1
|
Podyacheva E, Toropova Y. The Role of NAD+, SIRTs Interactions in Stimulating and Counteracting Carcinogenesis. Int J Mol Sci 2023; 24:ijms24097925. [PMID: 37175631 PMCID: PMC10178434 DOI: 10.3390/ijms24097925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
The World Health Organization has identified oncological diseases as one of the most serious health concerns of the current century. Current research on oncogenesis is focused on the molecular mechanisms of energy-biochemical reprogramming in cancer cell metabolism, including processes contributing to the Warburg effect and the pro-oncogenic and anti-oncogenic roles of sirtuins (SIRTs) and poly-(ADP-ribose) polymerases (PARPs). However, a clear understanding of the interaction between NAD+, SIRTs in cancer development, as well as their effects on carcinogenesis, has not been established, and literature data vary greatly. This work aims to provide a summary and structure of the available information on NAD+, SIRTs interactions in both stimulating and countering carcinogenesis, and to discuss potential approaches for pharmacological modulation of these interactions to achieve an anticancer effect.
Collapse
Affiliation(s)
- Ekaterina Podyacheva
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341 Saint-Petersburg, Russia
| | - Yana Toropova
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341 Saint-Petersburg, Russia
| |
Collapse
|
2
|
Anticancer Activities of Novel Nicotinamide Phosphoribosyltransferase Inhibitors in Hematological Malignancies. Molecules 2023; 28:molecules28041897. [PMID: 36838885 PMCID: PMC9967653 DOI: 10.3390/molecules28041897] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/20/2023] [Accepted: 02/02/2023] [Indexed: 02/19/2023] Open
Abstract
Targeting cancer cells that are highly dependent on the nicotinamide adenine dinucleotide (NAD+) metabolite is a promising therapeutic strategy. Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme catalyzing NAD+ production. Despite the high efficacy of several developed NAMPT inhibitors (i.e., FK866 (APO866)) in preclinical studies, their clinical activity was proven to be limited. Here, we report the synthesis of new NAMPT Inhibitors, JJ08, FEI191 and FEI199, which exhibit a broad anticancer activity in vitro. Results show that these compounds are potent NAMPT inhibitors that deplete NAD+ and NADP(H) after 24 h of drug treatment, followed by an increase in reactive oxygen species (ROS) accumulation. The latter event leads to ATP loss and mitochondrial depolarization with induction of apoptosis and necrosis. Supplementation with exogenous NAD+ precursors or catalase (ROS scavenger) abrogates the cell death induced by the new compounds. Finally, in vivo administration of the new NAMPT inhibitors in a mouse xenograft model of human Burkitt lymphoma delays tumor growth and significantly prolongs mouse survival. The most promising results are collected with JJ08, which completely eradicates tumor growth. Collectively, our findings demonstrate the efficient anticancer activity of the new NAMPT inhibitor JJ08 and highlight a strong interest for further evaluation of this compound in hematological malignancies.
Collapse
|
3
|
CD38-Induced Metabolic Dysfunction Primes Multiple Myeloma Cells for NAD +-Lowering Agents. Antioxidants (Basel) 2023; 12:antiox12020494. [PMID: 36830052 PMCID: PMC9952390 DOI: 10.3390/antiox12020494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/18/2023] Open
Abstract
Cancer cells fuel growth and energy demands by increasing their NAD+ biosynthesis dependency, which therefore represents an exploitable vulnerability for anti-cancer strategies. CD38 is a NAD+-degrading enzyme that has become crucial for anti-MM therapies since anti-CD38 monoclonal antibodies represent the backbone for treatment of newly diagnosed and relapsed multiple myeloma patients. Nevertheless, further steps are needed to enable a full exploitation of these strategies, including deeper insights of the mechanisms by which CD38 promotes tumorigenesis and its metabolic additions that could be selectively targeted by therapeutic strategies. Here, we present evidence that CD38 upregulation produces a pervasive intracellular-NAD+ depletion, which impairs mitochondrial fitness and enhances oxidative stress; as result, genetic or pharmacologic approaches that aim to modify CD38 surface-level prime MM cells to NAD+-lowering agents. The molecular mechanism underlying this event is an alteration in mitochondrial dynamics, which decreases mitochondria efficiency and triggers energetic remodeling. Overall, we found that CD38 handling represents an innovative strategy to improve the outcomes of NAD+-lowering agents and provides the rationale for testing these very promising agents in clinical studies involving MM patients.
Collapse
|
4
|
Śniegocka M, Liccardo F, Fazi F, Masciarelli S. Understanding ER homeostasis and the UPR to enhance treatment efficacy of acute myeloid leukemia. Drug Resist Updat 2022; 64:100853. [PMID: 35870226 DOI: 10.1016/j.drup.2022.100853] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Protein biogenesis, maturation and degradation are tightly regulated processes that are governed by a complex network of signaling pathways. The endoplasmic reticulum (ER) is responsible for biosynthesis and maturation of secretory proteins. Circumstances that alter cellular protein homeostasis, determine accumulation of misfolded and unfolded proteins in the ER, a condition defined as ER stress. In case of stress, the ER activates an adaptive response called unfolded protein response (UPR), a series of pathways of major relevance for cancer biology. The UPR plays a preeminent role in adaptation of tumor cells to the harsh conditions that they experience, due to high rates of proliferation, metabolic abnormalities and hostile environment scarce in oxygen and nutrients. Furthermore, the UPR is among the main adaptive cell stress responses contributing to the development of resistance to drugs and chemotherapy. Clinical management of Acute Myeloid Leukemia (AML) has improved significantly in the last decade, thanks to development of molecular targeted therapies. However, the emergence of treatment-resistant clones renders the rate of AML cure dismal. Moreover, different cell populations that constitute the bone marrow niche recently emerged as a main determinant leading to drug resistance. Herein we summarize the most relevant literature regarding the role played by the UPR in expansion of AML and ability to develop drug resistance and we discuss different possible modalities to overturn this adaptive response against leukemia. To this aim, we also describe the interconnection of the UPR with other cellular stress responses regulating protein homeostasis. Finally, we review the newest findings about the crosstalk between AML cells and cells of the bone marrow niche, under physiological conditions and in response to therapies, discussing in particular the importance of the niche in supporting survival of AML cells by favoring protein homeostasis.
Collapse
Affiliation(s)
- Martyna Śniegocka
- Department of Anatomical, Histological, Forensic & Orthopedic Sciences, Section of Histology & Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Francesca Liccardo
- Department of Anatomical, Histological, Forensic & Orthopedic Sciences, Section of Histology & Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic & Orthopedic Sciences, Section of Histology & Medical Embryology, Sapienza University of Rome, Rome, Italy.
| | - Silvia Masciarelli
- Department of Anatomical, Histological, Forensic & Orthopedic Sciences, Section of Histology & Medical Embryology, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
5
|
Subedi A, Liu Q, Ayyathan DM, Sharon D, Cathelin S, Hosseini M, Xu C, Voisin V, Bader GD, D'Alessandro A, Lechman ER, Dick JE, Minden MD, Wang JCY, Chan SM. Nicotinamide phosphoribosyltransferase inhibitors selectively induce apoptosis of AML stem cells by disrupting lipid homeostasis. Cell Stem Cell 2021; 28:1851-1867.e8. [PMID: 34293334 DOI: 10.1016/j.stem.2021.06.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 05/05/2021] [Accepted: 06/22/2021] [Indexed: 12/29/2022]
Abstract
Current treatments for acute myeloid leukemia (AML) are often ineffective in eliminating leukemic stem cells (LSCs), which perpetuate the disease. Here, we performed a metabolic drug screen to identify LSC-specific vulnerabilities and found that nicotinamide phosphoribosyltransferase (NAMPT) inhibitors selectively killed LSCs, while sparing normal hematopoietic stem and progenitor cells. Treatment with KPT-9274, a NAMPT inhibitor, suppressed the conversion of saturated fatty acids to monounsaturated fatty acids, a reaction catalyzed by the stearoyl-CoA desaturase (SCD) enzyme, resulting in apoptosis of AML cells. Transcriptomic analysis of LSCs treated with KPT-9274 revealed an upregulation of sterol regulatory-element binding protein (SREBP)-regulated genes, including SCD, which conferred partial protection against NAMPT inhibitors. Inhibition of SREBP signaling with dipyridamole enhanced the cytotoxicity of KPT-9274 on LSCs in vivo. Our work demonstrates that altered lipid homeostasis plays a key role in NAMPT inhibitor-induced apoptosis and identifies NAMPT inhibition as a therapeutic strategy for targeting LSCs in AML.
Collapse
Affiliation(s)
- Amit Subedi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Qiang Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Dhanoop M Ayyathan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - David Sharon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Severine Cathelin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Mohsen Hosseini
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Changjiang Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada
| | - Veronique Voisin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada
| | - Gary D Bader
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Eric R Lechman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medicine, University of Toronto, ON, Canada; Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON, Canada
| | - Jean C Y Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medicine, University of Toronto, ON, Canada; Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON, Canada
| | - Steven M Chan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medicine, University of Toronto, ON, Canada; Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
6
|
Ghanem MS, Monacelli F, Nencioni A. Advances in NAD-Lowering Agents for Cancer Treatment. Nutrients 2021; 13:1665. [PMID: 34068917 PMCID: PMC8156468 DOI: 10.3390/nu13051665] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is an essential redox cofactor, but it also acts as a substrate for NAD-consuming enzymes, regulating cellular events such as DNA repair and gene expression. Since such processes are fundamental to support cancer cell survival and proliferation, sustained NAD production is a hallmark of many types of neoplasms. Depleting intratumor NAD levels, mainly through interference with the NAD-biosynthetic machinery, has emerged as a promising anti-cancer strategy. NAD can be generated from tryptophan or nicotinic acid. In addition, the "salvage pathway" of NAD production, which uses nicotinamide, a byproduct of NAD degradation, as a substrate, is also widely active in mammalian cells and appears to be highly exploited by a subset of human cancers. In fact, research has mainly focused on inhibiting the key enzyme of the latter NAD production route, nicotinamide phosphoribosyltransferase (NAMPT), leading to the identification of numerous inhibitors, including FK866 and CHS-828. Unfortunately, the clinical activity of these agents proved limited, suggesting that the approaches for targeting NAD production in tumors need to be refined. In this contribution, we highlight the recent advancements in this field, including an overview of the NAD-lowering compounds that have been reported so far and the related in vitro and in vivo studies. We also describe the key NAD-producing pathways and their regulation in cancer cells. Finally, we summarize the approaches that have been explored to optimize the therapeutic response to NAMPT inhibitors in cancer.
Collapse
Affiliation(s)
- Moustafa S. Ghanem
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
7
|
Féral K, Jaud M, Philippe C, Di Bella D, Pyronnet S, Rouault-Pierre K, Mazzolini L, Touriol C. ER Stress and Unfolded Protein Response in Leukemia: Friend, Foe, or Both? Biomolecules 2021; 11:biom11020199. [PMID: 33573353 PMCID: PMC7911881 DOI: 10.3390/biom11020199] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 12/15/2022] Open
Abstract
The unfolded protein response (UPR) is an evolutionarily conserved adaptive signaling pathway triggered by a stress of the endoplasmic reticulum (ER) lumen compartment, which is initiated by the accumulation of unfolded proteins. This response, mediated by three sensors-Inositol Requiring Enzyme 1 (IRE1), Activating Transcription Factor 6 (ATF6), and Protein Kinase RNA-Like Endoplasmic Reticulum Kinase (PERK)—allows restoring protein homeostasis and maintaining cell survival. UPR represents a major cytoprotective signaling network for cancer cells, which frequently experience disturbed proteostasis owing to their rapid proliferation in an usually unfavorable microenvironment. Increased basal UPR also participates in the resistance of tumor cells against chemotherapy. UPR activation also occurs during hematopoiesis, and growing evidence supports the critical cytoprotective role played by ER stress in the emergence and proliferation of leukemic cells. In case of severe or prolonged stress, pro-survival UPR may however evolve into a cell death program called terminal UPR. Interestingly, a large number of studies have revealed that the induction of proapoptotic UPR can also strongly contribute to the sensitization of leukemic cells to chemotherapy. Here, we review the current knowledge on the consequences of the deregulation of UPR signaling in leukemias and their implications for the treatment of these diseases.
Collapse
Affiliation(s)
- Kelly Féral
- Inserm UMR1037-Cancer Research Center of Toulouse, 2 avenue Hubert Curien, Oncopole entrée C, CS 53717, 31037 Toulouse, France; (K.F.); (M.J.); (S.P.)
- Université Toulouse III Paul-Sabatier, F-31000 Toulouse, France
| | - Manon Jaud
- Inserm UMR1037-Cancer Research Center of Toulouse, 2 avenue Hubert Curien, Oncopole entrée C, CS 53717, 31037 Toulouse, France; (K.F.); (M.J.); (S.P.)
- Université Toulouse III Paul-Sabatier, F-31000 Toulouse, France
| | - Céline Philippe
- Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK; (C.P.); (D.D.B.); (K.R.-P.)
| | - Doriana Di Bella
- Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK; (C.P.); (D.D.B.); (K.R.-P.)
| | - Stéphane Pyronnet
- Inserm UMR1037-Cancer Research Center of Toulouse, 2 avenue Hubert Curien, Oncopole entrée C, CS 53717, 31037 Toulouse, France; (K.F.); (M.J.); (S.P.)
- Université Toulouse III Paul-Sabatier, F-31000 Toulouse, France
| | - Kevin Rouault-Pierre
- Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK; (C.P.); (D.D.B.); (K.R.-P.)
| | - Laurent Mazzolini
- Inserm UMR1037-Cancer Research Center of Toulouse, 2 avenue Hubert Curien, Oncopole entrée C, CS 53717, 31037 Toulouse, France; (K.F.); (M.J.); (S.P.)
- CNRS ERL5294, CRCT, F-31037 Toulouse, France
- Correspondence: (L.M.); (C.T.)
| | - Christian Touriol
- Inserm UMR1037-Cancer Research Center of Toulouse, 2 avenue Hubert Curien, Oncopole entrée C, CS 53717, 31037 Toulouse, France; (K.F.); (M.J.); (S.P.)
- Université Toulouse III Paul-Sabatier, F-31000 Toulouse, France
- Correspondence: (L.M.); (C.T.)
| |
Collapse
|
8
|
Polyakova YV, Zavodovsky BV, Sivordova LE, Akhverdyan YR, Zborovskaya IA. Visfatin and Rheumatoid Arthritis: Pathogenetic Implications and Clinical Utility. Curr Rheumatol Rev 2020; 16:224-239. [DOI: 10.2174/1573397115666190409112621] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/15/2019] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
Objective:
Analysis and generalization of data related to visfatin involvement in the
pathogenesis of inflammation at various stages of rheumatoid arthritis.
Data Synthesis:
Visfatin is an adipocytokine which has also been identified in non-adipose tissues.
It influences directly on the maturation of B cells, which are involved in autoantibody production
and T cell activation. Visfatin can promote inflammation via regulation of pro-inflammatory cytokines
including TNF, IL-1β and IL-6. The concentration of circulating visfatin in rheumatoid arthritis
patients is higher compared to healthy individuals. Several studies suggest that visfatin level is
associated with rheumatoid arthritis activity, and its elevation may precede clinical signs of the relapse.
In murine collagen-induced arthritis, visfatin levels were also found to be elevated both in
inflamed synovial cells and in joint vasculature. Visfatin blockers have been shown to confer fast
and long-term attenuation of pathological processes; however, most of their effects are transient.
Other factors responsible for hyperactivation of the immune system can participate in this process
at a later stage. Treatment of rheumatoid arthritis with a combination of these blockers and inhibitors
of other mediators of inflammation can potentially improve treatment outcomes compared to
current therapeutic strategies. Recent advances in the treatment of experimental arthritis in mice as
well as the application of emerging treatment strategies obtained from oncology for rheumatoid arthritis
management could be a source of novel adipokine-mediated anti-rheumatic drugs.
Conclusion:
The ongoing surge of interest in anticytokine therapy makes further study of visfatin
highly relevant as it may serve as a base for innovational RA treatment.
Collapse
Affiliation(s)
- Yulia V. Polyakova
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Boris V. Zavodovsky
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Larisa E. Sivordova
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Yuri R. Akhverdyan
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Irina A. Zborovskaya
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| |
Collapse
|
9
|
Galli U, Colombo G, Travelli C, Tron GC, Genazzani AA, Grolla AA. Recent Advances in NAMPT Inhibitors: A Novel Immunotherapic Strategy. Front Pharmacol 2020; 11:656. [PMID: 32477131 PMCID: PMC7235340 DOI: 10.3389/fphar.2020.00656] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is a cofactor of many enzymatic reactions as well as being a substrate for a number of NAD-consuming enzymes (e.g., PARPS, sirtuins, etc). NAD can be synthesized de novo starting from tryptophan, nicotinamide, nicotinic acid, or nicotinamide riboside from the diet. On the other hand, the nicotinamide that is liberated by NAD-consuming enzymes can be salvaged to re-form NAD. In this former instance, nicotinamide phosphoribosyltransferase (NAMPT) is the bottleneck enzyme. In the many cells in which the salvage pathway is predominant, NAMPT, therefore, represents an important controller of intracellular NAD concentrations, and as a consequence of energy metabolism. It is, therefore, not surprising that NAMPT is over expressed by tumoral cells, which take advantage from this to sustain growth rate and tumor progression. This has led to the initiation of numerous medicinal chemistry programs to develop NAMPT inhibitors in the context of oncology. More recently, however, it has been shown that NAMPT inhibitors do not solely target the tumor but also have an effect on the immune system. To add complexity, this enzyme can also be secreted by cells, and in the extracellular space it acts as a cytokine mainly through the activation of Toll like Receptor 4 (TLR4), although it has not been clarified yet if this is the only receptor responsible for its actions. While specific small molecules have been developed only against the intracellular form of NAMPT, growing evidences sustain the possibility to target the extracellular form. In this contribution, the most recent evidences on the medicinal chemistry of NAMPT will be reviewed, together with the key elements that sustain the hypothesis of NAMPT targeting and the drawbacks so far encountered.
Collapse
Affiliation(s)
- Ubaldina Galli
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Giorgia Colombo
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Cristina Travelli
- Department of Pharmaceutical Sciences, University of Pavia, Pavia, Italy
| | - Gian Cesare Tron
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Ambra A Grolla
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
10
|
Selective targeting of NAMPT by KPT-9274 in acute myeloid leukemia. Blood Adv 2020; 3:242-255. [PMID: 30692102 DOI: 10.1182/bloodadvances.2018024182] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 12/06/2018] [Indexed: 12/30/2022] Open
Abstract
Treatment options for acute myeloid leukemia (AML) remain extremely limited and associated with significant toxicity. Nicotinamide phosphoribosyltransferase (NAMPT) is involved in the generation of NAD+ and a potential therapeutic target in AML. We evaluated the effect of KPT-9274, a p21-activated kinase 4/NAMPT inhibitor that possesses a unique NAMPT-binding profile based on in silico modeling compared with earlier compounds pursued against this target. KPT-9274 elicited loss of mitochondrial respiration and glycolysis and induced apoptosis in AML subtypes independent of mutations and genomic abnormalities. These actions occurred mainly through the depletion of NAD+, whereas genetic knockdown of p21-activated kinase 4 did not induce cytotoxicity in AML cell lines or influence the cytotoxic effect of KPT-9274. KPT-9274 exposure reduced colony formation, increased blast differentiation, and diminished the frequency of leukemia-initiating cells from primary AML samples; KPT-9274 was minimally cytotoxic toward normal hematopoietic or immune cells. In addition, KPT-9274 improved overall survival in vivo in 2 different mouse models of AML and reduced tumor development in a patient-derived xenograft model of AML. Overall, KPT-9274 exhibited broad preclinical activity across a variety of AML subtypes and warrants further investigation as a potential therapeutic agent for AML.
Collapse
|
11
|
Pramono AA, Rather GM, Herman H, Lestari K, Bertino JR. NAD- and NADPH-Contributing Enzymes as Therapeutic Targets in Cancer: An Overview. Biomolecules 2020; 10:biom10030358. [PMID: 32111066 PMCID: PMC7175141 DOI: 10.3390/biom10030358] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 12/14/2022] Open
Abstract
Actively proliferating cancer cells require sufficient amount of NADH and NADPH for biogenesis and to protect cells from the detrimental effect of reactive oxygen species. As both normal and cancer cells share the same NAD biosynthetic and metabolic pathways, selectively lowering levels of NAD(H) and NADPH would be a promising strategy for cancer treatment. Targeting nicotinamide phosphoribosyltransferase (NAMPT), a rate limiting enzyme of the NAD salvage pathway, affects the NAD and NADPH pool. Similarly, lowering NADPH by mutant isocitrate dehydrogenase 1/2 (IDH1/2) which produces D-2-hydroxyglutarate (D-2HG), an oncometabolite that downregulates nicotinate phosphoribosyltransferase (NAPRT) via hypermethylation on the promoter region, results in epigenetic regulation. NADPH is used to generate D-2HG, and is also needed to protect dihydrofolate reductase, the target for methotrexate, from degradation. NAD and NADPH pools in various cancer types are regulated by several metabolic enzymes, including methylenetetrahydrofolate dehydrogenase, serine hydroxymethyltransferase, and aldehyde dehydrogenase. Thus, targeting NAD and NADPH synthesis under special circumstances is a novel approach to treat some cancers. This article provides the rationale for targeting the key enzymes that maintain the NAD/NADPH pool, and reviews preclinical studies of targeting these enzymes in cancers.
Collapse
Affiliation(s)
- Alvinsyah Adhityo Pramono
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; (A.A.P.); (G.M.R.)
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Gulam M. Rather
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; (A.A.P.); (G.M.R.)
| | - Herry Herman
- Division of Oncology, Department of Orthopaedic Surgery, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, Indonesia;
| | - Keri Lestari
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Joseph R. Bertino
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; (A.A.P.); (G.M.R.)
- Department of Pharmacology and Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Correspondence: ; Tel.: +1-(732)-235-8510
| |
Collapse
|
12
|
Liu Y, Tian L, Li Y, Chen Y, Chen Y, Liu L, Wang S. Photoactive Oligo( p-phenylene vinylene) Material for Functional Regulation of Induced Pluripotent Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2020; 12:3438-3444. [PMID: 31877011 DOI: 10.1021/acsami.9b19331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In recent years, rapid development of induced pluripotent stem cell (iPSC) technology has provided good technical support for the study of human cardiovascular disease (CVD). In this work, a mimetic cell membrane and drug carrier OPFL system containing photoactive oligo(p-phenylene vinylene) functionalized with phospholipid units (OPV-lipid) was prepared for functional regulation of iPSC-derived cardiomyocytes. OPFL bound to the cell membrane of iPSC-derived human cardiomyocytes and significantly enhanced delivery of cyclosporin A (CsA) into these cells, which promoted the regulation of mitochondrial calcium levels and membrane potential by CsA. This led to the protection of the mitochondrial structure and function, thus reducing apoptosis of iPSC-derived cardiomyocytes and achieving the effect of treating CVD. OPFL not only acts as a fluorescent probe for cell imaging and visualization of the drug delivery process but also provides a tool to deliver lipid-insoluble drugs throughout the cell membrane. Benefiting from good biocompatibility, facile operation, and a visible and controllable cell uptake process, OPFL has good potential to be a powerful tool in future basic and clinical research applications involving iPSCs.
Collapse
Affiliation(s)
- Yuqi Liu
- Cardiac Department, National Key Laboratory of Kidney Diseases, Department of Cardiology & National Clinical Research Center of Geriatric Disease, Beijing Key Laboratory of Chronic Heart Failure Precision Medicine , First Center of Chinese PLA General Hospital , Beijing 100853 , China
| | - Liuyang Tian
- Cardiac Department, National Key Laboratory of Kidney Diseases, Department of Cardiology & National Clinical Research Center of Geriatric Disease, Beijing Key Laboratory of Chronic Heart Failure Precision Medicine , First Center of Chinese PLA General Hospital , Beijing 100853 , China
- Department of Cardiology , Tianjin Union Medical Center , Tianjin 300121 , China
| | - Yang Li
- Cardiac Department, National Key Laboratory of Kidney Diseases, Department of Cardiology & National Clinical Research Center of Geriatric Disease, Beijing Key Laboratory of Chronic Heart Failure Precision Medicine , First Center of Chinese PLA General Hospital , Beijing 100853 , China
| | - Yanyan Chen
- Key Laboratory of Organic Solids, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Yundai Chen
- Cardiac Department, National Key Laboratory of Kidney Diseases, Department of Cardiology & National Clinical Research Center of Geriatric Disease, Beijing Key Laboratory of Chronic Heart Failure Precision Medicine , First Center of Chinese PLA General Hospital , Beijing 100853 , China
| | - Libing Liu
- Key Laboratory of Organic Solids, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Shu Wang
- Key Laboratory of Organic Solids, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , P. R. China
| |
Collapse
|
13
|
Heske CM. Beyond Energy Metabolism: Exploiting the Additional Roles of NAMPT for Cancer Therapy. Front Oncol 2020; 9:1514. [PMID: 32010616 PMCID: PMC6978772 DOI: 10.3389/fonc.2019.01514] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
Tumor cells have increased requirements for NAD+. Thus, many cancers exhibit an increased reliance on NAD+ production pathways. This dependence may be exploited therapeutically through pharmacological targeting of NAMPT, the rate-limiting enzyme in the NAD+ salvage pathway. Despite promising preclinical data using NAMPT inhibitors in cancer models, early NAMPT inhibitors showed limited efficacy in several early phase clinical trials, necessitating the identification of strategies, such as drug combinations, to enhance their efficacy. While the effect of NAMPT inhibitors on impairment of energy metabolism in cancer cells has been well-described, more recent insights have uncovered a number of additional targetable cellular processes that are impacted by inhibition of NAMPT. These include sirtuin function, DNA repair machinery, redox homeostasis, molecular signaling, cellular stemness, and immune processes. This review highlights the recent findings describing the effects of NAMPT inhibitors on the non-metabolic functions of malignant cells, with a focus on how this information can be leveraged clinically. Combining NAMPT inhibitors with other therapies that target NAD+-dependent processes or selecting tumors with specific vulnerabilities that can be co-targeted with NAMPT inhibitors may represent opportunities to exploit the multiple functions of this enzyme for greater therapeutic benefit.
Collapse
Affiliation(s)
- Christine M Heske
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
14
|
Oita RC, Camp SM, Ma W, Ceco E, Harbeck M, Singleton P, Messana J, Sun X, Wang T, Garcia JGN. Novel Mechanism for Nicotinamide Phosphoribosyltransferase Inhibition of TNF-α-mediated Apoptosis in Human Lung Endothelial Cells. Am J Respir Cell Mol Biol 2019; 59:36-44. [PMID: 29337590 DOI: 10.1165/rcmb.2017-0155oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) exists as both intracellular NAMPT and extracellular NAMPT (eNAMPT) proteins. eNAMPT is secreted into the blood and functions as a cytokine/enzyme (cytozyme) that activates NF-κB signaling via ligation of Toll-like receptor 4 (TLR4), further serving as a biomarker for inflammatory lung disorders such as acute respiratory distress syndrome. In contrast, intracellular NAMPT is involved in nicotinamide mononucleotide synthesis and has been implicated in the regulation of cellular apoptosis, although the exact mechanisms for this regulation are poorly understood. We examined the role of NAMPT in TNF-α-induced human lung endothelial cell (EC) apoptosis and demonstrated that reduced NAMPT expression (siRNA) increases EC susceptibility to TNF-α-induced apoptosis as reflected by PARP-1 cleavage and caspase-3 activation. In contrast, overexpression of NAMPT served to reduce degrees of TNF-α-induced EC apoptosis. Inhibition of nicotinamide mononucleotide synthesis by FK866 (a selective NAMPT enzymatic inhibitor) failed to alter TNF-α-induced human lung EC apoptosis, suggesting that NAMPT-dependent NAD+ generation is unlikely to be involved in regulation of TNF-α-induced EC apoptosis. We next confirmed that TNF-α-induced EC apoptosis is attributable to NAMPT secretion into the EC culture media and subsequent eNAMPT ligation of TLR4 on the EC membrane surface. Silencing of NAMPT expression, direct neutralization of secreted eNAMPT by an NAMPT-specific polyclonal antibody (preventing TLR4 ligation), or direct TLR4 antagonism all served to significantly increase EC susceptibility to TNF-α-induced EC apoptosis. Together, these studies provide novel insights into NAMPT contributions to lung inflammatory events and to novel mechanisms of EC apoptosis regulation.
Collapse
Affiliation(s)
- Radu C Oita
- 1 Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona Health Sciences, University of Arizona, Tucson, Arizona
| | - Sara M Camp
- 1 Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona Health Sciences, University of Arizona, Tucson, Arizona
| | - Wenli Ma
- 1 Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona Health Sciences, University of Arizona, Tucson, Arizona
| | - Ermelinda Ceco
- 2 Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Mark Harbeck
- 2 Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | | | - Joe Messana
- 1 Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona Health Sciences, University of Arizona, Tucson, Arizona
| | - Xiaoguang Sun
- 1 Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona Health Sciences, University of Arizona, Tucson, Arizona
| | - Ting Wang
- 1 Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona Health Sciences, University of Arizona, Tucson, Arizona
| | - Joe G N Garcia
- 1 Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona Health Sciences, University of Arizona, Tucson, Arizona
| |
Collapse
|
15
|
Demarest TG, Babbar M, Okur MN, Dan X, Croteau DL, Fakouri NB, Mattson MP, Bohr VA. NAD+Metabolism in Aging and Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2019. [DOI: 10.1146/annurev-cancerbio-030518-055905] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aging is a major risk factor for many types of cancer, and the molecular mechanisms implicated in aging, progeria syndromes, and cancer pathogenesis display considerable similarities. Maintaining redox homeostasis, efficient signal transduction, and mitochondrial metabolism is essential for genome integrity and for preventing progression to cellular senescence or tumorigenesis. NAD+is a central signaling molecule involved in these and other cellular processes implicated in age-related diseases and cancer. Growing evidence implicates NAD+decline as a major feature of accelerated aging progeria syndromes and normal aging. Administration of NAD+precursors such as nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN) offer promising therapeutic strategies to improve health, progeria comorbidities, and cancer therapies. This review summarizes insights from the study of aging and progeria syndromes and discusses the implications and therapeutic potential of the underlying molecular mechanisms involved in aging and how they may contribute to tumorigenesis.
Collapse
Affiliation(s)
- Tyler G. Demarest
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Mansi Babbar
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Mustafa N. Okur
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Xiuli Dan
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Deborah L. Croteau
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Nima B. Fakouri
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Vilhelm A. Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| |
Collapse
|
16
|
Overexpression of miR-182 inhibits ossification of ligamentum flavum cells by targeting NAMPT. Exp Cell Res 2018; 367:119-131. [DOI: 10.1016/j.yexcr.2018.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 12/31/2022]
|
17
|
Grohmann T, Penke M, Petzold-Quinque S, Schuster S, Richter S, Kiess W, Garten A. Inhibition of NAMPT sensitizes MOLT4 leukemia cells for etoposide treatment through the SIRT2-p53 pathway. Leuk Res 2018; 69:39-46. [PMID: 29653431 DOI: 10.1016/j.leukres.2018.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 03/31/2018] [Accepted: 04/04/2018] [Indexed: 10/17/2022]
Abstract
NAMPT (Nicotinamide phosphoribosyltransferase) catalyses the rate-limiting step in the NAD biosynthesis from nicotinamide and thereby regulates the activity of NAD-dependent enzymes. Cancer cells are highly dependent on NAD for energy and DNA repair processes and are assumed to be more susceptible to an inhibition of NAD synthesis than non-transformed cells. We aimed to investigate whether or not inhibition of NAMPT with its specific inhibitor FK866 can sensitize leukemia cells for chemotherapeutic agents. NAMPT protein abundance, enzymatic activity and NAD concentrations were significantly higher in Jurkat and Molt-4 leukemia cell lines compared to normal peripheral blood mononuclear cells. Combination of etoposide and FK866 caused increased cell death in leukemia cell lines compared to etoposide alone. Etoposide decreased protein abundance of NAD-dependent deacetylases SIRTUIN1. After combining etoposide and FK866 treatment SIRTUIN2 was further decreased and accumulation and acetylation of the downstream target p53 was further enhanced in MOLT4 cells. Concomitantly, protein abundance of p21 and cleaved BAX was increased. Targeting NAMPT could be a novel therapeutic strategy to enhance the efficacy of chemotherapeutic agents such as etoposide against leukemia.
Collapse
Affiliation(s)
- Theresa Grohmann
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany
| | - Melanie Penke
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany
| | - Stefanie Petzold-Quinque
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany
| | - Susanne Schuster
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany
| | - Sandy Richter
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany
| | - Wieland Kiess
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany
| | - Antje Garten
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany; University of Birmingham, Institute of Metabolism and Systems Research (IMSR), Birmingham, UK.
| |
Collapse
|
18
|
Thongon N, Zucal C, D'Agostino VG, Tebaldi T, Ravera S, Zamporlini F, Piacente F, Moschoi R, Raffaelli N, Quattrone A, Nencioni A, Peyron JF, Provenzani A. Cancer cell metabolic plasticity allows resistance to NAMPT inhibition but invariably induces dependence on LDHA. Cancer Metab 2018. [PMID: 29541451 PMCID: PMC5844108 DOI: 10.1186/s40170-018-0174-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Inhibitors of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in NAD+ biosynthesis from nicotinamide, exhibit anticancer effects in preclinical models. However, continuous exposure to NAMPT inhibitors, such as FK866, can induce acquired resistance. Methods We developed FK866-resistant CCRF-CEM (T cell acute lymphoblastic leukemia) and MDA MB231 (breast cancer) models, and by exploiting an integrated approach based on genetic, biochemical, and genome wide analyses, we annotated the drug resistance mechanisms. Results Acquired resistance to FK866 was independent of NAMPT mutations but rather was based on a shift towards a glycolytic metabolism and on lactate dehydrogenase A (LDHA) activity. In addition, resistant CCRF-CEM cells, which exhibit high quinolinate phosphoribosyltransferase (QPRT) activity, also exploited amino acid catabolism as an alternative source for NAD+ production, becoming addicted to tryptophan and glutamine and sensitive to treatment with the amino acid transport inhibitor JPH203 and with l-asparaginase, which affects glutamine exploitation. Vice versa, in line with their low QPRT expression, FK866-resistant MDA MB231 did not rely on amino acids for their resistance phenotype. Conclusions Our study identifies novel mechanisms of resistance to NAMPT inhibition, which may be useful to design more rational strategies for targeting cancer metabolism.
Collapse
Affiliation(s)
- Natthakan Thongon
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| | - Chiara Zucal
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| | | | - Toma Tebaldi
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| | - Silvia Ravera
- 2Department of Pharmacy, Biochemistry Laboratory, University of Genova, Genova, Italy
| | - Federica Zamporlini
- 3Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | | | - Ruxanda Moschoi
- 5Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM U1065, Nice, France
| | - Nadia Raffaelli
- 3Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Alessandro Quattrone
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| | - Alessio Nencioni
- 4Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Jean-Francois Peyron
- 5Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM U1065, Nice, France
| | - Alessandro Provenzani
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| |
Collapse
|
19
|
Targeting Mitochondrial Bioenergetics as a Therapeutic Strategy for Chronic Lymphocytic Leukemia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2426712. [PMID: 29682155 PMCID: PMC5851432 DOI: 10.1155/2018/2426712] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 11/06/2017] [Indexed: 12/31/2022]
Abstract
Altered cellular metabolism is considered a hallmark of cancer and is fast becoming an avenue for therapeutic intervention. Mitochondria have recently been viewed as an important cellular compartment that fuels the metabolic demands of cancer cells. Mitochondria are the major source of ATP and metabolites necessary to fulfill the bioenergetics and biosynthetic demands of cancer cells. Furthermore, mitochondria are central to cell death and the main source for generation of reactive oxygen species (ROS). Overall, the growing evidence now suggests that mitochondrial bioenergetics, biogenesis, ROS production, and adaptation to intrinsic oxidative stress are elevated in chronic lymphocytic leukemia (CLL). Hence, recent studies have shown that mitochondrial metabolism could be targeted for cancer therapy. This review focuses the recent advancements in targeting mitochondrial metabolism for the treatment of CLL.
Collapse
|
20
|
Interplay between P-Glycoprotein Expression and Resistance to Endoplasmic Reticulum Stressors. Molecules 2018; 23:molecules23020337. [PMID: 29415493 PMCID: PMC6017601 DOI: 10.3390/molecules23020337] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/13/2022] Open
Abstract
Multidrug resistance (MDR) is a phenotype of cancer cells with reduced sensitivity to a wide range of unrelated drugs. P-glycoprotein (P-gp)—a drug efflux pump (ABCB1 member of the ABC transporter gene family)—is frequently observed to be a molecular cause of MDR. The drug-efflux activity of P-gp is considered as the underlying mechanism of drug resistance against P-gp substrates and results in failure of cancer chemotherapy. Several pathological impulses such as shortages of oxygen and glucose supply, alterations of calcium storage mechanisms and/or processes of protein N-glycosylation in the endoplasmic reticulum (ER) leads to ER stress (ERS), characterized by elevation of unfolded protein cell content and activation of the unfolded protein response (UPR). UPR is responsible for modification of protein folding pathways, removal of misfolded proteins by ER associated protein degradation (ERAD) and inhibition of proteosynthesis. However, sustained ERS may result in UPR-mediated cell death. Neoplastic cells could escape from the death pathway induced by ERS by switching UPR into pro survival mechanisms instead of apoptosis. Here, we aimed to present state of the art information about consequences of P-gp expression on mechanisms associated with ERS development and regulation of the ERAD system, particularly focused on advances in ERS-associated therapy of drug resistant malignancies.
Collapse
|
21
|
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: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
22
|
Inhibitor of Nicotinamide Phosphoribosyltransferase Sensitizes Glioblastoma Cells to Temozolomide via Activating ROS/JNK Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1450843. [PMID: 28097126 PMCID: PMC5206411 DOI: 10.1155/2016/1450843] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/10/2016] [Indexed: 12/16/2022]
Abstract
Overcoming temozolomide (TMZ) resistance is a great challenge in glioblastoma (GBM) treatment. Nicotinamide phosphoribosyltransferase (NAMPT) is a rate-limiting enzyme in the biosynthesis of nicotinamide adenine dinucleotide and has a crucial role in cancer cell metabolism. In this study, we investigated whether FK866 and CHS828, two specific NAMPT inhibitors, could sensitize GBM cells to TMZ. Low doses of FK866 and CHS828 (5 nM and 10 nM, resp.) alone did not significantly decrease cell viability in U251-MG and T98 GBM cells. However, they significantly increased the antitumor action of TMZ in these cells. In U251-MG cells, administration of NAMPT inhibitors increased the TMZ (100 μM)-induced apoptosis and LDH release from GBM cells. NAMPT inhibitors remarkably enhanced the activities of caspase-1, caspase-3, and caspase-9. Moreover, NAMPT inhibitors increased reactive oxygen species (ROS) production and superoxide anion level but reduced the SOD activity and total antioxidative capacity in GBM cells. Treatment of NAMPT inhibitors increased phosphorylation of c-Jun and JNK. Administration of JNK inhibitor SP600125 or ROS scavenger tocopherol with TMZ and NAMPT inhibitors substantially attenuated the sensitization of NAMPT inhibitor on TMZ antitumor action. Our data indicate a potential value of NAMPT inhibitors in combined use with TMZ for GBM treatment.
Collapse
|
23
|
Sociali G, Raffaghello L, Magnone M, Zamporlini F, Emionite L, Sturla L, Bianchi G, Vigliarolo T, Nahimana A, Nencioni A, Raffaelli N, Bruzzone S. Antitumor effect of combined NAMPT and CD73 inhibition in an ovarian cancer model. Oncotarget 2016; 7:2968-84. [PMID: 26658104 PMCID: PMC4823084 DOI: 10.18632/oncotarget.6502] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/16/2015] [Indexed: 12/02/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is a crucial enzyme in the biosynthesis of intracellular NAD+. NAMPT inhibitors have potent anticancer activity in several preclinical models by depleting NAD+ and ATP levels. Recently, we demonstrated that CD73 enables the utilization of extracellular NAD+/nicotinamide mononucleotide (NMN) by converting them to Nicotinamide riboside (NR), which can cross the plasmamembrane and fuel intracellular NAD+ biosynthesis in human cells. These processes are herein confirmed to also occur in a human ovarian carcinoma cell line (OVCAR-3), by means of CD73 or NRK1 specific silencing. Next, we investigated the anti-tumor activity of the simultaneous inhibition of NAMPT (with FK866) and CD73 (with α, β-methylene adenosine 5′-diphosphate, APCP), in an in vivo human ovarian carcinoma model. Interestingly, the combined therapy was found to significantly decrease intratumor NAD+, NMN and ATP levels, compared with single treatments. In addition, the concentration of these nucleotides in ascitic exudates was more remarkably reduced in animals treated with both FK866 and APCP compared with single treatments. Importantly, tumors treated with FK866 in combination with APCP contained a statistically significant lower proportion of Ki67 positive proliferating cells and a higher percentage of necrotic area. Finally, a slight but significant increase in animal survival in response to the combined therapy, compared to the single agents, could be demonstrated. Our results indicate that the pharmacological inhibition of CD73 enzymatic activity could be considered as a means to potentiate the anti-cancer effects of NAMPT inhibitors.
Collapse
Affiliation(s)
- Giovanna Sociali
- Department of Experimental Medicine, Section of Biochemistry, and CEBR, University of Genova, 16132 Genova, Italy
| | | | - Mirko Magnone
- Department of Experimental Medicine, Section of Biochemistry, and CEBR, University of Genova, 16132 Genova, Italy
| | - Federica Zamporlini
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Laura Emionite
- Animal Facility, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, 16132 Genova, Italy
| | - Laura Sturla
- Department of Experimental Medicine, Section of Biochemistry, and CEBR, University of Genova, 16132 Genova, Italy
| | - Giovanna Bianchi
- Laboratorio di Oncologia Istituto G. Gaslini, 16147 Genova, Italy
| | - Tiziana Vigliarolo
- Department of Experimental Medicine, Section of Biochemistry, and CEBR, University of Genova, 16132 Genova, Italy
| | - Aimable Nahimana
- Service and Central Laboratory of Hematology, University Hospital of Lausanne, 1011-CHUV, Lausanne, Switzerland
| | - Alessio Nencioni
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy.,IRCCS A.O.U. San Martino IST, Istituto Nazionale per la Ricerca sul Cancro, 16132 Genova, Italy
| | - Nadia Raffaelli
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine, Section of Biochemistry, and CEBR, University of Genova, 16132 Genova, Italy
| |
Collapse
|
24
|
Kennedy BE, Sharif T, Martell E, Dai C, Kim Y, Lee PWK, Gujar SA. NAD + salvage pathway in cancer metabolism and therapy. Pharmacol Res 2016; 114:274-283. [PMID: 27816507 DOI: 10.1016/j.phrs.2016.10.027] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 10/30/2016] [Indexed: 12/22/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential coenzyme for various physiological processes including energy metabolism, DNA repair, cell growth, and cell death. Many of these pathways are typically dysregulated in cancer cells, making NAD+ an intriguing target for cancer therapeutics. NAD+ is mainly synthesized by the NAD+ salvage pathway in cancer cells, and not surprisingly, the pharmacological targeting of the NAD+ salvage pathway causes cancer cell cytotoxicity in vitro and in vivo. Several studies have described the precise consequences of NAD+ depletion on cancer biology, and have demonstrated that NAD+ depletion results in depletion of energy levels through lowered rates of glycolysis, reduced citric acid cycle activity, and decreased oxidative phosphorylation. Additionally, depletion of NAD+ causes sensitization of cancer cells to oxidative damage by disruption of the anti-oxidant defense system, decreased cell proliferation, and initiation of cell death through manipulation of cell signaling pathways (e.g., SIRT1 and p53). Recently, studies have explored the effect of well-known cancer therapeutics in combination with pharmacological depletion of NAD+ levels, and found in many cases a synergistic effect on cancer cell cytotoxicity. In this context, we will discuss the effects of NAD+ salvage pathway inhibition on cancer cell biology and provide insight on this pathway as a novel anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Barry E Kennedy
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Tanveer Sharif
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Emma Martell
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Cathleen Dai
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Youra Kim
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Patrick W K Lee
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada; Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Shashi A Gujar
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada; Department of Pathology, Dalhousie University, Halifax, NS, Canada; Centre for Innovative and Collaborative Health Systems Research, IWK Health Centre, Halifax, NS, Canada.
| |
Collapse
|
25
|
Cea M, Soncini D, Gobbi M, Lemoli RM, Cagnetta A. Exploiting tumor vulnerabilities: NAD(+)-depleting agents combined with anti-tumor drugs as innovative strategy to treat hematological malignancies. Expert Rev Anticancer Ther 2016; 16:897-8. [PMID: 27433930 DOI: 10.1080/14737140.2016.1212664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Michele Cea
- a Chair of Hematology, Department of Medicine (DiMI) , University of Genoa, AOU, I.R.C.C.S. San Martino -IST , Genova , Italy
| | - Debora Soncini
- a Chair of Hematology, Department of Medicine (DiMI) , University of Genoa, AOU, I.R.C.C.S. San Martino -IST , Genova , Italy
| | - Marco Gobbi
- a Chair of Hematology, Department of Medicine (DiMI) , University of Genoa, AOU, I.R.C.C.S. San Martino -IST , Genova , Italy
| | - Roberto M Lemoli
- a Chair of Hematology, Department of Medicine (DiMI) , University of Genoa, AOU, I.R.C.C.S. San Martino -IST , Genova , Italy
| | - Antonia Cagnetta
- a Chair of Hematology, Department of Medicine (DiMI) , University of Genoa, AOU, I.R.C.C.S. San Martino -IST , Genova , Italy
| |
Collapse
|
26
|
Cea M, Cagnetta A, Acharya C, Acharya P, Tai YT, Yang C, Lovera D, Soncini D, Miglino M, Fraternali-Orcioni G, Mastracci L, Nencioni A, Montecucco F, Monacelli F, Ballestrero A, Hideshima T, Chauhan D, Gobbi M, Lemoli RM, Munshi N, Treon SP, Anderson KC. Dual NAMPT and BTK Targeting Leads to Synergistic Killing of Waldenström Macroglobulinemia Cells Regardless of MYD88 and CXCR4 Somatic Mutation Status. Clin Cancer Res 2016; 22:6099-6109. [PMID: 27287071 DOI: 10.1158/1078-0432.ccr-16-0630] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 05/12/2016] [Accepted: 05/29/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Nicotinamide phosphoribosyltransferase (Nampt) regulates intracellular NAD+ pool and is highly expressed in a number of malignancies. FK866, a selective inhibitor of Nampt, depletes intracellular NAD+ levels, thereby blocking cellular metabolism and triggering sensitization to other drugs and cell death. Here we characterized the antitumor effects of Nampt inhibition in Waldenström macroglobulinemia. EXPERIMENTAL DESIGN We investigated Nampt role in MW cells using both mRNA and protein expression analyses. We have also used loss-of-function approaches to investigate the growth and survival effects of Nampt on MW cells and further tested the anti-MW activity of dual Nampt and BTK inhibition in vitro and in vivo RESULTS: We found that Waldenström macroglobulinemia cells exhibit high levels of Nampt compared with normal B cells. Loss of function studies suggested a potential oncogenic role of Nampt in Waldenström macroglobulinemia cells, and BTK-inhibitor ibrutinib and FK866 resulted in a significant and synergistic anti-Waldenström macroglobulinemia cell death, regardless of MYD88 and CXCR4 mutational status. Cell death was associated with: (i) activation of caspase-3, PARP and downregulation of Mcl-1, (ii) enhanced intracellular ATP and NAD+ depletion, (iii) inhibition of NF-κB signaling, and (iv) inhibition of multiple prosurvival signaling pathways. In a murine xenograft Waldenström macroglobulinemia model, low-dose combination FK866 and ibrutinib is well tolerated, significantly inhibits tumor growth, and prolongs host survival. CONCLUSIONS Our results show intracellular NAD+ level as crucial for proliferation and survival of Waldenström macroglobulinemia cells, and provides the mechanistic preclinical rationale for targeting Nampt, either alone or with Ibrutinib, to overcome drug resistance and improve patient outcome in Waldenström macroglobulinemia. Clin Cancer Res; 22(24); 6099-109. ©2016 AACR.
Collapse
Affiliation(s)
- Michele Cea
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. .,Department of Medicine (DiMI), University of Genoa, AOU, I.R.C.C.S. San Martino-IST, Genova, Italy
| | - Antonia Cagnetta
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Department of Medicine (DiMI), University of Genoa, AOU, I.R.C.C.S. San Martino-IST, Genova, Italy
| | - Chirag Acharya
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Prakrati Acharya
- Mount Auburn Hospital, Harvard Medical School, Cambridge, Massachusetts
| | - Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Cao Yang
- Bing Center for Waldenstrom's Macroglobulinemia, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Davide Lovera
- Department of Medicine (DiMI), University of Genoa, AOU, I.R.C.C.S. San Martino-IST, Genova, Italy
| | - Debora Soncini
- Department of Medicine (DiMI), University of Genoa, AOU, I.R.C.C.S. San Martino-IST, Genova, Italy
| | - Maurizio Miglino
- Department of Medicine (DiMI), University of Genoa, AOU, I.R.C.C.S. San Martino-IST, Genova, Italy
| | | | - Luca Mastracci
- Department of Surgical and Diagnostic Sciences (DISC), Pathology Unit, University of Genoa and IRCCS AUO S. Martino-IST, Genova, Italy
| | - Alessio Nencioni
- Department of Medicine (DiMI), University of Genoa, AOU, I.R.C.C.S. San Martino-IST, Genova, Italy
| | - Fabrizio Montecucco
- Department of Medicine (DiMI), University of Genoa, AOU, I.R.C.C.S. San Martino-IST, Genova, Italy
| | - Fiammetta Monacelli
- Department of Medicine (DiMI), University of Genoa, AOU, I.R.C.C.S. San Martino-IST, Genova, Italy
| | - Alberto Ballestrero
- Department of Medicine (DiMI), University of Genoa, AOU, I.R.C.C.S. San Martino-IST, Genova, Italy
| | - Teru Hideshima
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Dharminder Chauhan
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Marco Gobbi
- Department of Medicine (DiMI), University of Genoa, AOU, I.R.C.C.S. San Martino-IST, Genova, Italy
| | - Roberto M Lemoli
- Department of Medicine (DiMI), University of Genoa, AOU, I.R.C.C.S. San Martino-IST, Genova, Italy
| | - Nikhil Munshi
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Steven P Treon
- Bing Center for Waldenstrom's Macroglobulinemia, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
27
|
Mesquita I, Varela P, Belinha A, Gaifem J, Laforge M, Vergnes B, Estaquier J, Silvestre R. Exploring NAD+ metabolism in host-pathogen interactions. Cell Mol Life Sci 2016; 73:1225-36. [PMID: 26718485 PMCID: PMC11108276 DOI: 10.1007/s00018-015-2119-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 11/27/2015] [Accepted: 12/14/2015] [Indexed: 01/01/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD(+)) is a vital molecule found in all living cells. NAD(+) intracellular levels are dictated by its synthesis, using the de novo and/or salvage pathway, and through its catabolic use as co-enzyme or co-substrate. The regulation of NAD(+) metabolism has proven to be an adequate drug target for several diseases, including cancer, neurodegenerative or inflammatory diseases. Increasing interest has been given to NAD(+) metabolism during innate and adaptive immune responses suggesting that its modulation could also be relevant during host-pathogen interactions. While the maintenance of NAD(+) homeostatic levels assures an adequate environment for host cell survival and proliferation, fluctuations in NAD(+) or biosynthetic precursors bioavailability have been described during host-pathogen interactions, which will interfere with pathogen persistence or clearance. Here, we review the double-edged sword of NAD(+) metabolism during host-pathogen interactions emphasizing its potential for treatment of infectious diseases.
Collapse
Affiliation(s)
- Inês Mesquita
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Varela
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Belinha
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Gaifem
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | - Baptiste Vergnes
- MIVEGEC (IRD 224-CNRS 5290-Université Montpellier), Institut de Recherche pour le Développement (IRD), Montpellier, France
| | - Jérôme Estaquier
- CNRS FR 3636, Université Paris Descartes, 75006, Paris, France.
- Centre de Recherche du CHU de Québec, Université Laval, Quebec, G1V 4G2, Canada.
| | - Ricardo Silvestre
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
28
|
Zare-Abdollahi D, Safari S, Movafagh A, Ghadiani M, Tabarraee M, Riazi-Isfahani S, Gorji S, Keyvan L, Gachkar L. Expression analysis of BECN1 in acute myeloid leukemia: association with distinct cytogenetic and molecular abnormalities. Int J Lab Hematol 2016; 38:125-32. [PMID: 26765290 DOI: 10.1111/ijlh.12454] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 11/03/2015] [Indexed: 11/26/2022]
Abstract
INTRODUCTION In acute myeloid leukemia (AML), it has been shown that AML-derived cells often remain sensitive to autophagy-inducing stimuli, leading to the idea that harnessing the autophagy can be pertinent to AML cytotoxic therapy. Despite this promising notion, to date, there is no comprehensive study addressing autophagy-related genes expression status in AML. As a critical mediator, BECN1 influences the onset and advance of autophagy and several studies have pointed to the BECN1 recurrent allelic deletion and expression variation in a broad range of tumors. To explore this caveat, we chose this alteration-prone gene to investigate in our study. METHODS We have analyzed the expression status of BECN1 in a series of 128 de novo AML patients using real-time quantitative polymerase chain reaction (qRT-PCR). RESULTS In our favorable subgroup, BECN1 expression did not alter (P = 0.301), but in intermediate and unfavorable patients, we have had BECN1 low expression compared to the normal controls (P = 0.008 and P < 0.001, respectively). We found evidence for the association of reduced expression of BECN1 with FLT3-ITD mutation (19 of 27 patients), monosomal karyotype (all of 11 patients), higher age, and WBC count. CONCLUSION Overall, remarkable association of reduced expression of BECN1 with FLT3-ITD mutation and monosomal karyotype and their functional relationship is interesting which should be addressed and verified in future studies.
Collapse
Affiliation(s)
- D Zare-Abdollahi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - S Safari
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - A Movafagh
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - M Ghadiani
- Department of Hemato-Oncology, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - M Tabarraee
- Department of Hemato-Oncology, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - S Riazi-Isfahani
- Department of Social Determinants of Health, National Institute of Health Research, Tehran University of Medical Sciences, Tehran, Iran
| | - S Gorji
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - L Keyvan
- Department of Genetics, South PNU Centre, Payame Noor University, Tehran, Iran
| | - L Gachkar
- Department of Infectious Diseases and Tropical Medicine, Cardiovascular Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Lerrer B, Gertler AA, Cohen HY. The complex role of SIRT6 in carcinogenesis. Carcinogenesis 2015; 37:108-18. [PMID: 26717993 DOI: 10.1093/carcin/bgv167] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 11/25/2015] [Indexed: 12/28/2022] Open
Abstract
SIRT6, a member of the mammalian sirtuins family, functions as a mono-ADP-ribosyl transferase and NAD(+)-dependent deacylase of both acetyl groups and long-chain fatty acyl groups. SIRT6 regulates diverse cellular functions such as transcription, genome stability, telomere integrity, DNA repair, inflammation and metabolic related diseases such as diabetes, obesity and cancer. In this review, we will discuss the implication of SIRT6 in the biology of cancer and the relevance to organism homeostasis and lifespan.
Collapse
Affiliation(s)
- Batia Lerrer
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Asaf A Gertler
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Haim Y Cohen
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| |
Collapse
|
30
|
EIF2A-dependent translational arrest protects leukemia cells from the energetic stress induced by NAMPT inhibition. BMC Cancer 2015; 15:855. [PMID: 26542945 PMCID: PMC4636066 DOI: 10.1186/s12885-015-1845-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 10/23/2015] [Indexed: 01/04/2023] Open
Abstract
Background Nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in NAD+ biosynthesis from nicotinamide, is one of the major factors regulating cancer cells metabolism and is considered a promising target for treating cancer. The prototypical NAMPT inhibitor FK866 effectively lowers NAD+ levels in cancer cells, reducing the activity of NAD+-dependent enzymes, lowering intracellular ATP, and promoting cell death. Results We show that FK866 induces a translational arrest in leukemia cells through inhibition of MTOR/4EBP1 signaling and of the initiation factors EIF4E and EIF2A. Specifically, treatment with FK866 is shown to induce 5′AMP-activated protein kinase (AMPK) activation, which, together with EIF2A phosphorylation, is responsible for the inhibition of protein synthesis. Notably, such an effect was also observed in patients’ derived primary leukemia cells including T-cell Acute Lymphoblastic Leukemia. Jurkat cells in which AMPK or LKB1 expression was silenced or in which a non-phosphorylatable EIF2A mutant was ectopically expressed showed enhanced sensitivity to the NAMPT inhibitor, confirming a key role for the LKB1-AMPK-EIF2A axis in cell fate determination in response to energetic stress via NAD+ depletion. Conclusions We identified EIF2A phosphorylation as a novel early molecular event occurring in response to NAMPT inhibition and mediating protein synthesis arrest. In addition, our data suggest that tumors exhibiting an impaired LBK1- AMPK- EIF2A response may be especially susceptible to NAMPT inhibitors and thus become an elective indication for this type of agents. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1845-1) contains supplementary material, which is available to authorized users.
Collapse
|