1
|
Hong SM, Lee A, Kim B, Lee J, Seon S, Ha Y, Ng JT, Yoon G, Lim SB, Morgan MJ, Cha J, Lee D, Kim Y. NAMPT-Driven M2 Polarization of Tumor-Associated Macrophages Leads to an Immunosuppressive Microenvironment in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303177. [PMID: 38308188 PMCID: PMC11005718 DOI: 10.1002/advs.202303177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 12/16/2023] [Indexed: 02/04/2024]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is a metabolic enzyme with key roles in inflammation. Previous studies have examined the consequences of its upregulated expression in cancer cells themselves, but studies are limited with respect to its role in the other cells within the tumor microenvironment (TME) during colorectal cancer (CRC) progression. Using single-cell RNA sequencing (scRNA-seq) data, it is founded that NAMPT is highly expressed in SPP1+ tumor-associated macrophages (TAMs), a unique subset of TAMs associated with immunosuppressive activity. A NAMPThigh gene signature in SPP1+ TAMs correlated with worse prognostic outcomes in CRC patients. The effect of Nampt deletion in the myeloid compartment of mice during CRC development is explored. NAMPT deficiency in macrophages resulted in HIF-1α destabilization, leading to reduction in M2-like TAM polarization. NAMPT deficiency caused significant decreases in the efferocytosis activity of macrophages, which enhanced STING signaling and the induction of type I IFN-response genes. Expression of these genes contributed to anti-tumoral immunity via potentiation of cytotoxic T cell activity in the TME. Overall, these findings suggest that NAMPT-initiated TAM-specific genes can be useful in predicting poor CRC patient outcomes; strategies aimed at targeting NAMPT may provide a promising therapeutic approach for building an immunostimulatory TME in CRC progression.
Collapse
Affiliation(s)
- Sun Mi Hong
- Department of BiochemistryAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
| | - A‐Yeon Lee
- Department of BiochemistryAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
| | - Byeong‐Ju Kim
- Department of BiochemistryAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
| | - Jeong‐Eun Lee
- Department of BiochemistryAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
| | - Su‐Yeon Seon
- Department of BiochemistryAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
| | - Yu‐Jin Ha
- Department of BiochemistryAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
| | - Jestlin Tianthing Ng
- Department of BiochemistryAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
| | - Gyesoon Yoon
- Department of BiochemistryAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
| | - Su Bin Lim
- Department of BiochemistryAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
| | - Michael J. Morgan
- Department of Natural SciencesNortheastern State UniversityTahlequahOK74464USA
| | - Jong‐Ho Cha
- Department of Biomedical SciencesCollege of MedicineInha UniversityIncheon22212South Korea
- Department of Biomedical Science and EngineeringGraduate SchoolInha UniversityIncheon22212South Korea
| | - Dakeun Lee
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
- Department of PathologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
| | - You‐Sun Kim
- Department of BiochemistryAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐guSuwonGyeonggi‐do16499Republic of Korea
| |
Collapse
|
2
|
Zhou C, Liu Y, Li Y, Shi L. Recent advances and prospects in nanomaterials for bacterial sepsis management. J Mater Chem B 2023; 11:10778-10792. [PMID: 37901894 DOI: 10.1039/d3tb02220j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
Bacterial sepsis is a life-threatening condition caused by bacteria entering the bloodstream and triggering an immune response, underscoring the importance of early recognition and prompt treatment. Nanomedicine holds promise for addressing sepsis through improved diagnostics, nanoparticle biosensors for detection and imaging, enhanced antibiotic delivery, combating resistance, and immune modulation. However, challenges remain in ensuring safety, regulatory compliance, scalability, and cost-effectiveness before clinical implementation. Further research is needed to optimize design, efficacy, safety, and regulatory strategies for effective utilization of nanomedicines in bacterial sepsis diagnosis and treatment. This review highlights the significant potential of nanomedicines, including improved drug delivery, enhanced diagnostics, and immunomodulation for bacterial sepsis. It also emphasizes the need for further research to optimize design, efficacy, safety profiles, and address regulatory challenges to facilitate clinical translation.
Collapse
Affiliation(s)
- Chaoyang Zhou
- Department of Critical Care Medicine, The People's Hospital of Yuhuan, Taizhou, Zhejiang 317600, China.
| | - Yong Liu
- Department of Critical Care Medicine, The People's Hospital of Yuhuan, Taizhou, Zhejiang 317600, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China.
| | - Yuanfeng Li
- Department of Critical Care Medicine, The People's Hospital of Yuhuan, Taizhou, Zhejiang 317600, China.
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
3
|
Perez-Sanchez C, Escudero-Contreras A, Cerdó T, Sánchez-Mendoza LM, Llamas-Urbano A, la Rosa IAD, Pérez-Rodriguez M, Muñoz-Barrera L, Del Carmen Abalos-Aguilera M, Barbarroja N, Calvo J, Ortega-Castro R, Ruiz-Vilchez D, Moreno JA, Burón MI, González-Reyes JA, Collantes-Estevez E, Lopez-Pedrera C, Villalba JM. Preclinical Characterization of Pharmacologic NAD + Boosting as a Promising Therapeutic Approach in Rheumatoid Arthritis. Arthritis Rheumatol 2023; 75:1749-1761. [PMID: 37094367 DOI: 10.1002/art.42528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/08/2023] [Accepted: 04/04/2023] [Indexed: 04/26/2023]
Abstract
OBJECTIVE We analyzed NAD+ metabolism in patients with rheumatoid arthritis (RA), its association with disease activity and clinical outcomes of RA, and the therapeutic potential of pharmacologic NAD+ boosting. METHODS Our study included 253 participants. In the first cohort, comprising 153 RA patients and 56 healthy donors, we assessed NAD+ levels and NAD+ -related gene pathways. We analyzed 92 inflammatory molecules by proximity extension assay. In the second cohort, comprising 44 RA patients starting anti-tumor necrosis factor (anti-TNF) drugs, we evaluated changes in NAD+ levels and their association with clinical response after 3 months. Mechanistic studies were performed ex vivo on peripheral blood mononuclear cells (PBMCs) from patients with RA to test the beneficial effects of NAD+ boosters, such as nicotinamide and nicotinamide riboside. RESULTS Reduced NAD+ levels were found in RA samples, in line with altered activity and expression of genes involved in NAD+ consumption (sirtuins, poly[ADP-ribose] polymerase, CD38), transport (connexin 43), and biosynthesis (NAMPT, NMNATs). Unsupervised clustering analysis identified a group of RA patients with the highest inflammatory profile, the lowest NAD+ levels, and the highest disease activity (as shown by the Disease Activity Score in 28 joints). NAD+ levels were modulated by anti-TNF therapy in parallel with the clinical response. In vitro studies using PBMCs from RA patients showed that nicotinamide riboside and nicotinamide increased NAD+ levels via NAMPT and NMNAT and reduced their prooxidative, proapoptotic, and proinflammatory status. CONCLUSION RA patients display altered NAD+ metabolism, directly linked to their inflammatory and disease activity status, which was reverted by anti-TNF therapy. The preclinical beneficial effects of NAD+ boosters, as shown in leukocytes from RA patients, along with their proven clinical safety, might pave the way for the development of clinical trials using these compounds.
Collapse
Affiliation(s)
- Carlos Perez-Sanchez
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, and Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, Spain; Cobiomic Bioscience
| | | | - Tomás Cerdó
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Luz Marina Sánchez-Mendoza
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, Córdoba, Spain
| | - Adrián Llamas-Urbano
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Iván Arias-de la Rosa
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Miguel Pérez-Rodriguez
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, Córdoba, Spain
| | - Laura Muñoz-Barrera
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | | | - Nuria Barbarroja
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Jerusalem Calvo
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Rafaela Ortega-Castro
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Desiree Ruiz-Vilchez
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Juan Antonio Moreno
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, and Laboratory GE-06, IMIBIC, Nephrology Service, Reina Sofia University Hospital, ceiA3, Córdoba, Spain
| | - María Isabel Burón
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, Córdoba, Spain
| | - José Antonio González-Reyes
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, Córdoba, Spain
| | - Eduardo Collantes-Estevez
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Chary Lopez-Pedrera
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - José Manuel Villalba
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, Córdoba, Spain
| |
Collapse
|
4
|
Fang J, Hou P, Liu S, Zuo M, Liu Z, Chen W, Han Y, Li Y, Wang T, Feng C, Li P, Shao C, Shi Y. NAD + salvage governs the immunosuppressive capacity of mesenchymal stem cells. Cell Mol Immunol 2023; 20:1171-1185. [PMID: 37580400 PMCID: PMC10541442 DOI: 10.1038/s41423-023-01073-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/24/2023] [Indexed: 08/16/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) possess robust immunoregulatory functions and are promising therapeutics for inflammatory disorders. This capacity is not innate but is activated or 'licensed' by inflammatory cytokines. The licensing mechanism remains unclear. Here, we examined whether inflammatory cytokines metabolically reprogrammed MSCs to confer this immunoregulatory capacity. In response to stimulation by inflammatory cytokines, MSCs exhibited a dramatic increase in the consumption of glucose, which was accompanied by an enhanced use of nicotinamide adenine dinucleotide (NAD+) and increased expression of nicotinamide phosphoribosyltransferase (NAMPT), a central enzyme in the salvage pathway for NAD+ production. When NAD+ synthesis was blocked by inhibiting or depleting NAMPT, the immunosuppressive function of MSCs induced by inflammatory cytokines was greatly attenuated. Consequently, when NAD+ metabolism in MSCs was perturbed, their therapeutic benefit was decreased in mice suffering from inflammatory bowel disease and acute liver injury. Further analysis revealed that NAMPT-driven production of NAD+ was critical for the inflammatory cytokine-induced increase in glycolysis in MSCs. Furthermore, the increase in glycolysis led to succinate accumulation in the tricarboxylic acid cycle, which led to hypoxia-inducible factor 1α (HIF-1α) stabilization and subsequently increased the transcription of key glycolytic genes, thereby persistently maintaining glycolytic flux. This study demonstrated that unlike its proinflammatory role in immune cells, NAD+ metabolism governs the anti-inflammatory function of MSCs during inflammation.
Collapse
Affiliation(s)
- Jiankai Fang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Pengbo Hou
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Shisong Liu
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Muqiu Zuo
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Zhanhong Liu
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Wangwang Chen
- Laboratory Animal Center, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yuyi Han
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Yanan Li
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Tingting Wang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chao Feng
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Peishan Li
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Changshun Shao
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China.
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
5
|
Li Q, Lu J, Li J, Zhang B, Wu Y, Ying T. Antibody-based cancer immunotherapy by targeting regulatory T cells. Front Oncol 2023; 13:1157345. [PMID: 37182149 PMCID: PMC10174253 DOI: 10.3389/fonc.2023.1157345] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
Regulatory T cells (Tregs) are among the most abundant suppressive cells, which infiltrate and accumulate in the tumor microenvironment, leading to tumor escape by inducing anergy and immunosuppression. Their presence has been correlated with tumor progression, invasiveness and metastasis. Targeting tumor-associated Tregs is an effective addition to current immunotherapy approaches, but it may also trigger autoimmune diseases. The major limitation of current therapies targeting Tregs in the tumor microenvironment is the lack of selective targets. Tumor-infiltrating Tregs express high levels of cell surface molecules associated with T-cell activation, such as CTLA4, PD-1, LAG3, TIGIT, ICOS, and TNF receptor superfamily members including 4-1BB, OX40, and GITR. Targeting these molecules often attribute to concurrent depletion of antitumor effector T-cell populations. Therefore, novel approaches need to improve the specificity of targeting Tregs in the tumor microenvironment without affecting peripheral Tregs and effector T cells. In this review, we discuss the immunosuppressive mechanisms of tumor-infiltrating Tregs and the status of antibody-based immunotherapies targeting Tregs.
Collapse
Affiliation(s)
- Quanxiao Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun Lu
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| | - Baohong Zhang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yanling Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Tianlei Ying, ; Yanling Wu,
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Tianlei Ying, ; Yanling Wu,
| |
Collapse
|
6
|
Ye M, Zhao Y, Wang Y, Xie R, Tong Y, Sauer JD, Gong S. NAD(H)-loaded nanoparticles for efficient sepsis therapy via modulating immune and vascular homeostasis. NATURE NANOTECHNOLOGY 2022; 17:880-890. [PMID: 35668170 PMCID: PMC10044491 DOI: 10.1038/s41565-022-01137-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 04/08/2022] [Indexed: 05/14/2023]
Abstract
Sepsis is a life-threatening organ dysfunction responsible for nearly 270,000 deaths annually in the United States alone. Nicotinamide adenine dinucleotide (NAD+), an immunomodulator, can potentially treat sepsis; however, clinical application of NAD+ is hindered by its inability to be directly taken up by cells. To address this challenge, a family of nanoparticles (NPs) loaded with either NAD+ or the reduced form of NAD+ (NADH), hereafter NAD(H)-loaded NPs, were engineered to enable direct cellular transport and replenishment of NAD(H). The NAD(H)-loaded NPs improved cellular energy supply, suppressed inflammation and prevented inflammation-induced cell pyroptosis and apoptosis. Therefore, the NPs can help maintain immune homoeostasis and vascular function, two key factors in the pathogenesis of sepsis. The NAD(H)-loaded NPs demonstrated excellent therapeutic efficacies in treating endotoxemia and multidrug-resistant pathogen-induced bacteremia. In addition, the NAD(H)-loaded NPs prevented caecal ligation and puncture-induced multiorgan injury and improved outcomes of secondary Pseudomonas aeruginosa infections following caecal ligation and puncture, thus potentially leading to a highly innovative and translational approach to treat sepsis efficiently and safely.
Collapse
Affiliation(s)
- Mingzhou Ye
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Yi Zhao
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Yuyuan Wang
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Ruosen Xie
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Yao Tong
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Shaoqin Gong
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
7
|
D'Amico F, Costantino G, Salvatorelli L, Ramondetta A, De Pasquale R, Sortino MA, Merlo S. Inverse correlation between the expression of AMPK/SIRT1 and NAMPT in psoriatic skin: A pilot study. Adv Med Sci 2022; 67:262-268. [PMID: 35839539 DOI: 10.1016/j.advms.2022.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/23/2022] [Accepted: 07/04/2022] [Indexed: 11/01/2022]
Abstract
PURPOSE Epidermal hyperplasia and the involvement of immune cells characterize the clinical picture of psoriasis. Among the several factors involved, attention has been focused on sirtuin 1 (SIRT1) - a deacetylase endowed with a variety of functions including the control of metabolic and inflammatory processes-, and on nicotinamide phosphoribosyltransferase (NAMPT), important for SIRT1 activation and involved in inflammatory events. The aim of the study was to analyze changes of SIRT1 and NAMPT expression in psoriatic skin. PATIENTS AND METHODS Samples from healthy controls and psoriatic patients were subjected to immunohistochemical analysis. RESULTS A strong downregulation of SIRT1 expression was observed in skin samples from psoriatic patients compared to healthy controls. This was accompanied by a parallel reduction of adenosine monophosphate-activated kinase (AMPK) expression and, more strikingly, by the disappearance of cells immunolabeled for its active, phosphorylated form (pAMPK). In both cases, analysis of the distribution of immunopositive cells revealed a shift towards reduced intensity of staining. In contrast, NAMPT expression was upregulated in psoriatic samples in line with its pro-inflammatory role. This was again more visible with an intensity-based distribution analysis that evidenced a shift towards more intensely immunostained cell populations. CONCLUSIONS The present data correlate in the same samples the expression of SIRT1, pAMPK/AMPK and NAMPT in psoriasis and open the way for novel pharmacological targets in the treatment of the disease.
Collapse
Affiliation(s)
- Fabio D'Amico
- Department of Biomedical and Biotechnological Sciences, Section of Pathology, University of Catania, Catania, Italy
| | - Giuseppe Costantino
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy; PhD Program in Neuroscience and Education, University of Foggia, Foggia, Italy
| | - Lucia Salvatorelli
- Department of Medical and Surgical Sciences and Advanced Technologies, Section of Anatomic Pathology, School of Medicine, University of Catania, Catania, Italy
| | | | - Rocco De Pasquale
- Department of General Surgery and Medico-Surgical Specialties, University of Catania, Catania, Italy
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.
| | - Sara Merlo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| |
Collapse
|
8
|
Drapela S, Ilter D, Gomes AP. Metabolic reprogramming: a bridge between aging and tumorigenesis. Mol Oncol 2022; 16:3295-3318. [PMID: 35666002 PMCID: PMC9490145 DOI: 10.1002/1878-0261.13261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/07/2022] [Accepted: 05/23/2022] [Indexed: 12/01/2022] Open
Abstract
Aging is the most robust risk factor for cancer development, with more than 60% of cancers occurring in those aged 60 and above. However, how aging and tumorigenesis are intertwined is poorly understood and a matter of significant debate. Metabolic changes are hallmarks of both aging and tumorigenesis. The deleterious consequences of aging include dysfunctional cellular processes, the build‐up of metabolic byproducts and waste molecules in circulation and within tissues, and stiffer connective tissues that impede blood flow and oxygenation. Collectively, these age‐driven changes lead to metabolic reprogramming in different cell types of a given tissue that significantly affects their cellular functions. Here, we put forward the idea that metabolic changes that happen during aging help create a favorable environment for tumorigenesis. We review parallels in metabolic changes that happen during aging and how these changes function both as adaptive mechanisms that enable the development of malignant phenotypes in a cell‐autonomous manner and as mechanisms that suppress immune surveillance, collectively creating the perfect environment for cancers to thrive. Hence, antiaging therapeutic strategies that target the metabolic reprogramming that occurs as we age might provide new opportunities to prevent cancer initiation and/or improve responses to standard‐of‐care anticancer therapies.
Collapse
Affiliation(s)
- Stanislav Drapela
- Department of Molecular Oncology, H. Lee Moffit Cancer Center & Research Institute, Tampa, FL, USA
| | - Didem Ilter
- Department of Molecular Oncology, H. Lee Moffit Cancer Center & Research Institute, Tampa, FL, USA
| | - Ana P Gomes
- Department of Molecular Oncology, H. Lee Moffit Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
9
|
Picerno A, Castellano G, Curci C, Kopaczka K, Stasi A, Pertosa GB, Sabbà C, Gesualdo L, Gramignoli R, Sallustio F. The Icarus Flight of Perinatal Stem and Renal Progenitor Cells Within Immune System. Front Immunol 2022; 13:840146. [PMID: 35355984 PMCID: PMC8959820 DOI: 10.3389/fimmu.2022.840146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/11/2022] [Indexed: 12/02/2022] Open
Abstract
Our immune system actively fights bacteria and viruses, and it must strike a delicate balance between over- and under-reaction, just like Daedalus and Icarus in Greek mythology, who could not escape their imprisonment by flying too high or too low. Both human amniotic epithelial and mesenchymal stromal cells and the conditioned medium generated from their culture exert multiple immunosuppressive activities. They have strong immunomodulatory properties that are influenced by the types and intensity of inflammatory stimuli present in the microenvironment. Notably, very recently, the immunomodulatory activity of human adult renal stem/progenitor cells (ARPCs) has been discovered. ARPCs cause a decrease in Tregs and CD3+ CD4- CD8- (DN) T cells in the early stages of inflammation, encouraging inflammation, and an increase in the late stages of inflammation, favoring inflammation quenching. If the inflammatory trigger continues, however, ARPCs cause a further increase in DN T cells to avoid the development of a harmful inflammatory state. As in the flight of Daedalus and Icarus, who could not fly too high or too low to not destroy their wings by the heat of the sun or the humidity of the sea, in response to an inflammatory environment, stem cells seem to behave by paying attention to regulating T cells in the balance between immune tolerance and autoimmunity. Recognizing the existence of both suppressive and stimulatory properties, and the mechanisms that underpin the duality of immune reaction, will aid in the development of active immunotherapeutic approaches that manipulate the immune system to achieve therapeutic benefit.
Collapse
Affiliation(s)
- Angela Picerno
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Bari, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Renal Transplant Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Claudia Curci
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Katarzyna Kopaczka
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Alessandra Stasi
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Battista Pertosa
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Carlo Sabbà
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Bari, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Fabio Sallustio
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|
10
|
Funk-Hilsdorf TC, Behrens F, Grune J, Simmons S. Dysregulated Immunity in Pulmonary Hypertension: From Companion to Composer. Front Physiol 2022; 13:819145. [PMID: 35250621 PMCID: PMC8891568 DOI: 10.3389/fphys.2022.819145] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/20/2022] [Indexed: 12/26/2022] Open
Abstract
Pulmonary hypertension (PH) represents a grave condition associated with high morbidity and mortality, emphasizing a desperate need for innovative and targeted therapeutic strategies. Cumulative evidence suggests that inflammation and dysregulated immunity interdependently affect maladaptive organ perfusion and congestion as hemodynamic hallmarks of the pathophysiology of PH. The role of altered cellular and humoral immunity in PH gains increasing attention, especially in pulmonary arterial hypertension (PAH), revealing novel mechanistic insights into the underlying immunopathology. Whether these immunophysiological aspects display a universal character and also hold true for other types of PH (e.g., PH associated with left heart disease, PH-LHD), or whether there are unique immunological signatures depending on the underlying cause of disease are points of consideration and discussion. Inflammatory mediators and cellular immune circuits connect the local inflammatory landscape in the lung and heart through inter-organ communication, involving, e.g., the complement system, sphingosine-1-phosphate (S1P), cytokines and subsets of, e.g., monocytes, macrophages, natural killer (NK) cells, dendritic cells (DCs), and T- and B-lymphocytes with distinct and organ-specific pro- and anti-inflammatory functions in homeostasis and disease. Perivascular macrophage expansion and monocyte recruitment have been proposed as key pathogenic drivers of vascular remodeling, the principal pathological mechanism in PAH, pinpointing toward future directions of anti-inflammatory therapeutic strategies. Moreover, different B- and T-effector cells as well as DCs may play an important role in the pathophysiology of PH as an imbalance of T-helper-17-cells (TH17) activated by monocyte-derived DCs, a potentially protective role of regulatory T-cells (Treg) and autoantibody-producing plasma cells occur in diverse PH animal models and human PH. This article highlights novel aspects of the innate and adaptive immunity and their interaction as disease mediators of PH and its specific subtypes, noticeable inflammatory mediators and summarizes therapeutic targets and strategies arising thereby.
Collapse
Affiliation(s)
- Teresa C. Funk-Hilsdorf
- Junior Research Group “Immunodynamics”, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Felix Behrens
- Junior Research Group “Immunodynamics”, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Jana Grune
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Szandor Simmons
- Junior Research Group “Immunodynamics”, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- *Correspondence: Szandor Simmons,
| |
Collapse
|
11
|
Yang Y, Yue S, Qiao Y, Zhang P, Jiang N, Ning Z, Liu C, Hou Y. Activable Multi-Modal Nanoprobes for Imaging Diagnosis and Therapy of Tumors. Front Chem 2021; 8:572471. [PMID: 33912535 PMCID: PMC8075363 DOI: 10.3389/fchem.2020.572471] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 12/18/2020] [Indexed: 01/05/2023] Open
Abstract
Malignant tumors have become one of the major causes of human death, but there remains a lack of effective methods for tiny tumor diagnosis, metastasis warning, clinical efficacy prediction, and effective treatment. In this context, localizing tiny tumors via imaging and non-invasively extracting molecular information related to tumor proliferation, invasion, metastasis, and drug resistance from the tumor microenvironment have become the most fundamental tasks faced by cancer researchers. Tumor-associated microenvironmental physiological parameters, such as hypoxia, acidic extracellular pH, protease, reducing conditions, and so forth, have much to do with prognostic indicators for cancer progression, and impact therapeutic administrations. By combining with various novel nanoparticle-based activatable probes, molecular imaging technologies can provide a feasible approach to visualize tumor-associated microenvironment parameters noninvasively and realize accurate treatment of tumors. This review focuses on the recent achievements in the design of “smart” nanomedicine responding to the tumor microenvironment-related features and highlights state-of- the-art technology in tumor imaging diagnosis and therapy.
Collapse
Affiliation(s)
- Yan Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Saisai Yue
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yuanyuan Qiao
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Peisen Zhang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Ni Jiang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zhenbo Ning
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Chunyan Liu
- Institute of Atmospheric Physics, Chinese Academy of Sciences, Beijing, China
| | - Yi Hou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.,Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Berberine-induced TFEB deacetylation by SIRT1 promotes autophagy in peritoneal macrophages. Aging (Albany NY) 2021; 13:7096-7119. [PMID: 33639613 PMCID: PMC7993719 DOI: 10.18632/aging.202566] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 01/13/2021] [Indexed: 12/14/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease that commonly affects the elderly and is characterized by vascular damage, macrophage infiltration, and plaque formation. Moreover, it increases the risk of cardiovascular disease. The pathogenesis of atherosclerosis involves an interplay between macrophage autophagy and apoptosis. A recently discovered transcription factor, transcription factor EB (TFEB) is known to activate autophagy in macrophages. Sirtuin deacetylase 1 (SIRT1), a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase, activates several transcription factors, including TFEB. We studied the effects of berberine on the NAD+ synthesis pathway and interactions between SIRT1 and TFEB. We also studied the effects of berberine-induced TFEB activation via SIRT1 on autophagy and apoptosis of peritoneal macrophages. We found that berberine promoted autophagy of peritoneal macrophages by activating SIRT1 via the NAD+ synthesis pathway and, in turn, promoting TFEB nuclear translocation and deacetylation. The functional regulation of SIRT1 and TFEB by berberine could be exploited as a potential therapeutic strategy for atherosclerosis.
Collapse
|
13
|
Navas LE, Carnero A. NAD + metabolism, stemness, the immune response, and cancer. Signal Transduct Target Ther 2021; 6:2. [PMID: 33384409 PMCID: PMC7775471 DOI: 10.1038/s41392-020-00354-w] [Citation(s) in RCA: 195] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/11/2020] [Accepted: 09/27/2020] [Indexed: 02/07/2023] Open
Abstract
NAD+ was discovered during yeast fermentation, and since its discovery, its important roles in redox metabolism, aging, and longevity, the immune system and DNA repair have been highlighted. A deregulation of the NAD+ levels has been associated with metabolic diseases and aging-related diseases, including neurodegeneration, defective immune responses, and cancer. NAD+ acts as a cofactor through its interplay with NADH, playing an essential role in many enzymatic reactions of energy metabolism, such as glycolysis, oxidative phosphorylation, fatty acid oxidation, and the TCA cycle. NAD+ also plays a role in deacetylation by sirtuins and ADP ribosylation during DNA damage/repair by PARP proteins. Finally, different NAD hydrolase proteins also consume NAD+ while converting it into ADP-ribose or its cyclic counterpart. Some of these proteins, such as CD38, seem to be extensively involved in the immune response. Since NAD cannot be taken directly from food, NAD metabolism is essential, and NAMPT is the key enzyme recovering NAD from nicotinamide and generating most of the NAD cellular pools. Because of the complex network of pathways in which NAD+ is essential, the important role of NAD+ and its key generating enzyme, NAMPT, in cancer is understandable. In the present work, we review the role of NAD+ and NAMPT in the ways that they may influence cancer metabolism, the immune system, stemness, aging, and cancer. Finally, we review some ongoing research on therapeutic approaches.
Collapse
Affiliation(s)
- Lola E Navas
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBER de Cancer, Sevilla, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain. .,CIBER de Cancer, Sevilla, Spain.
| |
Collapse
|
14
|
Hinrichs A, Riedel EO, Klymiuk N, Blutke A, Kemter E, Längin M, Dahlhoff M, Keßler B, Kurome M, Zakhartchenko V, Jemiller EM, Ayares D, Bidlingmaier M, Flenkenthaler F, Hrabĕ de Angelis M, Arnold GJ, Reichart B, Fröhlich T, Wolf E. Growth hormone receptor knockout to reduce the size of donor pigs for preclinical xenotransplantation studies. Xenotransplantation 2020; 28:e12664. [PMID: 33241624 DOI: 10.1111/xen.12664] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Many genetically multi-modified donor lines for xenotransplantation have a background of domestic pigs with rapid body and organ growth. The intrinsic growth potential of porcine xeno-organs may impair their long-term function after orthotopic transplantation in non-human primate models. Since growth hormone is a major stimulator of postnatal growth, we deleted its receptor (GHR-KO) to reduce the size of donor pigs in one step. METHODS Heart weight and proteome profile of myocardium were investigated in GHR-KO and control pigs. GHR-KO mutations were introduced using CRISPR/Cas9 in an α1,3-galactosyltransferase (GGTA1)-deficient background expressing the human cluster of differentiation (hCD46) and human thrombomodulin (hTHBD) to generate quadruple-modified (4GM) pigs. RESULTS At age 6 months, GHR-KO pigs had a 61% reduced body weight and a 63% reduced heart weight compared with controls. The mean minimal diameter of cardiomyocytes was 28% reduced. A holistic proteome study of myocardium samples from the two groups did not reveal prominent differences. Two 4GM founder sows had low serum insulin-like growth factor 1 (IGF1) levels (24 ± 1 ng/mL) and reached body weights of 70.3 and 73.4 kg at 9 months. Control pigs with IGF1 levels of 228 ± 24 ng/mL reached this weight range three months earlier. The 4GM sows showed normal sexual development and were mated with genetically multi-modified boars. Offspring revealed the expected Mendelian transmission of the genetic modifications and consistent expression of the transgenes. CONCLUSION GHR-KO donor pigs can be used at an age beyond the steepest phase of their growth curve, potentially reducing the problem of xeno-organ overgrowth in preclinical studies.
Collapse
Affiliation(s)
- Arne Hinrichs
- Department of Veterinary Sciences, Chair for Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Evamaria O Riedel
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Nikolai Klymiuk
- Department of Veterinary Sciences, Chair for Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Andreas Blutke
- Institute of Experimental Genetics, Helmholtz Zentrum München, Chair of Experimental Genetics, Technical University of Munich, Neuherberg, Germany
| | - Elisabeth Kemter
- Department of Veterinary Sciences, Chair for Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Matthias Längin
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Maik Dahlhoff
- Department of Veterinary Sciences, Chair for Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Barbara Keßler
- Department of Veterinary Sciences, Chair for Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Mayuko Kurome
- Department of Veterinary Sciences, Chair for Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Valeri Zakhartchenko
- Department of Veterinary Sciences, Chair for Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Eva-Maria Jemiller
- Department of Veterinary Sciences, Chair for Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | | | - Martin Bidlingmaier
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Florian Flenkenthaler
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Martin Hrabĕ de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München, Chair of Experimental Genetics, Technical University of Munich, Neuherberg, Germany
| | - Georg J Arnold
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Bruno Reichart
- Walter Brendel Center for Experimental Medicine, LMU Munich, Munich, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Eckhard Wolf
- Department of Veterinary Sciences, Chair for Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany.,Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| |
Collapse
|
15
|
Li M, Kirtane AR, Kiyokawa J, Nagashima H, Lopes A, Tirmizi ZA, Lee CK, Traverso G, Cahill DP, Wakimoto H. Local Targeting of NAD + Salvage Pathway Alters the Immune Tumor Microenvironment and Enhances Checkpoint Immunotherapy in Glioblastoma. Cancer Res 2020; 80:5024-5034. [PMID: 32998997 DOI: 10.1158/0008-5472.can-20-1094] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/17/2020] [Accepted: 09/25/2020] [Indexed: 12/21/2022]
Abstract
The aggressive primary brain tumor glioblastoma (GBM) is characterized by aberrant metabolism that fuels its malignant phenotype. Diverse genetic subtypes of malignant glioma are sensitive to selective inhibition of the NAD+ salvage pathway enzyme nicotinamide phosphoribosyltransferase (NAMPT). However, the potential impact of NAD+ depletion on the brain tumor microenvironment has not been elaborated. In addition, systemic toxicity of NAMPT inhibition remains a significant concern. Here we show that microparticle-mediated intratumoral delivery of NAMPT inhibitor GMX1778 induces specific immunologic changes in the tumor microenvironment of murine GBM, characterized by upregulation of immune checkpoint PD-L1, recruitment of CD3+, CD4+, and CD8+ T cells, and reduction of M2-polarized immunosuppressive macrophages. NAD+ depletion and autophagy induced by NAMPT inhibitors mediated the upregulation of PD-L1 transcripts and cell surface protein levels in GBM cells. NAMPT inhibitor modulation of the tumor immune microenvironment was therefore combined with PD-1 checkpoint blockade in vivo, significantly increasing the survival of GBM-bearing animals. Thus, the therapeutic impacts of NAMPT inhibition extended beyond neoplastic cells, shaping surrounding immune effectors. Microparticle delivery and release of NAMPT inhibitor at the tumor site offers a safe and robust means to alter an immune tumor microenvironment that could potentiate checkpoint immunotherapy for glioblastoma. SIGNIFICANCE: Microparticle-mediated local inhibition of NAMPT modulates the tumor immune microenvironment and acts cooperatively with anti-PD-1 checkpoint blockade, offering a combination immunotherapy strategy for the treatment of GBM.
Collapse
Affiliation(s)
- Ming Li
- Department of Neurosurgery, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Ameya R Kirtane
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Juri Kiyokawa
- Department of Neurosurgery, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Hiroaki Nagashima
- Department of Neurosurgery, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Aaron Lopes
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Zain A Tirmizi
- Department of Neurosurgery, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Christine K Lee
- Department of Neurosurgery, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel P Cahill
- Department of Neurosurgery, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
16
|
Radenkovic D, Reason, Verdin E. Clinical Evidence for Targeting NAD Therapeutically. Pharmaceuticals (Basel) 2020; 13:E247. [PMID: 32942582 PMCID: PMC7558103 DOI: 10.3390/ph13090247] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) pharmacology is a promising class of treatments for age-related conditions that are likely to have a favorable side effect profile for human use, given the widespread use of the NAD precursor vitamin B3 supplements. However, despite several decades of active investigation and numerous possible biochemical mechanisms of action suggested, only a small number of randomized and adequately powered clinical trials of NAD upregulation as a therapeutic strategy have taken place. We conducted a systematic review of the literature, following the PRISMA guidelines, in an attempt to determine whether or not the human clinical trials performed to date support the potential benefits of NAD supplementation in a range of skin, metabolic and age-related conditions. In addition, we sought medical indications that have yielded the most promising results in the limited studies to date. We conclude that promising, yet still speculative, results have been reported for the treatment of psoriasis and enhancement of skeletal muscle activity. However, further trials are required to determine the optimal method of raising NAD levels, identifying the target conditions, and comparisons to the present standard of care for these conditions. Lastly, pharmacological methods that increase NAD levels should also be directly compared to physiological means of raising NAD levels, such as exercise programs and dietary interventions that are tailored to older individuals, and which may be more effective.
Collapse
Affiliation(s)
- Dina Radenkovic
- Health Longevity Performance Optimisation Institute, Cambridge CB22 5NE, UK
| | - Reason
- Fight Aging!, 4736 Onondaga Blvd, PMB 179, Syracuse, NY 13219, USA;
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA;
| |
Collapse
|
17
|
Anderson G, Betancort Medina SR. Autism Spectrum Disorders: Role of Pre- and Post-Natal GammaDelta (γδ) T Cells and Immune Regulation. Curr Pharm Des 2020; 25:4321-4330. [PMID: 31682211 DOI: 10.2174/1381612825666191102170125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 10/31/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND It is widely accepted that alterations in immune functioning are an important aspect of the pathoetiology and pathophysiology of autism spectrum disorders (ASD). A relatively under-explored aspect of these alterations is the role of gammaDelta (γδ) T cells, prenatally and in the postnatal gut, which seem important hubs in driving the course of ASD. METHODS The present article describes the role of γδ T cells in ASD, including their interactions with other immune cells shown to be altered in this spectrum of conditions, including natural killer cells and mast cells. RESULTS Other risk factors in ASD, such as decreased vitamins A & D, as well as toxin-associated activation of the aryl hydrocarbon receptor, may also be intimately linked to γδ T cells, and alterations in the regulation of these cells. A growing body of data has highlighted an important role for alterations in mitochondria functioning in the regulation of immune cells, including natural killer cells and mast cells. This is an area that requires investigation in γδ T cells and their putative subtypes. CONCLUSION It is also proposed that maternal stress may act through alterations in the maternal microbiome, leading to changes in how the balance of short-chain fatty acids, such as butyrate, which may act to regulate the placenta and foetal development. Following an overview of previous research on immune, especially γδ T cells, effects in ASD, the future research implications are discussed in detail.
Collapse
Affiliation(s)
- George Anderson
- CRC Scotland & London, Eccleston Square, London, United Kingdom
| | | |
Collapse
|
18
|
Maes M, Anderson G, Betancort Medina SR, Seo M, Ojala JO. Integrating Autism Spectrum Disorder Pathophysiology: Mitochondria, Vitamin A, CD38, Oxytocin, Serotonin and Melatonergic Alterations in the Placenta and Gut. Curr Pharm Des 2020; 25:4405-4420. [PMID: 31682209 DOI: 10.2174/1381612825666191102165459] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 10/31/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND A diverse array of data has been associated with autism spectrum disorder (ASD), reflecting the complexity of its pathophysiology as well as its heterogeneity. Two important hubs have emerged, the placenta/prenatal period and the postnatal gut, with alterations in mitochondria functioning crucial in both. METHODS Factors acting to regulate mitochondria functioning in ASD across development are reviewed in this article. RESULTS Decreased vitamin A, and its retinoic acid metabolites, lead to a decrease in CD38 and associated changes that underpin a wide array of data on the biological underpinnings of ASD, including decreased oxytocin, with relevance both prenatally and in the gut. Decreased sirtuins, poly-ADP ribose polymerase-driven decreases in nicotinamide adenine dinucleotide (NAD+), hyperserotonemia, decreased monoamine oxidase, alterations in 14-3-3 proteins, microRNA alterations, dysregulated aryl hydrocarbon receptor activity, suboptimal mitochondria functioning, and decreases in the melatonergic pathways are intimately linked to this. Many of the above processes may be modulating, or mediated by, alterations in mitochondria functioning. Other bodies of data associated with ASD may also be incorporated within these basic processes, including how ASD risk factors such as maternal obesity and preeclampsia, as well as more general prenatal stressors, modulate the likelihood of offspring ASD. CONCLUSION Such a mitochondria-focussed integrated model of the pathophysiology of ASD has important preventative and treatment implications.
Collapse
Affiliation(s)
- Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - George Anderson
- CRC Scotland & London, Eccleston Square, London, United Kingdom
| | | | - Moonsang Seo
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Johanna O Ojala
- Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
19
|
Audrito V, Messana VG, Deaglio S. NAMPT and NAPRT: Two Metabolic Enzymes With Key Roles in Inflammation. Front Oncol 2020; 10:358. [PMID: 32266141 PMCID: PMC7096376 DOI: 10.3389/fonc.2020.00358] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) and nicotinate phosphoribosyltransferase (NAPRT) are two intracellular enzymes that catalyze the first step in the biosynthesis of NAD from nicotinamide and nicotinic acid, respectively. By fine tuning intracellular NAD levels, they are involved in the regulation/reprogramming of cellular metabolism and in the control of the activity of NAD-dependent enzymes, including sirtuins, PARPs, and NADases. However, during evolution they both acquired novel functions as extracellular endogenous mediators of inflammation. It is well-known that cellular stress and/or damage induce release in the extracellular milieu of endogenous molecules, called alarmins or damage-associated molecular patterns (DAMPs), which modulate immune functions through binding pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs), and activate inflammatory responses. Increasing evidence suggests that extracellular (e)NAMPT and eNAPRT are novel soluble factors with cytokine/adipokine/DAMP-like actions. Elevated eNAMPT were reported in several metabolic and inflammatory disorders, including obesity, diabetes, and cancer, while eNAPRT is emerging as a biomarker of sepsis and septic shock. This review will discuss available data concerning the dual role of this unique family of enzymes.
Collapse
Affiliation(s)
- Valentina Audrito
- Laboratory of Tumor Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Vincenzo Gianluca Messana
- Laboratory of Tumor Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Deaglio
- Laboratory of Tumor Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
20
|
Singhal A, Cheng CY. Host NAD+ metabolism and infections: therapeutic implications. Int Immunol 2020; 31:59-67. [PMID: 30329059 DOI: 10.1093/intimm/dxy068] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is both a crucial coenzyme and a cosubstrate for various metabolic reactions in all living cells. Maintenance of NAD+ levels is essential for cell energy homeostasis, survival, proliferation and function. Mounting evidence points to NAD+ as one of the major modulators of immuno-metabolic circuits, thus regulating immune responses and functions. Recent studies delineate impaired host NAD+ metabolism during chronic infections and inflammation, suggesting NAD+ replenishment as an avenue to ameliorate deleterious inflammatory responses. Here, we discuss aspects of NAD+ biosynthesis and consumption, NAD+ biology during infections and how NAD+ metabolism can be intervened with pharmacologically to enhance the host's immunological fitness against pathogens.
Collapse
Affiliation(s)
- Amit Singhal
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Vaccine and Infectious Disease Research Centre (VIDRC), Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Catherine Youting Cheng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
21
|
Chadha S, Wang L, Hancock WW, Beier UH. Sirtuin-1 in immunotherapy: A Janus-headed target. J Leukoc Biol 2019; 106:337-343. [PMID: 30605226 PMCID: PMC7477756 DOI: 10.1002/jlb.2ru1118-422r] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/13/2018] [Accepted: 12/16/2018] [Indexed: 12/16/2022] Open
Abstract
Sirtuin-1 (Sirt1), a member of the NAD-dependent sirtuin family of histone/protein deacetylases (HDAC), is an important target for immunotherapy due to its role in deacetylating the transcription factors Foxp3 and thymic retinoid acid receptor related orphan receptor gamma (RORγt). Sirt1 inhibition can increase Foxp3 acetylation and promote the production and functions of Foxp3+ T-regulatory (Treg) cells, whereas the acetylation of RORγt decreases its transcriptional activity DNA binding and decreases the differentiation of proinflammatory Th17 cells. Pharmacologic inhibitors of Sirt1 increase allograft survival and decrease autoimmune colitis and experimental allergic encephalomyelitis. However, in contrast to its role in T cells, Sirt1 has anti-inflammatory effects in myeloid cells, and, context dependent, in Th17 cells. Here, inhibition of Sirt1 can have proinflammatory effects. In addition to effects arising from the central role of Sirt1 in cellular metabolism and NAD-dependent reactions, such proinflammatory effects further complicate the potential of Sirt1 for therapeutic immunosuppression. This review aims to reconcile the opposing literature on pro- and anti-inflammatory effects of Sirt1, provides an overview of the role of Sir1 in the immune system, and discusses the pros and cons associated with inhibiting Sirt1 for control of inflammation and immune responses.
Collapse
Affiliation(s)
- Sakshum Chadha
- Division of Nephrology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, University of Pennsylvania, Philadelphia, PA 19104, USA
- Current address: Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Liqing Wang
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, and Biesecker Center for Pediatric Liver Disease, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wayne W. Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, and Biesecker Center for Pediatric Liver Disease, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ulf H. Beier
- Division of Nephrology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
22
|
Audrito V, Managò A, Gaudino F, Sorci L, Messana VG, Raffaelli N, Deaglio S. NAD-Biosynthetic and Consuming Enzymes as Central Players of Metabolic Regulation of Innate and Adaptive Immune Responses in Cancer. Front Immunol 2019; 10:1720. [PMID: 31402913 PMCID: PMC6671870 DOI: 10.3389/fimmu.2019.01720] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/09/2019] [Indexed: 12/15/2022] Open
Abstract
Cancer cells, particularly in solid tumors, are surrounded by non-neoplastic elements, including endothelial and stromal cells, as well as cells of immune origin, which can support tumor growth by providing the right conditions. On the other hand, local hypoxia, and lack of nutrients induce tumor cells to reprogram their metabolism in order to survive, proliferate, and disseminate: the same conditions are also responsible for building a tumor-suppressive microenvironment. In addition to tumor cells, it is now well-recognized that metabolic rewiring occurs in all cellular components of the tumor microenvironment, affecting epigenetic regulation of gene expression and influencing differentiation/proliferation decisions of these cells. Nicotinamide adenine dinucleotide (NAD) is an essential co-factor for energy transduction in metabolic processes. It is also a key component of signaling pathways, through the regulation of NAD-consuming enzymes, including sirtuins and PARPs, which can affect DNA plasticity and accessibility. In addition, both NAD-biosynthetic and NAD-consuming enzymes can be present in the extracellular environment, adding a new layer of complexity to the system. In this review we will discuss the role of the “NADome” in the metabolic cross-talk between cancer and infiltrating immune cells, contributing to cancer growth and immune evasion, with an eye to therapeutic implications.
Collapse
Affiliation(s)
- Valentina Audrito
- Department of Medical Sciences, University of Turin, Turin, Italy.,Italian Institute for Genomic Medicine, Turin, Italy
| | - Antonella Managò
- Department of Medical Sciences, University of Turin, Turin, Italy.,Italian Institute for Genomic Medicine, Turin, Italy
| | - Federica Gaudino
- Department of Medical Sciences, University of Turin, Turin, Italy.,Italian Institute for Genomic Medicine, Turin, Italy
| | - Leonardo Sorci
- Division of Bioinformatics and Biochemistry, Department of Materials, Environmental Sciences and Urban Planning, Polytechnic University of Marche, Ancona, Italy
| | - Vincenzo Gianluca Messana
- Department of Medical Sciences, University of Turin, Turin, Italy.,Italian Institute for Genomic Medicine, Turin, Italy
| | - Nadia Raffaelli
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin, Italy.,Italian Institute for Genomic Medicine, Turin, Italy
| |
Collapse
|
23
|
Petin K, Weiss R, Müller G, Garten A, Grahnert A, Sack U, Hauschildt S. NAD metabolites interfere with proliferation and functional properties of THP-1 cells. Innate Immun 2019; 25:280-293. [PMID: 31053044 PMCID: PMC6830904 DOI: 10.1177/1753425919844587] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Over the past few years the NAD-related compounds nicotinamide (NAM),
nicotinamide riboside (NR) and 1-methylnicotinamide (MNA) have been established
as important molecules in signalling pathways that contribute to metabolic
functions of many cells, including those of the immune system. Among immune
cells, monocytes/macrophages, which are the major players of inflammatory
processes, are especially susceptible to the anti-inflammatory action of NAM.
Here we asked whether NAM and the two other compounds have the potential to
regulate differentiation and LPS-induced biological answers of the monocytic
cell line THP-1. We show that treatment of THP-1 cells with NAM, NR and MNA
resulted in growth retardation accompanied by enrichment of cells in the
G0/G1-phase independent of p21 and p53. NAM and NR caused an increase in
intracellular NAD concentrations and SIRT1 and PARP1 mRNA expression was found
to be enhanced. The compounds failed to up-regulate the expression of the cell
surface differentiation markers CD38, CD11b and CD14. They modulated the
reactive oxygen species production and primed the cells to respond less
effectively to the LPS induced TNF-α production. Our data show that the NAD
metabolites interfere with early events associated with differentiation of THP-1
cells along the monocytic path and that they affect LPS-induced biological
responses of the cell line.
Collapse
Affiliation(s)
- Katharina Petin
- 1 Institute of Clinical Immunology, Leipzig University, Germany
| | - Ronald Weiss
- 1 Institute of Clinical Immunology, Leipzig University, Germany
| | - Gerd Müller
- 2 Department of Molecular Oncology, Leipzig University, Germany
| | - Antje Garten
- 3 Centre for Paediatric Research Leipzig (CPL), Leipzig University, Germany.,4 Institute of Metabolism and Systems Research, University of Birmingham, UK
| | - Anja Grahnert
- 1 Institute of Clinical Immunology, Leipzig University, Germany
| | - Ulrich Sack
- 1 Institute of Clinical Immunology, Leipzig University, Germany
| | | |
Collapse
|
24
|
|
25
|
Khan A, Shin OS, Na J, Kim JK, Seong RK, Park MS, Noh JY, Song JY, Cheong HJ, Park YH, Kim WJ. A Systems Vaccinology Approach Reveals the Mechanisms of Immunogenic Responses to Hantavax Vaccination in Humans. Sci Rep 2019; 9:4760. [PMID: 30886186 PMCID: PMC6423257 DOI: 10.1038/s41598-019-41205-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 02/27/2019] [Indexed: 01/08/2023] Open
Abstract
Hantavax is an inactivated vaccine for hemorrhagic fever with renal syndrome (HFRS). The immunogenic responses have not been elucidated yet. Here we conducted a cohort study in which 20 healthy subjects were administered four doses of Hantavax during 13-months period. Pre- and post- vaccinated peripheral blood mononuclear cells (PBMCs) and sera were analysed by transcriptomic and metabolomic profilings, respectively. Based on neutralizing antibody titers, subjects were subsequently classified into three groups; non responders (NRs), low responders (LRs) and high responders (HRs). Post vaccination differentially expressed genes (DEGs) associated with innate immunity and cytokine pathways were highly upregulated. DEG analysis revealed a significant induction of CD69 expression in the HRs. High resolution metabolomics (HRM) analysis showed that correlated to the antibody response, cholesteryl nitrolinoleate, octanoyl-carnitine, tyrosine, ubiquinone-9, and benzoate were significantly elevated in HRs, while chenodeoxycholic acid and methyl palmitate were upregulated in NRs and LRs, compared with HRs. Additionally, gene-metabolite interaction revealed upregulated gene-metabolite couplings in, folate biosynthesis, nicotinate and nicotinamide, arachidonic acid, thiamine and pyrimidine metabolism in a dose dependent manner in HR group. Collectively, our data provide new insight into the underlying mechanisms of the Hantavax-mediated immunogenicity in humans.
Collapse
Affiliation(s)
- Adnan Khan
- Metabolomics Laboratory, Korea University College of Pharmacy, Sejeong city, Republic of Korea
| | - Ok Sarah Shin
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jinhyuk Na
- Metabolomics Laboratory, Korea University College of Pharmacy, Sejeong city, Republic of Korea
| | - Jae Kwan Kim
- Metabolomics Laboratory, Korea University College of Pharmacy, Sejeong city, Republic of Korea
| | - Rak-Kyun Seong
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Man-Seong Park
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ji Yun Noh
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Joon Young Song
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hee Jin Cheong
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Youngja Hwang Park
- Metabolomics Laboratory, Korea University College of Pharmacy, Sejeong city, Republic of Korea.
| | - Woo Joo Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea. .,Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
26
|
Minhas PS, Liu L, Moon PK, Joshi AU, Dove C, Mhatre S, Contrepois K, Wang Q, Lee BA, Coronado M, Bernstein D, Snyder MP, Migaud M, Majeti R, Mochly-Rosen D, Rabinowitz JD, Andreasson KI. Macrophage de novo NAD + synthesis specifies immune function in aging and inflammation. Nat Immunol 2018; 20:50-63. [PMID: 30478397 DOI: 10.1038/s41590-018-0255-3] [Citation(s) in RCA: 312] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/10/2018] [Indexed: 11/09/2022]
Abstract
Recent advances highlight a pivotal role for cellular metabolism in programming immune responses. Here, we demonstrate that cell-autonomous generation of nicotinamide adenine dinucleotide (NAD+) via the kynurenine pathway (KP) regulates macrophage immune function in aging and inflammation. Isotope tracer studies revealed that macrophage NAD+ derives substantially from KP metabolism of tryptophan. Genetic or pharmacological blockade of de novo NAD+ synthesis depleted NAD+, suppressed mitochondrial NAD+-dependent signaling and respiration, and impaired phagocytosis and resolution of inflammation. Innate immune challenge triggered upstream KP activation but paradoxically suppressed cell-autonomous NAD+ synthesis by limiting the conversion of downstream quinolinate to NAD+, a profile recapitulated in aging macrophages. Increasing de novo NAD+ generation in immune-challenged or aged macrophages restored oxidative phosphorylation and homeostatic immune responses. Thus, KP-derived NAD+ operates as a metabolic switch to specify macrophage effector responses. Breakdown of de novo NAD+ synthesis may underlie declining NAD+ levels and rising innate immune dysfunction in aging and age-associated diseases.
Collapse
Affiliation(s)
- Paras S Minhas
- Department of Neurology & Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA.,Neurosciences Graduate Program, Stanford University, Stanford, CA, USA
| | - Ling Liu
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.,Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Peter K Moon
- Department of Neurology & Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Amit U Joshi
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Christopher Dove
- Department of Hematology, Stanford School of Medicine, Stanford, CA, USA
| | - Siddhita Mhatre
- Department of Neurology & Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Kevin Contrepois
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Qian Wang
- Department of Neurology & Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Brittany A Lee
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Michael Coronado
- Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA
| | - Daniel Bernstein
- Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA
| | - Michael P Snyder
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Marie Migaud
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Ravindra Majeti
- Department of Hematology, Stanford School of Medicine, Stanford, CA, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.,Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Katrin I Andreasson
- Department of Neurology & Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA. .,Stanford Neuroscience Institute, Stanford University, Stanford, CA, USA. .,Stanford Immunology Program, Stanford University, Stanford, CA, USA.
| |
Collapse
|
27
|
Horenstein AL, Morandi F, Bracci C, Pistoia V, Malavasi F. Functional insights into nucleotide-metabolizing ectoenzymes expressed by bone marrow-resident cells in patients with multiple myeloma. Immunol Lett 2018; 205:40-50. [PMID: 30447309 DOI: 10.1016/j.imlet.2018.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 11/09/2018] [Indexed: 12/18/2022]
Abstract
Human myeloma cells grow in a hypoxic acidic niche in the bone marrow. Cross talk among cellular components of this closed niche generates extracellular adenosine, which promotes tumor cell survival. This is achieved through the binding of adenosine to purinergic receptors into complexes that function as an autocrine/paracrine signal factor with immune regulatory activities that i) down-regulate the functions of most immune effector cells and ii) enhance the activity of cells that suppress anti-tumor immune responses, thus facilitating the escape of malignant myeloma cells from immune surveillance. Here we review recent findings confirming that the dominant phenotype for survival of tumor cells is that where the malignant cells have been metabolically reprogrammed for the generation of lactic acidosis in the bone marrow niche. Adenosine triphosphate and nicotinamide-adenine dinucleotide extruded from tumor cells, along with cyclic adenosine monophosphate, are the main intracellular energetic/messenger molecules that serve as leading substrates in the extracellular space for membrane-bound ectonucleotidases metabolizing purine nucleotides to signaling adenosine. Within this mechanistic framework, the adenosinergic substrate conversion can vary significantly according to the metabolic environment. Indeed, the neoplastic expansion of plasma cells exploits both enzymatic networks and hypoxic acidic conditions for migrating and homing to a protected niche and for evading the immune response. The expression of multiple specific adenosine receptors in the niche completes the profile of a complex regulatory framework whose signals modify multiple myeloma and host immune responses.
Collapse
Affiliation(s)
- A L Horenstein
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Italy; CeRMS, University of Torino, Torino, Italy.
| | - F Morandi
- Stem Cell Laboratory and Cell Therapy Center, Istituto Giannina Gaslini, Genova, Italy
| | - C Bracci
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Italy; CeRMS, University of Torino, Torino, Italy
| | - V Pistoia
- Immunology Area, Pediatric Hospital Bambino Gesù, Rome, Italy
| | - F Malavasi
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Italy; CeRMS, University of Torino, Torino, Italy
| |
Collapse
|
28
|
P2Y 11 Receptors: Properties, Distribution and Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1051:107-122. [PMID: 29134605 DOI: 10.1007/5584_2017_89] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The P2Y11 receptor is a G protein-coupled receptor that is stimulated by endogenous purine nucleotides, particularly ATP. Amongst P2Y receptors it has several unique properties; (1) it is the only human P2Y receptor gene that contains an intron in the coding sequence; (2) the gene does not appear to be present in the rodent genome; (3) it couples to stimulation of both phospholipase C and adenylyl cyclase. Its absence in mice and rats, along with a limited range of selective pharmacological tools, has hampered the development of our knowledge and understanding of its properties and functions. Nonetheless, through a combination of careful use of the available tools, suppression of receptor expression using siRNA and genetic screening for SNPs, possible functions of native P2Y11 receptors have been identified in a variety of human cells and tissues. Many are in blood cells involved in inflammatory responses, consistent with extracellular ATP being a damage-associated signalling molecule in the immune system. Thus proposed potential therapeutic applications relate, in the main, to modulation of acute and chronic inflammatory responses.
Collapse
|
29
|
Extracellular nicotinamide phosphoribosyltransferase (eNAMPT) is a novel marker for patients with BRAF-mutated metastatic melanoma. Oncotarget 2018; 9:18997-19005. [PMID: 29721178 PMCID: PMC5922372 DOI: 10.18632/oncotarget.24871] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/28/2018] [Indexed: 01/09/2023] Open
Abstract
Metastatic melanoma carrying BRAF mutations represent a still unmet medical need as success of BRAF inhibitors is limited by development of resistance. Nicotinamide phosphoribosyltransferase (NAMPT) is a key enzyme in NAD biosynthesis. An extracellular form (eNAMPT) possesses cytokine-like functions and is up-regulated in inflammatory disorders, including cancer. Here we show that eNAMPT is actively released in culture supernatants of melanoma cell lines. Furthermore, cells that become resistant to BRAF inhibitors (BiR) show a significant increase of eNAMPT levels. Plasma from mice xenografted with BiR cell lines contain higher eNAMPT levels compared to tumor-free animals. Consistently, eNAMPT levels are elevated in 113 patients with BRAF-mutated metastatic melanoma compared to 50 with localized disease or to 38 healthy donors, showing a direct correlation with markers of tumor burden, such as LDH, or aggressive disease (such as PD-L1). eNAMPT concentrations decrease in response to therapy with BRAF/MEK inhibitors, but increase again at progression, as inferred from the serial analysis of 50 patients. Lastly, high eNAMPT levels correlate with a significantly shorter overall survival. Our findings suggest that eNAMPT is a novel marker of tumor burden and response to therapy in patients with metastatic melanoma carrying BRAF mutations.
Collapse
|
30
|
Adzic M, Nedeljkovic N. Unveiling the Role of Ecto-5'-Nucleotidase/CD73 in Astrocyte Migration by Using Pharmacological Tools. Front Pharmacol 2018; 9:153. [PMID: 29545748 PMCID: PMC5837971 DOI: 10.3389/fphar.2018.00153] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/13/2018] [Indexed: 01/25/2023] Open
Abstract
CD73 is a bifunctional glycosylphosphatidylinositol (GPI)-anchored membrane protein which functions as ecto-5′-nucleotidase and a membrane receptor for extracellular matrix protein (ECM). A large body of evidence demonstrates a critical involvement of altered purine metabolism and particularly, increased expression of CD73 in a number of human disorders, including cancer and immunodeficiency. Massive up-regulation of CD73 was also found in reactive astrocytes in several experimental models of human neuropathologies. In all the pathological contexts studied so far, the increased expression of CD73 has been associated with the altered ability of cells to adhere and/or migrate. Thus, we hypothesized that increased expression of CD73 in reactive astrocytes has a role in the process of astrocyte adhesion and migration. In the present study, the involvement of CD73 in astrocyte migration was investigated in the scratch wound assay (SW), using primary astrocyte culture prepared from neonatal rat cortex. The cultures were treated with one of the following pharmacological inhibitors which preferentially target individual functions of CD73: (a) α,β-methylene ADP (APCP), which inhibits the catalytic activity of CD73 (b) polyclonal anti-CD73 antibodies, which bind to the internal epitope of CD73 molecule and mask their surface exposure and (c) small interfering CD73-RNA (siCD73), which silences the expression of CD73 gene. It was concluded that approaches that reduce surface expression of CD73 increase migration velocity and promote wound closure in the scratch wound assay, while inhibition of the enzyme activity by APCP induces redistribution of CD73 molecules at the cell surface, thus indirectly affecting cell adhesion and migration. Application of anti-CD73 antibodies induces a decrease in CD73 activity and membrane expression, through CD73 molecules shedding and their release to the culture media. In addition, all applied pharmacological inhibitors differentially affect other aspects of astrocyte function in vitro, including reduced cell proliferation, altered expression of adenosine receptors and increased expression of ERK1/2. Altogether these data imply that CD73 participates in cell adhesion/migration and transmits extracellular signals through interactions with ECM.
Collapse
Affiliation(s)
- Marija Adzic
- Department of General Physiology and Biophysics, Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia.,Centre for Laser Microscopy, Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Nadezda Nedeljkovic
- Department of General Physiology and Biophysics, Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
31
|
Chaker M, Minden A, Chen S, Weiss RH, Chini EN, Mahipal A, Azmi AS. Rho GTPase effectors and NAD metabolism in cancer immune suppression. Expert Opin Ther Targets 2017; 22:9-17. [PMID: 29207896 DOI: 10.1080/14728222.2018.1413091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Sustained proliferative signaling and de-regulated cellular bioenergetics are two of the chief hallmarks of cancer. Alterations in the Ras pathway and its downstream effectors are among the major drivers for uncontrolled cell growth in many cancers. The GTPases are one of the signaling molecules that activate crucial signal transducing pathways downstream of Ras through several effector proteins. The GTPases (GTP bound) interact with several effectors and modulate a number of different biological pathways including those that regulate cytoskeleton, cellular motility, cytokinesis, proliferation, apoptosis, transcription and nuclear signaling. Similarly, the altered glycolytic pathway, the so-called 'Warburg effect', rewires tumor cell metabolism to support the biosynthetic requirements of uncontrolled proliferation. There exists strong evidence for the critical role of the glycolytic pathway's rate limiting enzymes in promoting immunosuppression. Areas covered: We review the emerging roles of GTPase effector proteins particularly the p21 activated kinase 4 (PAK4) and nicotinamide biosynthetic pathway enzyme nicotinamide phosphoribosyltransferase (NAMPT) as signaling molecules in immune surveillance and the immune response. Expert opinion: In this expert opinion article we highlight the recent information on the role of GTPases and the metabolic enzymes on the immune microenvironment and propose some unique immune therapeutic opportunities.
Collapse
Affiliation(s)
- Mahmoud Chaker
- a Department of Oncology , Wayne State University School of Medicine, Karmanos Cancer Institute , Detroit , MI , USA
| | - Audrey Minden
- b Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology , Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey , Piscataway , NJ , USA
| | - Suzie Chen
- b Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology , Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey , Piscataway , NJ , USA
| | - Robert H Weiss
- c Division of Nephrology, Department of Internal Medicine , University of California , Davis , CA , USA.,d Cancer Center , University of California , Davis , CA , USA.,e Medical Service , VA Northern California Health Care System , Mather , CA , USA
| | - Eduardo N Chini
- f Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology , Oncology Research, GI Signaling Center, Mayo Clinic College of Medicine , Rochester , MN , USA
| | - Amit Mahipal
- f Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology , Oncology Research, GI Signaling Center, Mayo Clinic College of Medicine , Rochester , MN , USA
| | - Asfar S Azmi
- a Department of Oncology , Wayne State University School of Medicine, Karmanos Cancer Institute , Detroit , MI , USA
| |
Collapse
|
32
|
Kotova PD, Bystrova MF, Rogachevskaja OA, Khokhlov AA, Sysoeva VY, Tkachuk VA, Kolesnikov SS. Coupling of P2Y receptors to Ca 2+ mobilization in mesenchymal stromal cells from the human adipose tissue. Cell Calcium 2017; 71:1-14. [PMID: 29604959 DOI: 10.1016/j.ceca.2017.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/25/2017] [Accepted: 11/06/2017] [Indexed: 12/19/2022]
Abstract
The purinergic transduction was examined in mesenchymal stromal cells (MSCs) from the human adipose tissue, and several nucleotides, including ATP, UTP, and ADP, were found to mobilize cytosolic Ca2+. Transcripts for multiple purinoreceptors were detected in MSC preparations, including A1, A2A, A2B, P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y13, P2Y14, P2X2, P2X4, and P2X7. Cellular responses to nucleotides were insignificantly sensitive to bath Ca2+, pointing at a minor contribution of Ca2+ entry, and were suppressed by U73122 and 2-APB, implicating the phosphoinositide cascade in coupling P2Y receptors to Ca2+ release. While individual cells were sensitive to several P2Y agonists, responsiveness to a given nucleotide varied from cell to cell, suggesting that particular MSCs could employ different sets of purinoreceptors. Caged Ca2+ stimulated Ca2+-induced Ca2+ release (CICR) that was mediated largely by IP3 receptors, and resultant Ca2+ transients were similar to nucleotide responses by magnitude and kinetics. A variety of findings hinted at CICR to be a universal mechanism that finalizes Ca2+ signaling initiated by agonists in MSCs. Individual MSCs responded to nucleotides in an all-or-nothing manner. Presumably just CICR provided invariant Ca2+ responses observed in MSCs at different nucleotide concentrations. The effects of isoform specific agonists and antagonists suggested that both P2Y1 and P2Y13 were obligatory for ADP responses, while P2Y4 and P2Y11 served as primary UTP and ATP receptors, respectively. Extracellular NAD+ stimulated Ca2+ signaling in each ATP-responsive MSC by involving P2Y11. The overall data indicate that extracellular nucleotides and NAD+ can serve as autocrine/paracrine factors regulating MSC functions.
Collapse
Affiliation(s)
- Polina D Kotova
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutional Street 3, Pushchino, Moscow Region, 142290, Russia
| | - Marina F Bystrova
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutional Street 3, Pushchino, Moscow Region, 142290, Russia
| | - Olga A Rogachevskaja
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutional Street 3, Pushchino, Moscow Region, 142290, Russia
| | - Alexander A Khokhlov
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutional Street 3, Pushchino, Moscow Region, 142290, Russia
| | - Veronika Yu Sysoeva
- Department of Biochemistry and Molecular Medicine, Faculty of Basic Medicine, Lomonosov Moscow State University, Russia
| | - Vsevolod A Tkachuk
- Department of Biochemistry and Molecular Medicine, Faculty of Basic Medicine, Lomonosov Moscow State University, Russia
| | - Stanislav S Kolesnikov
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutional Street 3, Pushchino, Moscow Region, 142290, Russia.
| |
Collapse
|
33
|
Muller-Steffner H, Jacques SA, Kuhn I, Schultz MD, Botta D, Osswald P, Maechling C, Lund FE, Kellenberger E. Efficient Inhibition of SmNACE by Coordination Complexes Is Abolished by S. mansoni Sequestration of Metal. ACS Chem Biol 2017; 12:1787-1795. [PMID: 28481502 DOI: 10.1021/acschembio.7b00186] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
SmNACE is a NAD catabolizing enzyme expressed on the outer tegument of S. mansoni, a human parasite that is one of the major agents of the neglected tropical disease schistosomiasis. Recently, we identified aroylhydrazone derivatives capable of inhibiting the recombinant form of the enzyme with variable potency (IC50 ranging from 88 μM to 33 nM). In the present study, we investigated the mechanism of action of the least potent micromolar inhibitor (compound 1) and the most potent nanomolar inhibitor (compound 2) in the series on both the recombinant and native SmNACE enzymes. Using mass spectroscopy, spectrophotometry, and activity assays under different experimental conditions, we demonstrated that the >3 log gain in potency against recombinant SmNACE by this class of compounds is dependent on the formation of a coordination complex with metal cations, such as Ni(II), Zn(II), and Fe(II), that are loaded on the protein surface. Testing the compounds on live parasites, we observed that only the weak micromolar compound 1 was active on the native enzyme. We showed that S. mansoni effectively sequesters the metal from the coordination complex, resulting in the loss of inhibitory activity of the potent nanomolar compound 2. Importantly, the modeling of the transition complex between Zn(II) and compound 2 enabled the discovery of a new metal-independent aroylhydrazone analogue, which is now the most potent and selective inhibitor of native SmNACE known.
Collapse
Affiliation(s)
- Hélène Muller-Steffner
- Laboratoire des Systèmes Chimiques Fonctionnels, CAMB UMR 7199 CNRS-Université de Strasbourg, MEDALIS Drug Discovery Center,
Faculté de Pharmacie, 67400 Illkirch, France
| | - Sylvain A. Jacques
- Laboratoire d’Innovation Thérapeutique, LIT UMR 7200 CNRS-Université de Strasbourg, MEDALIS Drug Discovery Center,
Faculté de Pharmacie, 67400 Illkirch, France
| | - Isabelle Kuhn
- Laboratoire des Systèmes Chimiques Fonctionnels, CAMB UMR 7199 CNRS-Université de Strasbourg, MEDALIS Drug Discovery Center,
Faculté de Pharmacie, 67400 Illkirch, France
| | - Michael D. Schultz
- Department
of Microbiology, University of Alabama at Birmingham, 276 BBRB Box
11, 1720 Second Avenue South, Birmingham, Alabama, United States
| | - Davide Botta
- Department
of Microbiology, University of Alabama at Birmingham, 276 BBRB Box
11, 1720 Second Avenue South, Birmingham, Alabama, United States
| | - Paul Osswald
- Laboratoire des Systèmes Chimiques Fonctionnels, CAMB UMR 7199 CNRS-Université de Strasbourg, MEDALIS Drug Discovery Center,
Faculté de Pharmacie, 67400 Illkirch, France
| | - Clarisse Maechling
- Laboratoire d’Innovation Thérapeutique, LIT UMR 7200 CNRS-Université de Strasbourg, MEDALIS Drug Discovery Center,
Faculté de Pharmacie, 67400 Illkirch, France
| | - Frances E. Lund
- Department
of Microbiology, University of Alabama at Birmingham, 276 BBRB Box
11, 1720 Second Avenue South, Birmingham, Alabama, United States
| | - Esther Kellenberger
- Laboratoire d’Innovation Thérapeutique, LIT UMR 7200 CNRS-Université de Strasbourg, MEDALIS Drug Discovery Center,
Faculté de Pharmacie, 67400 Illkirch, France
| |
Collapse
|
34
|
Williams AC, Hill LJ. Meat and Nicotinamide: A Causal Role in Human Evolution, History, and Demographics. Int J Tryptophan Res 2017; 10:1178646917704661. [PMID: 28579800 PMCID: PMC5417583 DOI: 10.1177/1178646917704661] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/15/2017] [Indexed: 01/15/2023] Open
Abstract
Hunting for meat was a critical step in all animal and human evolution. A key brain-trophic element in meat is vitamin B3 / nicotinamide. The supply of meat and nicotinamide steadily increased from the Cambrian origin of animal predators ratcheting ever larger brains. This culminated in the 3-million-year evolution of Homo sapiens and our overall demographic success. We view human evolution, recent history, and agricultural and demographic transitions in the light of meat and nicotinamide intake. A biochemical and immunological switch is highlighted that affects fertility in the 'de novo' tryptophan-to-kynurenine-nicotinamide 'immune tolerance' pathway. Longevity relates to nicotinamide adenine dinucleotide consumer pathways. High meat intake correlates with moderate fertility, high intelligence, good health, and longevity with consequent population stability, whereas low meat/high cereal intake (short of starvation) correlates with high fertility, disease, and population booms and busts. Too high a meat intake and fertility falls below replacement levels. Reducing variances in meat consumption might help stabilise population growth and improve human capital.
Collapse
Affiliation(s)
- Adrian C Williams
- Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Lisa J Hill
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
35
|
Wang HY, Ma JL, Yin BC, Ye BC. Nicotinamide adenine dinucleotide detection based on silver nanoclusters stabilized by a dumbbell-shaped probe. Analyst 2017; 142:1765-1771. [PMID: 28425549 DOI: 10.1039/c7an00293a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We have developed a novel method for detecting nicotinamide adenine dinucleotide (NAD+) based on fluorescent silver nanoclusters (AgNCs) stabilized by a dumbbell-shaped DNA template containing two cytosine-loops joined in a dsDNA stem. The design involves two types of components: a dumbbell-shaped DNA template and three enzymes. In the presence of NAD+ as a cofactor, Escherichia coli DNA ligase (E.coli DNA ligase) catalyzes template ligation to generate a sealed (no terminal nucleotides) dumbbell-shaped structure, preventing digestion by exonuclease III (Exo III) and exonuclease I (Exo I). The loop regions of the intact template serve as sites for the deposition of highly fluorescent AgNCs. In the absence of NAD+, the ligation reaction does not occur, and the unsealed dumbbell-shaped template is digested into mononucleotides via cooperation of Exo III and Exo I. The destruction of the DNA template results in the agglomeration of AgNCs into silver nanoparticles with low fluorescence. The fluorescence enhancement depends on the ligation and digestion of the DNA template, allowing quantitative detection of NAD+ in the range of 0.5 nM-5000 nM with a detection limit of ∼0.25 nM.
Collapse
Affiliation(s)
- Hong-Ya Wang
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science & Technology, Shanghai, 200237, China.
| | - Jin-Liang Ma
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science & Technology, Shanghai, 200237, China.
| | - Bin-Cheng Yin
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science & Technology, Shanghai, 200237, China.
| | - Bang-Ce Ye
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science & Technology, Shanghai, 200237, China. and School of Chemistry and Chemical Engineering, Shihezi University, Xinjiang, 832000, China
| |
Collapse
|
36
|
Chen J, Sysol JR, Singla S, Zhao S, Yamamura A, Valdez-Jasso D, Abbasi T, Shioura KM, Sahni S, Reddy V, Sridhar A, Gao H, Torres J, Camp SM, Tang H, Ye SQ, Comhair S, Dweik R, Hassoun P, Yuan JXJ, Garcia JGN, Machado RF. Nicotinamide Phosphoribosyltransferase Promotes Pulmonary Vascular Remodeling and Is a Therapeutic Target in Pulmonary Arterial Hypertension. Circulation 2017; 135:1532-1546. [PMID: 28202489 DOI: 10.1161/circulationaha.116.024557] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 02/06/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pulmonary arterial hypertension is a severe and progressive disease, a hallmark of which is pulmonary vascular remodeling. Nicotinamide phosphoribosyltransferase (NAMPT) is a cytozyme that regulates intracellular nicotinamide adenine dinucleotide levels and cellular redox state, regulates histone deacetylases, promotes cell proliferation, and inhibits apoptosis. We hypothesized that NAMPT promotes pulmonary vascular remodeling and that inhibition of NAMPT could attenuate pulmonary hypertension. METHODS Plasma, mRNA, and protein levels of NAMPT were measured in the lungs and isolated pulmonary artery endothelial cells from patients with pulmonary arterial hypertension and in the lungs of rodent models of pulmonary hypertension. Nampt+/- mice were exposed to 10% hypoxia and room air for 4 weeks, and the preventive and therapeutic effects of NAMPT inhibition were tested in the monocrotaline and Sugen hypoxia models of pulmonary hypertension. The effects of NAMPT activity on proliferation, migration, apoptosis, and calcium signaling were tested in human pulmonary artery smooth muscle cells. RESULTS Plasma and mRNA and protein levels of NAMPT were increased in the lungs and isolated pulmonary artery endothelial cells from patients with pulmonary arterial hypertension, as well as in lungs of rodent models of pulmonary hypertension. Nampt+/- mice were protected from hypoxia-mediated pulmonary hypertension. NAMPT activity promoted human pulmonary artery smooth muscle cell proliferation via a paracrine effect. In addition, recombinant NAMPT stimulated human pulmonary artery smooth muscle cell proliferation via enhancement of store-operated calcium entry by enhancing expression of Orai2 and STIM2. Last, inhibition of NAMPT activity attenuated monocrotaline and Sugen hypoxia-induced pulmonary hypertension in rats. CONCLUSIONS Our data provide evidence that NAMPT plays a role in pulmonary vascular remodeling and that its inhibition could be a potential therapeutic target for pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Jiwang Chen
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Justin R Sysol
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Sunit Singla
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Shuangping Zhao
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Aya Yamamura
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Daniela Valdez-Jasso
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Taimur Abbasi
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Krystyna M Shioura
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Sakshi Sahni
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Vamsi Reddy
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Arvind Sridhar
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Hui Gao
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Jaime Torres
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Sara M Camp
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Haiyang Tang
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Shui Q Ye
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Suzy Comhair
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Raed Dweik
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Paul Hassoun
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Jason X-J Yuan
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Joe G N Garcia
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.).
| | - Roberto F Machado
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.).
| |
Collapse
|
37
|
Zimmermann H. Extracellular ATP and other nucleotides-ubiquitous triggers of intercellular messenger release. Purinergic Signal 2015; 12:25-57. [PMID: 26545760 DOI: 10.1007/s11302-015-9483-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 10/29/2015] [Indexed: 12/21/2022] Open
Abstract
Extracellular nucleotides, and ATP in particular, are cellular signal substances involved in the control of numerous (patho)physiological mechanisms. They provoke nucleotide receptor-mediated mechanisms in select target cells. But nucleotides can considerably expand their range of action. They function as primary messengers in intercellular communication by stimulating the release of other extracellular messenger substances. These in turn activate additional cellular mechanisms through their own receptors. While this applies also to other extracellular messengers, its omnipresence in the vertebrate organism is an outstanding feature of nucleotide signaling. Intercellular messenger substances released by nucleotides include neurotransmitters, hormones, growth factors, a considerable variety of other proteins including enzymes, numerous cytokines, lipid mediators, nitric oxide, and reactive oxygen species. Moreover, nucleotides activate or co-activate growth factor receptors. In the case of hormone release, the initially paracrine or autocrine nucleotide-mediated signal spreads through to the entire organism. The examples highlighted in this commentary suggest that acting as ubiquitous triggers of intercellular messenger release is one of the major functional roles of extracellular nucleotides. While initiation of messenger release by nucleotides has been unraveled in many contexts, it may have been overlooked in others. It can be anticipated that additional nucleotide-driven messenger functions will be uncovered with relevance for both understanding physiology and development of therapy.
Collapse
Affiliation(s)
- Herbert Zimmermann
- Institute of Cell Biology and Neuroscience, Molecular and Cellular Neurobiology, Goethe University, Max-von-Laue-Str. 13, Frankfurt am Main, Germany.
| |
Collapse
|
38
|
NAD⁺-Metabolizing Ectoenzymes in Remodeling Tumor-Host Interactions: The Human Myeloma Model. Cells 2015; 4:520-37. [PMID: 26393653 PMCID: PMC4588049 DOI: 10.3390/cells4030520] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/09/2015] [Accepted: 09/14/2015] [Indexed: 11/17/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD⁺) is an essential co-enzyme reported to operate both intra- and extracellularly. In the extracellular space, NAD⁺ can elicit signals by binding purinergic P2 receptors or it can serve as the substrate for a chain of ectoenzymes. As a substrate, it is converted to adenosine (ADO) and then taken up by the cells, where it is transformed and reincorporated into the intracellular nucleotide pool. Nucleotide-nucleoside conversion is regulated by membrane-bound ectoenzymes. CD38, the main mammalian enzyme that hydrolyzes NAD⁺, belongs to the ectoenzymatic network generating intracellular Ca(2+)-active metabolites. Within this general framework, the extracellular conversion of NAD⁺ can vary significantly according to the tissue environment or pathological conditions. Accumulating evidence suggests that tumor cells exploit such a network for migrating and homing to protected areas and, even more importantly, for evading the immune response. We report on the experience of this lab to exploit human multiple myeloma (MM), a neoplastic expansion of plasma cells, as a model to investigate these issues. MM cells express high levels of surface CD38 and grow in an environment prevalently represented by closed niches hosted in the bone marrow (BM). An original approach of this study derives from the recent use of the clinical availability of therapeutic anti-CD38 monoclonal antibodies (mAbs) in perturbing tumor viability and enzymatic functions in conditions mimicking what happens in vivo.
Collapse
|
39
|
Chiang SH, Harrington WW, Luo G, Milliken NO, Ulrich JC, Chen J, Rajpal DK, Qian Y, Carpenter T, Murray R, Geske RS, Stimpson SA, Kramer HF, Haffner CD, Becherer JD, Preugschat F, Billin AN. Genetic Ablation of CD38 Protects against Western Diet-Induced Exercise Intolerance and Metabolic Inflexibility. PLoS One 2015; 10:e0134927. [PMID: 26287487 PMCID: PMC4546114 DOI: 10.1371/journal.pone.0134927] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 07/15/2015] [Indexed: 01/01/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a key cofactor required for essential metabolic oxidation-reduction reactions. It also regulates various cellular activities, including gene expression, signaling, DNA repair and calcium homeostasis. Intracellular NAD+ levels are tightly regulated and often respond rapidly to nutritional and environmental changes. Numerous studies indicate that elevating NAD+ may be therapeutically beneficial in the context of numerous diseases. However, the role of NAD+ on skeletal muscle exercise performance is poorly understood. CD38, a multi-functional membrane receptor and enzyme, consumes NAD+ to generate products such as cyclic-ADP-ribose. CD38 knockout mice show elevated tissue and blood NAD+ level. Chronic feeding of high-fat, high-sucrose diet to wild type mice leads to exercise intolerance and reduced metabolic flexibility. Loss of CD38 by genetic mutation protects mice from diet-induced metabolic deficit. These animal model results suggest that elevation of tissue NAD+ through genetic ablation of CD38 can profoundly alter energy homeostasis in animals that are maintained on a calorically-excessive Western diet.
Collapse
Affiliation(s)
- Shian-Huey Chiang
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
- * E-mail:
| | - W. Wallace Harrington
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Guizhen Luo
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Naphtali O. Milliken
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - John C. Ulrich
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Jing Chen
- QSCI Computational Biology, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Deepak K. Rajpal
- QSCI Computational Biology, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Ying Qian
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Tiffany Carpenter
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Rusty Murray
- Laboratory Animal Science, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Robert S. Geske
- Target & Pathway Validation, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Stephen A. Stimpson
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Henning F. Kramer
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Curt D. Haffner
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - J. David Becherer
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Frank Preugschat
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| | - Andrew N. Billin
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, Research Triangle Park, North Carolina, 27709, United States of America
| |
Collapse
|
40
|
Vidugiriene J, Leippe D, Sobol M, Vidugiris G, Zhou W, Meisenheimer P, Gautam P, Wennerberg K, Cali JJ. Bioluminescent cell-based NAD(P)/NAD(P)H assays for rapid dinucleotide measurement and inhibitor screening. Assay Drug Dev Technol 2015; 12:514-26. [PMID: 25506801 DOI: 10.1089/adt.2014.605] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Abstract The central role of nicotinamide adenine dinucleotides in cellular energy metabolism and signaling makes them important nodes that link the metabolic state of cells with energy homeostasis and gene regulation. In this study, we describe the implementation of cell-based bioluminescence assays for rapid and sensitive measurement of those important redox cofactors. We show that the sensitivity of the assays (limit of detection ∼0.5 nM) enables the selective detection of total amounts of nonphosphorylated or phosphorylated dinucleotides directly in cell lysates. The total amount of NAD+NADH or NADP+NADPH levels can be detected in as low as 300 or 600 cells/well, respectively. The signal remains linear up to 5,000 cells/well with the maximum signal-to-background ratios ranging from 100 to 200 for NAD+NADH and from 50 to 100 for NADP+NADPH detection. The assays are robust (Z' value >0.7) and the inhibitor response curves generated using a known NAD biosynthetic pathway inhibitor FK866 correlate well with the reported data. More importantly, by multiplexing the dinucleotide detection assays with a fluorescent nonmetabolic cell viability assay, we show that dinucleotide levels can be decreased dramatically (>80%) by FK866 treatment before changes in cell viability are detected. The utility of the assays to identify modulators of intracellular nicotinamide adenine dinucleotide levels was further confirmed using an oncology active compound library, where novel dinucleotide regulating compounds were identified. For example, the histone deacetylase inhibitor entinostat was a potent inhibitor of cellular nicotinamide adenine dinucleotides, whereas the selective estrogen receptor modulator raloxifene unexpectedly caused a twofold increase in cellular nicotinamide adenine dinucleotide levels.
Collapse
|
41
|
Effect of Leflunomide on the Abnormal Expression of Lipid Rafts and F-Actin in B Lymphocytes from Patients with Systemic Lupus Erythematosus. J Immunol Res 2015; 2015:832916. [PMID: 26090500 PMCID: PMC4452103 DOI: 10.1155/2015/832916] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 11/18/2022] Open
Abstract
Purposes. To investigate the possible changes in B cell subsets and in B cell expression patterns of lipid rafts (LRs) and F-actin in patients with SLE and whether leflunomide treatment may have effect on these changes. Methods. The B cell subsets and LRs expression were determined by flow cytometry and confocal microscopy, and F-actin expression was examined by confocal microscopy. Results. CD27+IgD+ B cell subsets were significantly decreased while CD38+CD95+ B cell subsets increased in SLE patients. The LRs levels of B cells were remarkably increased and positively correlated with SLEDAI and anti-dsDNA titer in SLE patients. The expression level of LRs was significantly higher in CD38+ B cells than CD38− B cells and negatively correlated with C3 levels. The increased expression of LRs was associated with reduced expression of F-actin in the B cells from active SLE patients. Furthermore, in vitro treatment of the cells with A771726 reduced the expression level of LRs, attenuated the overaggregation of LRs, and normalized the distribution of F-actin. Conclusions. There were abnormalities in B cell subsets and LRs and F-actin expression of B cell from SLE patients. Modulation of B cell expression of LRs and F-actin by LEF could be a potential therapeutic target for SLE.
Collapse
|
42
|
CD38 is expressed on inflammatory cells of the intestine and promotes intestinal inflammation. PLoS One 2015; 10:e0126007. [PMID: 25938500 PMCID: PMC4418770 DOI: 10.1371/journal.pone.0126007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 03/27/2015] [Indexed: 12/20/2022] Open
Abstract
The enzyme CD38 is expressed on a variety of hematopoietic and non-hematopoietic cells and is involved in diverse processes such as generation of calcium-mobilizing metabolites, cell activation, and chemotaxis. Here, we show that under homeostatic conditions CD38 is highly expressed on immune cells of the colon mucosa of C57BL/6 mice. Myeloid cells recruited to this tissue upon inflammation also express enhanced levels of CD38. To determine the role of CD38 in intestinal inflammation, we applied the dextran sulfate sodium (DSS) colitis model. Whereas wild-type mice developed severe colitis, CD38-/- mice had only mild disease following DSS-treatment. Histologic examination of the colon mucosa revealed pronounced inflammatory damage with dense infiltrates containing numerous granulocytes and macrophages in wild-type animals, while these findings were significantly attenuated in CD38-/- mice. Despite attenuated histological findings, the mRNA expression of inflammatory cytokines and chemokines was only marginally lower in the colons of CD38-/- mice as compared to wild-type mice. In conclusion, our results identify a function for CD38 in the control of inflammatory processes in the colon.
Collapse
|
43
|
Hasan MA, Sultan MT, Ahn WG, Kim YJ, Jang JH, Hong CW, Song DK. Scant Extracellular NAD Cleaving Activity of Human Neutrophils is Down-Regulated by fMLP via FPRL1. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:497-502. [PMID: 25598664 PMCID: PMC4296039 DOI: 10.4196/kjpp.2014.18.6.497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 08/26/2014] [Accepted: 09/05/2014] [Indexed: 11/15/2022]
Abstract
Extracellular nicotinamide adenine dinucleotide (NAD) cleaving activity of a particular cell type determines the rate of the degradation of extracellular NAD with formation of metabolites in the vicinity of the plasma membrane, which has important physiological consequences. It is yet to be elucidated whether intact human neutrophils have any extracellular NAD cleaving activity. In this study, with a simple fluorometric assay utilizing 1,N6-ethenoadenine dinucleotide (etheno-NAD) as the substrate, we have shown that intact peripheral human neutrophils have scant extracellular etheno-NAD cleaving activity, which is much less than that of mouse bone marrow neutrophils, mouse peripheral neutrophils, human monocytes and lymphocytes. With high performance liquid chromatography (HPLC), we have identified that ADP-ribose (ADPR) is the major extracellular metabolite of NAD degradation by intact human neutrophils. The scant extracellular etheno-NAD cleaving activity is decreased further by N-formyl-methionine-leucine-phenylalanine (fMLP), a chemoattractant for neutrophils. The fMLP-mediated decrease in the extracellular etheno-NAD cleaving activity is reversed by WRW4, a potent FPRL1 antagonist. These findings show that a much less extracellular etheno-NAD cleaving activity of intact human neutrophils compared to other immune cell types is down-regulated by fMLP via a low affinity fMLP receptor FPRL1.
Collapse
Affiliation(s)
- Md Ashraful Hasan
- Department of Pharmacology, Infectious Diseases Medical Research Center, College of Medicine, Hallym University, Chuncheon 200-702, Korea
| | - Md Tipu Sultan
- Department of Pharmacology, Infectious Diseases Medical Research Center, College of Medicine, Hallym University, Chuncheon 200-702, Korea
| | - Won-Gyun Ahn
- Department of Pharmacology, Infectious Diseases Medical Research Center, College of Medicine, Hallym University, Chuncheon 200-702, Korea
| | - Yeon-Ja Kim
- Department of Pharmacology, Infectious Diseases Medical Research Center, College of Medicine, Hallym University, Chuncheon 200-702, Korea
| | - Ji-Hye Jang
- Department of Pharmacology, Infectious Diseases Medical Research Center, College of Medicine, Hallym University, Chuncheon 200-702, Korea
| | - Chang-Won Hong
- Department of Pharmacology, Infectious Diseases Medical Research Center, College of Medicine, Hallym University, Chuncheon 200-702, Korea
| | - Dong-Keun Song
- Department of Pharmacology, Infectious Diseases Medical Research Center, College of Medicine, Hallym University, Chuncheon 200-702, Korea
| |
Collapse
|
44
|
Nicotinamide phosphoribosyltransferase/pre-B-cell colony enhancing factor/visfatin plasma levels and clinical outcome in patients with dilated cardiomyopathy. J Card Fail 2014; 21:330-8. [PMID: 25498756 DOI: 10.1016/j.cardfail.2014.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 11/30/2014] [Accepted: 12/03/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND Nicotinamide phosphoribosyltransferase (Nampt) is an enzyme involved in nicotinamide adenine dinucleotide biosynthesis. Nampt functions as gatekeeper of energy status and survival in cardiac myocytes in animal models of ischemia-reperfusion and might regulate inflammatory processes. Therefore, we performed for the 1st time a clinical study to determine the effects of Nampt on cardiac function in patients with nonischemic dilated (DCM) and inflammatory (DCMi) cardiomyopathy. METHODS AND RESULTS A total of 113 patients were enrolled in the study and classified into control (n = 25), DCM (n = 38), and DCMi (n = 50) groups. Cardiac functional and inflammatory parameters as well as plasma Nampt and cardiac mRNA and protein Nampt expression were determined at baseline and follow-up after 6 months. Patients with DCM (1.04 ± 0.8 ng/mL; P < .001) and DCMi (1.07 ± 0.7 ng/mL; P < .001) showed significantly increased Nampt plasma concentrations at baseline compared with the control group (0.57 ± 0.5 ng/mL). Patients with higher Nampt concentrations in both heart failure groups showed significant better improvement of cardiac functional parameters (correlation between Nampt plasma levels and the change of left ventricular ejection fraction after 6 months: DCM: r = 0.698, P < .001; DCMi: r = 0.503, P < .001). Moreover, cardiac inflammation did not influence Nampt expression, and Nampt concentrations did not modulate cardiac inflammation in DCMi. A multivariate linear regression model revealed high plasma Nampt expression to contribute to better improvement of cardiac function in patients of both heart failure groups. Moreover, heart failure patients with high plasma Nampt levels showed suppressed cardiac TNF-α and IL-6 mRNA expression after 6 months' follow-up as well as lower B-type natriuretic peptide levels compared with heart failure patients with low Nampt plasma concentrations. CONCLUSIONS High Nampt expression in patients with nonischemic DCM and DCMi is associated with a favorable outcome and improvement in functional status.
Collapse
|
45
|
Zhang HS, Chen XY, Wu TC, Zhang FJ. Tanshinone II A Inhibits Tat-Induced HIV-1 Transactivation Through Redox-Regulated AMPK/Nampt Pathway. J Cell Physiol 2014; 229:1193-201. [DOI: 10.1002/jcp.24552] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 01/07/2014] [Indexed: 12/14/2022]
Affiliation(s)
- Hong-Sheng Zhang
- College of Life Science & Bioengineering; Beijing University of Technology; Pingleyuan 100# District of Chaoyang Beijing China
| | - Xin-Yu Chen
- College of Life Science & Bioengineering; Beijing University of Technology; Pingleyuan 100# District of Chaoyang Beijing China
| | - Tong-Chao Wu
- College of Life Science & Bioengineering; Beijing University of Technology; Pingleyuan 100# District of Chaoyang Beijing China
| | - Feng-Juan Zhang
- College of Life Science & Bioengineering; Beijing University of Technology; Pingleyuan 100# District of Chaoyang Beijing China
| |
Collapse
|
46
|
Pecchi E, Priam S, Gosset M, Pigenet A, Sudre L, Laiguillon MC, Berenbaum F, Houard X. Induction of nerve growth factor expression and release by mechanical and inflammatory stimuli in chondrocytes: possible involvement in osteoarthritis pain. Arthritis Res Ther 2014; 16:R16. [PMID: 24438745 PMCID: PMC3978639 DOI: 10.1186/ar4443] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 01/03/2014] [Indexed: 01/05/2023] Open
Abstract
Introduction Nerve growth factor (NGF) level is increased in osteoarthritis (OA) joints and is involved in pain associated with OA. Stimuli responsible for NGF stimulation in chondrocytes are unknown. We investigated whether mechanical stress and proinflammatory cytokines may influence NGF synthesis by chondrocytes. Methods Primary cultures of human OA chondrocytes, newborn mouse articular chondrocytes or cartilage explants were stimulated by increasing amounts of IL-1β, prostaglandin E2 (PGE2), visfatin/nicotinamide phosphoribosyltransferase (NAMPT) or by cyclic mechanical compression (0.5 Hz, 1 MPa). Before stimulation, chondrocytes were pretreated with indomethacin, Apo866, a specific inhibitor of NAMPT enzymatic activity, or transfected by siRNA targeting visfatin/NAMPT. mRNA NGF levels were assessed by real-time quantitative PCR and NGF released into media was determined by ELISA. Results Unstimulated human and mouse articular chondrocytes expressed low levels of NGF (19.2 ± 8.7 pg/mL, 13.5 ± 1.0 pg/mL and 4.4 ± 0.8 pg/mL/mg tissue for human and mouse articular chondrocytes and costal explants, respectively). Mechanical stress induced NGF release in conditioned media. When stimulated by IL-1β or visfatin/NAMPT, a proinflammatory adipokine produced by chondocytes in response to IL-1β, a dose-dependent increase in NGF mRNA expression and NGF release in both human and mouse chondrocyte conditioned media was observed. Visfatin/NAMPT is also an intracellular enzyme acting as the rate-limiting enzyme of the generation of NAD. The expression of NGF induced by visfatin/NAMPT was inhibited by Apo866, whereas IL-1β-mediated NGF expression was not modified by siRNA targeting visfatin/NAMPT. Interestingly, PGE2, which is produced by chondrocytes in response to IL-1β and visfatin/NAMPT, did not stimulate NGF production. Consistently, indomethacin, a cyclooxygenase inhibitor, did not counteract IL-1β-induced NGF production. Conclusions These results show that mechanical stress, IL-1β and extracellular visfatin/NAMPT, all stimulated the expression and release of NGF by chondrocytes and thus suggest that the overexpression of visfatin/NAMPT and IL-1β in the OA joint and the increased mechanical loading of cartilage may mediate OA pain via the stimulation of NGF expression and release by chondrocytes.
Collapse
|
47
|
Antagonistic crosstalk between NF-κB and SIRT1 in the regulation of inflammation and metabolic disorders. Cell Signal 2013; 25:1939-48. [DOI: 10.1016/j.cellsig.2013.06.007] [Citation(s) in RCA: 582] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 06/04/2013] [Indexed: 02/07/2023]
|
48
|
Horenstein AL, Chillemi A, Zaccarello G, Bruzzone S, Quarona V, Zito A, Serra S, Malavasi F. A CD38/CD203a/CD73 ectoenzymatic pathway independent of CD39 drives a novel adenosinergic loop in human T lymphocytes. Oncoimmunology 2013; 2:e26246. [PMID: 24319640 PMCID: PMC3850273 DOI: 10.4161/onci.26246] [Citation(s) in RCA: 203] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 08/20/2013] [Accepted: 08/21/2013] [Indexed: 11/19/2022] Open
Abstract
The tumor microenvironment is characterized by of high levels of extracellular nucleotides that are metabolized through the dynamic and sequential action of cell surface enzymes (ectoenzymes). These ectoenzymes operate according to their spatial arrangement, as part of (1) continuous (molecules on the same cell) or (2) discontinuous (molecules on different cells) pathways, the latter being facilitated by restricted cellular microenvironment. The outcome of this catabolic activity is an increase in the local concentration of adenosine, a nucleoside involved in the control of inflammation and immune responses. The aim of the work presented here was to demonstrate that a previously unexplored enzymatic pathway may be an alternate route to produce extracellular adenosine. Our data show that this new axis is driven by the nucleotide-metabolizing ectoenzymes CD38 (an NAD+ nucleosidase), the ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (NPP1, also known as CD203a or PC-1) and the 5′ ectonucleotidase (5′-NT) CD73, while bypassing the canonical catabolic pathway mediated by the nucleoside tri- and diphosphohydrolase (NTPDase) CD39. To determine the relative contributions of these cell surface enzymes to the production of adenosine, we exploited a human T-cell model allowing for the modular expression of the individual components of this alternative pathway upon activation and transfection. The biochemical analysis of the products of these ectoenzymes by high-performance liquid chromatography (HPLC) fully substantiated our working hypothesis. This newly characterized pathway may facilitate the emergence of an adaptive immune response in selected cellular contexts. Considering the role for extracellular adenosine in the regulation of inflammation and immunogenicity, this pathway could constitute a novel strategy of tumor evasion, implying that these enzymes may represent ideal targets for antibody-mediated therapy.
Collapse
Affiliation(s)
- Alberto L Horenstein
- Laboratory of Immunogenetics; Department of Medical Sciences; University of Torino; Torino, Italy ; Research Center for Experimental Medicine; University of Torino; Torino, Italy ; Transplantation Immunology; "Città della Salute e della Scienza" Hospital; Torino, Italy
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Rosado MM, Bennici E, Novelli F, Pioli C. Beyond DNA repair, the immunological role of PARP-1 and its siblings. Immunology 2013; 139:428-37. [PMID: 23489378 DOI: 10.1111/imm.12099] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 02/16/2013] [Accepted: 03/11/2013] [Indexed: 12/14/2022] Open
Abstract
ADP-ribosylation is the addition of one or more (up to some hundreds) ADP-ribose moieties to acceptor proteins. There are two major families of enzymes that catalyse this reaction: extracellular ADP-ribosyl-transferases (ARTs), which are bound to the cell membrane by a glycosylphosphatidylinositol anchor or are secreted, and poly(ADP-ribose)-polymerases (PARPs), which are present in the cell nucleus and/or cytoplasm. Recent findings revealed a wide immunological role for ADP-ribosylating enzymes. ARTs, by sensing extracellular NAD concentration, can act as danger detectors. PARP-1, the prototypical representative of the PARP family, known to protect cells from genomic instability, is involved in the development of inflammatory responses and several forms of cell death. PARP-1 also plays a role in adaptive immunity by modulating the ability of dendritic cells to stimulate T cells or by directly affecting the differentiation and functions of T and B cells. Both PARP-1 and PARP-14 (CoaSt6) knockout mice were described to display reduced T helper type 2 cell differentiation and allergic responses. Our recent findings showed that PARP-1 is involved in the differentiation of Foxp3+ regulatory T (Treg) cells, suggesting a role for PARP-1 in tolerance induction. Also ARTs regulate Treg cell homeostasis by promoting Treg cell apoptosis during inflammatory responses. PARP inhibitors ameliorate immune-mediated diseases in several experimental models, including rheumatoid arthritis, colitis, experimental autoimmune encephalomyelitis and allergy. Together these findings show that ADP-ribosylating enzymes, in particular PARP-1, play a pivotal role in the regulation of immune responses and may represent a good target for new therapeutic approaches in immune-mediated diseases.
Collapse
Affiliation(s)
- Maria Manuela Rosado
- Laboratory of B cell development, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | | | | | | |
Collapse
|
50
|
Di Stefano M, Conforti L. Diversification of NAD biological role: the importance of location. FEBS J 2013; 280:4711-28. [PMID: 23848828 DOI: 10.1111/febs.12433] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Accepted: 07/08/2013] [Indexed: 02/03/2023]
Abstract
Over 100 years after its first discovery, several new aspects of the biology of the redox co-factor NAD are rapidly emerging. NAD, as well as its precursors, its derivatives, and its metabolic enzymes, have been recently shown to play a determinant role in a variety of biological functions, from the classical role in oxidative phosphorylation and redox reactions to a role in regulation of gene transcription, lifespan and cell death, from a role in neurotransmission to a role in axon degeneration, and from a function in regulation of glucose homeostasis to that of control of circadian rhythm. It is also becoming clear that this variety of specialized functions is regulated by the fine subcellular localization of NAD, its related nucleotides and its metabolic enzymatic machinery. Here we describe the known NAD biosynthetic and catabolic pathways, and review evidence supporting a specialized role for NAD metabolism in a subcellular compartment-dependent manner.
Collapse
Affiliation(s)
- Michele Di Stefano
- School of Biomedical Sciences, University of Nottingham Medical School, Queen's Medical Centre, UK
| | | |
Collapse
|