed for resistin in primary cell culture (P<0.05). Both visfatin (1ngmL-1) and resistin (1ngmL-1) increased AMP-activated protein kinase α phosphorylation in LβT2 cells after 5 or 10min treatment, up to 60min (P<0.04). Extracellular signal-regulated kinase 1/2 phosphorylation was transiently increased only after 5min resistin (1ngmL-1) treatment (P<0.01). In conclusion, visfatin and resistin are expressed in gonadotroph cells and they may affect mouse female fertility by regulating LH secretion at the level of the pituitary.
Collapse
Affiliation(s)
- Virginie Maillard
- UMR85 PRC, INRA, CNRS, IFCE, Université de Tours, 37380 Nouzilly, France
| | - Sébastien Elis
- UMR85 PRC, INRA, CNRS, IFCE, Université de Tours, 37380 Nouzilly, France
| | - Alice Desmarchais
- UMR85 PRC, INRA, CNRS, IFCE, Université de Tours, 37380 Nouzilly, France
| | - Céline Hivelin
- UMR85 PRC, INRA, CNRS, IFCE, Université de Tours, 37380 Nouzilly, France
| | - Lionel Lardic
- UMR85 PRC, INRA, CNRS, IFCE, Université de Tours, 37380 Nouzilly, France
| | - Didier Lomet
- UMR85 PRC, INRA, CNRS, IFCE, Université de Tours, 37380 Nouzilly, France
| | - Svetlana Uzbekova
- UMR85 PRC, INRA, CNRS, IFCE, Université de Tours, 37380 Nouzilly, France
| | - Philippe Monget
- UMR85 PRC, INRA, CNRS, IFCE, Université de Tours, 37380 Nouzilly, France
| | - Joëlle Dupont
- UMR85 PRC, INRA, CNRS, IFCE, Université de Tours, 37380 Nouzilly, France
| |
Collapse
|
31
|
Zangooei M, Nourbakhsh M, Ghahremani MH, Meshkani R, Khedri A, Shadboorestan A, Shokri Afra H, Shahmohamadnejad S, Mirmiranpour H, Khaghani S. Investigating the effect of visfatin on ERalpha phosphorylation (Ser118 and Ser167) and ERE-dependent transcriptional activity. EXCLI JOURNAL 2018; 17:516-525. [PMID: 30034315 PMCID: PMC6046625 DOI: 10.17179/excli2018-1299] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 05/25/2018] [Indexed: 01/09/2023]
Abstract
Obesity is associated with higher postmenopausal breast cancer incidence. Visfatin level alteration is one of the mechanisms by which obesity promotes cancer. Ligand-independent activation of estrogen receptor alpha (ERα) is also associated with carcinogenesis. The activity of ERα is modulated through phosphorylation on multiple sites by a number of protein kinases. Here we investigated the effect of visfatin as a novel adipocytokine on the phosphorylation and activity of ERα in MCF-7 breast cancer cells. We showed that exogenous administration of visfatin significantly increased the phosphorylation of ERα at serine 118 (Ser118) and 167 (Ser167) residues. Visfatin-induced Ser118 phosphorylation was diminished after treatment of cells with U0126 (MEK1/2 inhibitor). Furthermore, our results showed that visfatin-induced Ser167 phosphorylation is mediated through both MAPK and PI3K/Akt signaling pathways. Inhibition of the enzymatic activity of visfatin by FK866 had no effect on phosphorylation of ERα. We also showed that visfatin enhanced the estrogen response element (ERE)-dependent activity of ER in the presence of 17-β estradiol (E2). Additional study on T47D cells showed that visfatin also increased Ser118 and Ser167 phosphorylation of ERα and enhanced ERE-dependent activity in the presence of E2 in these cells.
Collapse
Affiliation(s)
- Mohammad Zangooei
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Azam Khedri
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Shadboorestan
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hajar Shokri Afra
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shiva Shahmohamadnejad
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Mirmiranpour
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahnaz Khaghani
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Dalamaga M, Christodoulatos GS, Mantzoros CS. The role of extracellular and intracellular Nicotinamide phosphoribosyl-transferase in cancer: Diagnostic and therapeutic perspectives and challenges. Metabolism 2018; 82:72-87. [PMID: 29330025 DOI: 10.1016/j.metabol.2018.01.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/23/2017] [Accepted: 01/04/2018] [Indexed: 02/07/2023]
Abstract
Nicotinamide phosphoribosyl-transferase (Nampt) or pre-B cell colony-enhancing factor or visfatin represents a pleiotropic molecule acting as an enzyme, a cytokine and a growth factor. Intracellular Nampt plays an important role in cellular bioenergetics and metabolism, particularly NAD biosynthesis. NAD biosynthesis is critical in DNA repair, oncogenic signal transduction, transcription, genomic integrity and apoptosis. Although its insulin-mimetic function remains a controversial issue, extracellular Nampt presents proliferative, anti-apoptotic, pro-inflammatory, pro-angiogenic and metastatic properties. Nampt is upregulated in many malignancies, including obesity-associated cancers, and is associated with worse prognosis. Serum Nampt may be a potential diagnostic and prognostic biomarker in cancer. Pharmacologic agents that neutralize Nampt or medications that decrease Nampt levels or downregulate signaling pathways downstream of Nampt may prove to be useful anti-cancer treatments. In particular, Nampt inhibitors as monotherapy or in combination therapy have displayed anti-cancer activity in vivo and in vitro. The aim of this review is to explore the role of Nampt in cancer pathophysiology as well as to synopsize the mechanisms underlying the association between extracellular and intracellular Nampt, and malignancy. Exploring the interplay of cellular bioenergetics, inflammation and adiposopathy is expected to be of importance in the development of preventive and therapeutic strategies against cancer.
Collapse
Affiliation(s)
- Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527 Athens, Greece.
| | - Gerasimos Socrates Christodoulatos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527 Athens, Greece; Department of Microbiology, KAT Hospital, Nikis 2, Kifisia, 14561 Athens, Greece
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
Ferreira A, Lamy M, Margarida Rocha M, Silva G, Bandeiras TM, Barbas A. Production and characterization of a novel Delta-like 1 functional unit as a tool for Notch pathway activation and generation of a specific antibody. Protein Expr Purif 2018; 146:8-16. [PMID: 29366964 DOI: 10.1016/j.pep.2018.01.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/17/2018] [Accepted: 01/19/2018] [Indexed: 12/13/2022]
Abstract
Notch signalling is an evolutionary conserved cell-to-cell communication pathway crucial for development and tissue homeostasis. Abnormal Notch signalling by mutations or deregulated expression of its receptors and/or ligands can lead to cancer making it a potential therapeutic target. Delta-like1 (DLL1) is a ligand of the Notch pathway implicated in different types of cancer, including breast cancer. Herein, we produced rhDLL1-DE3, a novel soluble form of DLL1 protein, which contains the DSL domain and EGF1-3 repeats critical for Notch pathway activation. cDNA fragments of human DLL1, encoding truncated versions of DLL1 with regions required to activate Notch receptors, were cloned and expressed as histidine-fused proteins in bacterial and mammalian cells. Expression tests in mammalian cells showed almost exclusively expression of the rhDLL1-DE3 protein form comprising the minimal binding regions DSL to EGF3 to Notch receptors. The highest yield of rhDLL1-DE3 was obtained from E. coli inclusion bodies. The produced protein, with purity higher than 95% bound to human Notch1 recombinant protein, by both Biolayer interferometry and ELISA assays. Cellular assays revealed rhDLL1-DE3 was biologically active as it increased expression of Notch-dependent genes in inducible pluripotent and breast cancer cells. Moreover, rhDLL1-DE3 allowed the generation of polyclonal antibodies by immunization that efficiently recognized DLL1 proteins by immunoblot, and caused a significant decrease of Notch1 expression in MCF7 breast cancer cells. The rhDLL1-DE3 protein might thus be used for Notch pathway activation and to generate anti-DLL1 monoclonal antibodies by immunization or phage display technology to unveil the effect of DLL1 in breast cancer.
Collapse
Affiliation(s)
- Andreia Ferreira
- iBET - Instituto de Biologia Experimental e Tecnológica, Portugal
| | - Márcia Lamy
- iBET - Instituto de Biologia Experimental e Tecnológica, Portugal; FairJourney Biologics, Porto, Portugal
| | | | - Gabriela Silva
- iBET - Instituto de Biologia Experimental e Tecnológica, Portugal
| | - Tiago M Bandeiras
- iBET - Instituto de Biologia Experimental e Tecnológica, Portugal; ITQB - Instituto de Tecnologia Química e Biológica, Portugal
| | - Ana Barbas
- iBET - Instituto de Biologia Experimental e Tecnológica, Portugal; Bayer Portugal, Carnaxide, Portugal.
| |
Collapse
|
34
|
Behrouzfar K, Alaee M, Nourbakhsh M, Gholinejad Z, Golestani A. Extracellular NAMPT/visfatin causes p53 deacetylation via NAD production and SIRT1 activation in breast cancer cells. Cell Biochem Funct 2017; 35:327-333. [PMID: 28845527 DOI: 10.1002/cbf.3279] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/09/2017] [Accepted: 07/05/2017] [Indexed: 01/19/2023]
Abstract
Visfatin, which is secreted as an adipokine and cytokine, has been implicated in cancer development and progression. In this study, we investigated the NAD-producing ability of visfatin and its relationship with SIRT1 (silent information regulator 2) and p53 to clarify the role of visfatin in breast cancer. MCF-7 breast cancer cells were cultured and treated with visfatin. SIRT1 activity was assessed by measuring fluorescence intensity from fluoro-substrate peptide. To investigate the effect of visfatin on p53 acetylation, SDS-PAGE followed by western blotting was performed using specific antibodies against p53 and its acetylated form. Total NAD was measured both in cell lysate and the extracellular medium by colorimetric method. Visfatin increased both extracellular and intracellular NAD concentrations. It also induced proliferation of breast cancer cells, an effect that was abolished by inhibition of its enzymatic activity. Visfatin significantly increased SIRT1 activity, accompanied by induction of p53 deacetylation. In conclusion, the results show that extracellular visfatin produces NAD that causes upregulation of SIRT1 activity and p53 deacetylation. These findings explain the relationship between visfatin and breast cancer progression.
Collapse
Affiliation(s)
- Kiarash Behrouzfar
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Alaee
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zafar Gholinejad
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Golestani
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Banik U, Parasuraman S, Adhikary AK, Othman NH. Curcumin: the spicy modulator of breast carcinogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:98. [PMID: 28724427 PMCID: PMC5517797 DOI: 10.1186/s13046-017-0566-5] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 07/10/2017] [Indexed: 12/22/2022]
Abstract
Worldwide breast cancer is the most common cancer in women. For many years clinicians and the researchers are examining and exploring various therapeutic modalities for breast cancer. Yet the disease has remained unconquered and the quest for cure is still going on. Present-day strategy of breast cancer therapy and prevention is either combination of a number of drugs or a drug that modulates multiple targets. In this regard natural products are now becoming significant options. Curcumin exemplifies a promising natural anticancer agent for this purpose. This review primarily underscores the modulatory effect of curcumin on the cancer hallmarks. The focus is its anticancer effect in the complex pathways of breast carcinogenesis. Curcumin modulates breast carcinogenesis through its effect on cell cycle and proliferation, apoptosis, senescence, cancer spread and angiogenesis. Largely the NFkB, PI3K/Akt/mTOR, MAPK and JAK/STAT are the key signaling pathways involved. The review also highlights the curcumin mediated modulation of tumor microenvironment, cancer immunity, breast cancer stem cells and cancer related miRNAs. Using curcumin as a therapeutic and preventive agent in breast cancer is perplexed by its diverse biological activity, much of which remains inexplicable. The information reviewed here should point toward potential scope of future curcumin research in breast cancer.
Collapse
Affiliation(s)
- Urmila Banik
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.,Unit of Pathology, AIMST University, Faculty of Medicine, Semeling, 08100, Bedong, Kedah, Malaysia
| | - Subramani Parasuraman
- Unit of Pharmacology, AIMST University, Faculty of Pharmacy, Semeling, 08100, Bedong, Kedah, Malaysia
| | - Arun Kumar Adhikary
- Unit of Microbiology, AIMST University, Faculty of Medicine, Semeling, 08100, Bedong, Kedah, Malaysia
| | - Nor Hayati Othman
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
36
|
Gholinejad Z, Kheiripour N, Nourbakhsh M, Ilbeigi D, Behroozfar K, Hesari Z, Golestani A, Shabani M, Einollahi N. Extracellular NAMPT/Visfatin induces proliferation through ERK1/2 and AKT and inhibits apoptosis in breast cancer cells. Peptides 2017; 92:9-15. [PMID: 28442350 DOI: 10.1016/j.peptides.2017.04.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 04/12/2017] [Accepted: 04/19/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Visfatin is a novel adipokine and proinflammatory cytokine which is implicated in breast cancer progression. The exact proliferative and anti-apoptotic mechanisms of visfatin are still under debate. In this study, the effect of extracellular visfatin on proliferation and apoptosis of breast cancer cells were investigated considering key regulatory molecules in these procedures. METHODS BrdU (Bromodeoxyuridine) experiment was used to assess cell proliferation in response to visfatin treatment. Cell viability and apoptosis were assessed using MTT assay and flowcytometry, respectively. Phosphorylation levels of AKT and ERK1/2 as well as survivin levels and Poly ADP ribose polymerase (PARP) cleavage were investigated by western blot analysis. RESULTS Visfatin induced proliferation of MCF-7 and MDA-MB-231 cells, an effect that was repressed by using AKT and ERK1/2 inhibitors, indicating involvement of these two signaling pathways in the proliferative effect of visfatin. Similarly, phosphorylation of AKT and ERK1/2 were elevated by visfatin treatment. On the other hand, visfatin improved cell viability and prevented TNF-α-induced apoptosis as well as PARP cleavage. Visfatin also exerted a protective effect on survivin. CONCLUSION The results of this study suggest that visfatin induces breast cancer cell proliferation through AKT/PI3K and ERK/MAPK activation and protects against apoptosis in these cells. Thus increased visfatin levels may augment breast cancer development and attenuate treatment efficiency in breast cancer patients.
Collapse
Affiliation(s)
- Zafar Gholinejad
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nejat Kheiripour
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular - Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Davod Ilbeigi
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiarash Behroozfar
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Hesari
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Golestani
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Shabani
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Einollahi
- Department of Clinical Laboratory Sciences, Faculty of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Carbone F, Liberale L, Bonaventura A, Vecchiè A, Casula M, Cea M, Monacelli F, Caffa I, Bruzzone S, Montecucco F, Nencioni A. Regulation and Function of Extracellular Nicotinamide Phosphoribosyltransferase/Visfatin. Compr Physiol 2017; 7:603-621. [PMID: 28333382 DOI: 10.1002/cphy.c160029] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is an adipokine-enzyme, which was described as to play bioactivities both in the intracellular and in the extracellular environment. However, while the functions of intracellular NAMPT (iNAMPT) are well known, much less is known on extracellular NAMPT (eNAMPT), also called visfatin or pre-B cell colony-enhancing factor. iNAMPT catalyzes the rate-limiting step in the NAD+ biosynthesis pathway from nicotinamide. Its inhibition severely reduces intracellular NAD+ levels, achieving anti-inflammatory and anti-cancer effects. eNAMPT can be detected in the human circulation and in many extracellular environments. Studies show that eNAMPT can act as a growth factor, as an enzyme, and as a cytokine, but its true mechanism of secretion and its physiological functions are still debated. Increased levels of eNAMPT have been associated with different metabolic disorders and cancers. eNAMPT was demonstrated to modulate the pathways involved in the pathophysiology of obesity, diabetes, atherosclerosis, and cardiovascular events by regulating the oxidative stress response, apoptosis, and inflammation. In cancer, eNAMPT was shown to play a pivotal role in modulating cancer cell metabolism, in promoting epithelial-to-mesenchymal transition and in shaping the tumor microenvironment. In line with these functions, circulating eNAMPT levels are frequently increased in cancer patients. Given these pleiotropic roles of eNAMPT in human disease, this protein has attracted attention as a therapeutic target. In this narrative review, we will discuss recent evidence on eNAMPT-driven signalling, highlighting the emerging pathophysiological roles of this protein in different disorders and the potential therapeutic opportunities linked to its targeting. © 2017 American Physiological Society. Compr Physiol 7:603-621, 2017.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Alessandra Vecchiè
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Matteo Casula
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Michele Cea
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS AOU San Martino-IST, Genoa, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS AOU San Martino-IST, Genoa, Italy
| | - Irene Caffa
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS AOU San Martino-IST, Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Alessio Nencioni
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS AOU San Martino-IST, Genoa, Italy
| |
Collapse
|
38
|
Garten A, Schuster S, Penke M. Could NAMPT inhibition become a potential treatment option in hepatocellular carcinoma? Expert Rev Anticancer Ther 2017; 17:289-291. [PMID: 28271737 DOI: 10.1080/14737140.2017.1298447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Antje Garten
- a Institute of Metabolism and Systems Research, College of Medical and Dental Sciences , University of Birmingham , Birmingham , UK.,c Center for Pediatric Research, Hospital for Children and Adolescents , Leipzig University , Leipzig , Germany
| | - Susanne Schuster
- b Department of Pediatrics , University of California San Diego , La Jolla , CA , USA.,c Center for Pediatric Research, Hospital for Children and Adolescents , Leipzig University , Leipzig , Germany
| | - Melanie Penke
- c Center for Pediatric Research, Hospital for Children and Adolescents , Leipzig University , Leipzig , Germany
| |
Collapse
|
39
|
Dalamaga M, Christodoulatos GS. Visfatin, Obesity, and Cancer. ADIPOCYTOKINES, ENERGY BALANCE, AND CANCER 2017. [DOI: 10.1007/978-3-319-41677-9_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
40
|
Wang Y, Gao C, Zhang Y, Gao J, Teng F, Tian W, Yang W, Yan Y, Xue F. Visfatin stimulates endometrial cancer cell proliferation via activation of PI3K/Akt and MAPK/ERK1/2 signalling pathways. Gynecol Oncol 2016; 143:168-178. [PMID: 27473926 DOI: 10.1016/j.ygyno.2016.07.109] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/18/2016] [Accepted: 07/21/2016] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Endometrial carcinoma is one of the most common malignancies of the female reproductive system, but the aetiology and pathogenesis are not well understood, although adipokines such as visfatin may be involved. Our study provides insight into the mechanism underlying the tumorigenic effects of visfatin in endometrial carcinoma. METHODS We investigated the effect of visfatin on endometrial carcinoma cell proliferation, cell cycle, and apoptosis using well-differentiated Ishikawa cells and poorly differentiated KLE cells. We also assessed the effect of visfatin on tumour growth in vivo. RESULTS Visfatin stimulated the proliferation of both Ishikawa and KLE cells, and visfatin treatment promoted G1/S phase progression and inhibited endometrial carcinoma cell apoptosis. Visfatin promoted endometrial carcinoma tumour growth in BALB/c-nu mice. Transplanted tumour tissues from an endometrial carcinoma mouse model were analysed using immunohistochemical staining, which revealed much stronger positive signals for Ki-67 with over-abundant visfatin. Western blot analysis revealed that insulin receptor (IR), insulin receptor substrate (IRS)1/2 and key components of the phosphoinositide 3-kinase (PI3K)/AKT and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK)1/2 signalling pathways were highly expressed in endometrial carcinoma cells exposed to visfatin. Treated cells showed increased C-MYC and cyclin D1 and reduced caspase-3 expression. The effects of visfatin on proliferation and apoptosis were abrogated by treatment with the PI3K inhibitor LY294002 and MEK inhibitor U0126. CONCLUSIONS Visfatin promotes the malignant progression of endometrial carcinoma via activation of IR and PI3K/Akt and MAPK/ERK signalling. Visfatin may serve as a therapeutic target in the treatment of endometrial carcinoma.
Collapse
Affiliation(s)
- Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yanfang Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinping Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fei Teng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenyan Tian
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wen Yang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Ye Yan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
41
|
Grolla AA, Travelli C, Genazzani AA, Sethi JK. Extracellular nicotinamide phosphoribosyltransferase, a new cancer metabokine. Br J Pharmacol 2016; 173:2182-94. [PMID: 27128025 PMCID: PMC4919578 DOI: 10.1111/bph.13505] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/14/2016] [Accepted: 04/15/2016] [Indexed: 12/12/2022] Open
Abstract
In this review, we focus on the secreted form of nicotinamide phosphoribosyltransferase (NAMPT); extracellular NAMPT (eNAMPT), also known as pre-B cell colony-enhancing factor or visfatin. Although intracellular NAMPT is a key enzyme in controlling NAD metabolism, eNAMPT has been reported to function as a cytokine, with many roles in physiology and pathology. Circulating eNAMPT has been associated with several metabolic and inflammatory disorders, including cancer. Because cytokines produced in the tumour micro-environment play an important role in cancer pathogenesis, in part by reprogramming cellular metabolism, future improvements in cancer immunotherapy will require a better understanding of the crosstalk between cytokine action and tumour biology. In this review, the knowledge of eNAMPT in cancer will be discussed, focusing on its immunometabolic function as a metabokine, its secretion, its mechanism of action and possible roles in the cancer micro-environment.
Collapse
Affiliation(s)
- Ambra A Grolla
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale, Novara, Italy
| | - Cristina Travelli
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale, Novara, Italy
| | - Armando A Genazzani
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale, Novara, Italy
| | - Jaswinder K Sethi
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
42
|
Yang J, Zhang K, Song H, Wu M, Li J, Yong Z, Jiang S, Kuang X, Zhang T. Visfatin is involved in promotion of colorectal carcinoma malignancy through an inducing EMT mechanism. Oncotarget 2016; 7:32306-17. [PMID: 27058759 PMCID: PMC5078014 DOI: 10.18632/oncotarget.8615] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/28/2016] [Indexed: 02/05/2023] Open
Abstract
Increasing evidences suggested visfatin, a newly discovered obesity-induced adipocytokine, is involved in promotion of cancer malignancy and correlated with worse clinical prognosis. While its effects and mechanisms on progression of colorectal cancer (CRC) remain unclear. Our clinical data show that visfatin protein is over expressed, positive associated with lymph node metastasis, high-grade tumor, and poor prognosis in 87 CRC patients. The levels of plasma visfatin are significantly upregulated in Stage IV colon cancer. Visfatin can significantly promote the in vitro migration and invasion of CRC cells via induction epithelial mesenchymal transition (EMT). It can increase the expression and nuclear translocation of Snail, a key transcription factor in regulating EMT. While silencing of Snail attenuates visfatin induced EMT. Further studies reveal visfatin can inhibit the association of Snail with GSK-3β and subsequently suppress ubiquitylation of Snail. In addition, visfatin can increase the expression and nuclear translocation of β-catenin, elevate its binding with Snail promoter, and then increase the transcription of Snail. While inhibitor of PI3K/Akt, LY294002, abolishes visfatin induced up regulation of Snail, Vimentin (Vim), β-catenin, and phosphorylated GSK-3β. In summary, our data suggest that increased expression of visfatin are associated with a more aggressive phenotype of CRC patients. It can trigger the EMT of CRC cells via Akt/GSK-3β/β-catenin signals.
Collapse
Affiliation(s)
- Jing Yang
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Kun Zhang
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Haixing Song
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Mingbo Wu
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Jingyi Li
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Ziyi Yong
- School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Sheng Jiang
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Xi Kuang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Sichuan, China
| | - Tao Zhang
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| |
Collapse
|
43
|
Cabia B, Andrade S, Carreira MC, Casanueva FF, Crujeiras AB. A role for novel adipose tissue-secreted factors in obesity-related carcinogenesis. Obes Rev 2016; 17:361-76. [PMID: 26914773 DOI: 10.1111/obr.12377] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 12/15/2015] [Indexed: 12/12/2022]
Abstract
Obesity, a pandemic disease, is caused by an excessive accumulation of fat that can have detrimental effects on health. Adipose tissue plays a very important endocrine role, secreting different molecules that affect body physiology. In obesity, this function is altered, leading to a dysfunctional production of several factors, known as adipocytokines. This process has been linked to various comorbidities associated with obesity, such as carcinogenesis. In fact, several classical adipocytokines with increased levels in obesity have been demonstrated to exert a pro-carcinogenic role, including leptin, TNF-α, IL-6 and resistin, whereas others like adiponectin, with decreased levels in obesity, might have an anti-carcinogenic function. In this expanding field, new proteomic techniques and approaches have allowed the identification of novel adipocytokines, a number of which exhibit an altered production in obesity and type 2 diabetes and thus are related to adiposity. Many of these novel adipocytokines have also been identified in various tumour types, such as that of the breast, liver or endometrium, thereby increasing the list of potential contributors to carcinogenesis. This review is focused on the regulation of these novel adipocytokines by obesity, including apelin, endotrophin, FABP4, lipocalin 2, omentin-1, visfatin, chemerin, ANGPTL2 or osteopontin, emphasizing its involvement in tumorigenesis.
Collapse
Affiliation(s)
- B Cabia
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| | - S Andrade
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| | - M C Carreira
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| | - F F Casanueva
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| | - A B Crujeiras
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| |
Collapse
|
44
|
Hung AC, Lo S, Hou MF, Lee YC, Tsai CH, Chen YY, Liu W, Su YH, Lo YH, Wang CH, Wu SC, Hsieh YC, Hu SCS, Tai MH, Wang YM, Yuan SSF. Extracellular Visfatin-Promoted Malignant Behavior in Breast Cancer Is Mediated Through c-Abl and STAT3 Activation. Clin Cancer Res 2016; 22:4478-90. [PMID: 27036136 DOI: 10.1158/1078-0432.ccr-15-2704] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 03/22/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Visfatin is an adipocytokine involved in cellular metabolism, inflammation, and cancer. This study investigated the roles of extracellular visfatin in breast cancer, and explored underlying mechanisms in clinical and experimental settings. EXPERIMENTAL DESIGN Associations of serum visfatin with clinicopathologic characteristics and patient survival were assessed with Cox regression models and Kaplan-Meier analyses. Effects of extracellular visfatin on cultured breast cancer cells were examined, followed by in vivo investigation of tumor growth and metastasis in xenograft animal models. Imatinib and Stattic were used to inhibit c-Abl and STAT3 activation, respectively. RESULTS Breast cancer patients with high serum visfatin levels were associated with advanced tumor stage, increased tumor size and lymph node metastasis, and poor survival. Elevated phosphorylation of c-Abl and STAT3 in breast tumor tissues were correlated with high serum visfatin levels in patients. Visfatin-promoted in vitro cell viability and metastatic capability were suppressed by imatinib (c-Abl inhibitor) and Stattic (STAT3 inhibitor). Increased in vivo cell invasiveness was observed in zebrafish xenografted with visfatin-pretreated breast cancer cells. Tumor growth and lung metastasis occurred in visfatin-administered mice xenografted with breast cancer cells. Tail vein-injected mice with visfatin-pretreated breast cancer cells showed increased lung metastasis, which was suppressed by imatinib. CONCLUSIONS Serum visfatin levels in breast cancer patients reveal potential prognostic values, and our findings that visfatin promoted breast cancer through activation of c-Abl and STAT3 may provide an important molecular basis for future design of targeted therapies that take into account different serum visfatin levels in breast cancer. Clin Cancer Res; 22(17); 4478-90. ©2016 AACR.
Collapse
Affiliation(s)
- Amos C Hung
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Steven Lo
- Canniesburn Plastic Surgery Unit, Royal Infirmary, Glasgow, Scotland, United Kingdom
| | - Ming-Feng Hou
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Chen Lee
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan. Department of Anatomy, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Hao Tsai
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan. Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuan-Yin Chen
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wangta Liu
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Han Su
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Hsuan Lo
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chie-Hong Wang
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shiou-Chen Wu
- Department of Biological Science and Technology and Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Ya-Ching Hsieh
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Stephen Chu-Sung Hu
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan. Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Hong Tai
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Yun-Ming Wang
- Department of Biological Science and Technology and Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan.
| | - Shyng-Shiou F Yuan
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan. Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan. Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan. Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
45
|
|
46
|
Grolla AA, Torretta S, Gnemmi I, Amoruso A, Orsomando G, Gatti M, Caldarelli A, Lim D, Penengo L, Brunelleschi S, Genazzani AA, Travelli C. Nicotinamide phosphoribosyltransferase (NAMPT/PBEF/visfatin) is a tumoural cytokine released from melanoma. Pigment Cell Melanoma Res 2015; 28:718-29. [DOI: 10.1111/pcmr.12420] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 09/08/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Ambra A. Grolla
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Simone Torretta
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Ilaria Gnemmi
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Angela Amoruso
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Giuseppe Orsomando
- Section of Biochemistry; Department of Clinical Sciences; Polytechnic University of Marche; Ancona Italy
| | - Marco Gatti
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Antonio Caldarelli
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Lorenza Penengo
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
- Institute of Pharmacology and Toxicology; University of Zürich-Vetsuisse; Zürich Switzerland
| | - Sandra Brunelleschi
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Cristina Travelli
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| |
Collapse
|
47
|
Zhang Y, Zhang J, Liu Z, Liu Y, Tuo S. A network-based approach to identify disease-associated gene modules through integrating DNA methylation and gene expression. Biochem Biophys Res Commun 2015; 465:437-42. [PMID: 26282201 DOI: 10.1016/j.bbrc.2015.08.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 08/09/2015] [Indexed: 11/28/2022]
|
48
|
Garten A, Schuster S, Penke M, Gorski T, de Giorgis T, Kiess W. Physiological and pathophysiological roles of NAMPT and NAD metabolism. Nat Rev Endocrinol 2015. [PMID: 26215259 DOI: 10.1038/nrendo.2015.117] [Citation(s) in RCA: 487] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is a regulator of the intracellular nicotinamide adenine dinucleotide (NAD) pool. NAD is an essential coenzyme involved in cellular redox reactions and is a substrate for NAD-dependent enzymes. In various metabolic disorders and during ageing, levels of NAD are decreased. Through its NAD-biosynthetic activity, NAMPT influences the activity of NAD-dependent enzymes, thereby regulating cellular metabolism. In addition to its enzymatic function, extracellular NAMPT (eNAMPT) has cytokine-like activity. Abnormal levels of eNAMPT are associated with various metabolic disorders. NAMPT is able to modulate processes involved in the pathogenesis of obesity and related disorders such as nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM) by influencing the oxidative stress response, apoptosis, lipid and glucose metabolism, inflammation and insulin resistance. NAMPT also has a crucial role in cancer cell metabolism, is often overexpressed in tumour tissues and is an experimental target for antitumour therapies. In this Review, we discuss current understanding of the functions of NAMPT and highlight progress made in identifying the physiological role of NAMPT and its relevance in various human diseases and conditions, such as obesity, NAFLD, T2DM, cancer and ageing.
Collapse
Affiliation(s)
- Antje Garten
- Center for Pediatric Research Leipzig, Hospital for Children and Adolescents, University of Leipzig, Liebigstrasse 21, 04103 Leipzig, Germany
| | - Susanne Schuster
- Center for Pediatric Research Leipzig, Hospital for Children and Adolescents, University of Leipzig, Liebigstrasse 21, 04103 Leipzig, Germany
| | - Melanie Penke
- Center for Pediatric Research Leipzig, Hospital for Children and Adolescents, University of Leipzig, Liebigstrasse 21, 04103 Leipzig, Germany
| | - Theresa Gorski
- Center for Pediatric Research Leipzig, Hospital for Children and Adolescents, University of Leipzig, Liebigstrasse 21, 04103 Leipzig, Germany
| | - Tommaso de Giorgis
- Center for Pediatric Research Leipzig, Hospital for Children and Adolescents, University of Leipzig, Liebigstrasse 21, 04103 Leipzig, Germany
| | - Wieland Kiess
- Center for Pediatric Research Leipzig, Hospital for Children and Adolescents, University of Leipzig, Liebigstrasse 21, 04103 Leipzig, Germany
| |
Collapse
|
49
|
Lezhnina K, Kovalchuk O, Zhavoronkov AA, Korzinkin MB, Zabolotneva AA, Shegay PV, Sokov DG, Gaifullin NM, Rusakov IG, Aliper AM, Roumiantsev SA, Alekseev BY, Borisov NM, Buzdin AA. Novel robust biomarkers for human bladder cancer based on activation of intracellular signaling pathways. Oncotarget 2015; 5:9022-32. [PMID: 25296972 PMCID: PMC4253415 DOI: 10.18632/oncotarget.2493] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
We recently proposed a new bioinformatic algorithm called OncoFinder for quantifying the activation of intracellular signaling pathways. It was proved advantageous for minimizing errors of high-throughput gene expression analyses and showed strong potential for identifying new biomarkers. Here, for the first time, we applied OncoFinder for normal and cancerous tissues of the human bladder to identify biomarkers of bladder cancer. Using Illumina HT12v4 microarrays, we profiled gene expression in 17 cancer and seven non-cancerous bladder tissue samples. These experiments were done in two independent laboratories located in Russia and Canada. We calculated pathway activation strength values for the investigated transcriptomes and identified signaling pathways that were regulated differently in bladder cancer (BC) tissues compared with normal controls. We found, for both experimental datasets, 44 signaling pathways that serve as excellent new biomarkers of BC, supported by high area under the curve (AUC) values. We conclude that the OncoFinder approach is highly efficient in finding new biomarkers for cancer. These markers are mathematical functions involving multiple gene products, which distinguishes them from “traditional” expression biomarkers that only assess concentrations of single genes.
Collapse
Affiliation(s)
- Ksenia Lezhnina
- Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR. Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, 4401 University Drive, Lethbridge, AB, T1K 3M4. Canada Cancer and Aging Research Laboratories, Lethbridge, AB, Canada
| | - Alexander A Zhavoronkov
- Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia. Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD. Faculty of Biological and Medical Physics, Moscow Institute of Physics and Technology
| | | | - Anastasia A Zabolotneva
- Group for Genomic Regulation of Cell Signaling Systems, Shemyakn-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Peter V Shegay
- P.A. Herzen Moscow Oncological Research Institute, Moscow, Russia
| | | | - Nurshat M Gaifullin
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Moscow, Russia. Russian medical postgraduate academy,Moscow, Russia
| | - Igor G Rusakov
- P.A. Herzen Moscow Oncological Research Institute, Moscow, Russia
| | - Alexander M Aliper
- Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR. Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Sergey A Roumiantsev
- Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Boris Y Alekseev
- P.A. Herzen Moscow Oncological Research Institute, Moscow, Russia
| | - Nikolay M Borisov
- Laboratory of Systems Biology, A.I. Burnasyan Federal Medical Biophysical Center, Moscow, Russia
| | - Anton A Buzdin
- Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR. Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia. Group for Genomic Regulation of Cell Signaling Systems, Shemyakn-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
50
|
Ruggieri S, Orsomando G, Sorci L, Raffaelli N. Regulation of NAD biosynthetic enzymes modulates NAD-sensing processes to shape mammalian cell physiology under varying biological cues. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1138-49. [PMID: 25770681 DOI: 10.1016/j.bbapap.2015.02.021] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 12/25/2022]
Abstract
In addition to its role as a redox coenzyme, NAD is a substrate of various enzymes that split the molecule to either catalyze covalent modifications of target proteins or convert NAD into biologically active metabolites. The coenzyme bioavailability may be significantly affected by these reactions, with ensuing major impact on energy metabolism, cell survival, and aging. Moreover, through the activity of the NAD-dependent deacetylating sirtuins, NAD behaves as a beacon molecule that reports the cell metabolic state, and accordingly modulates transcriptional responses and metabolic adaptations. In this view, NAD biosynthesis emerges as a highly regulated process: it enables cells to preserve NAD homeostasis in response to significant NAD-consuming events and it can be modulated by various stimuli to induce, via NAD level changes, suitable NAD-mediated metabolic responses. Here we review the current knowledge on the regulation of mammalian NAD biosynthesis, with focus on the relevant rate-limiting enzymes. This article is part of a Special Issue entitled: Cofactor-dependent proteins: evolution, chemical diversity and bio-applications.
Collapse
Affiliation(s)
- Silverio Ruggieri
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Giuseppe Orsomando
- Department of Clinical Sciences, Section of Biochemistry, Polytechnic University of Marche, Ancona, Italy
| | - Leonardo Sorci
- Department of Clinical Sciences, Section of Biochemistry, Polytechnic University of Marche, Ancona, Italy
| | - Nadia Raffaelli
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy.
| |
Collapse
|