1
|
Lagunas-Rangel FA. Structural Insights Into centSIRT6: Bioinformatic Analysis of N308K and A313S Substitution Effects. Bioinform Biol Insights 2025; 19:11779322251339698. [PMID: 40416060 PMCID: PMC12099093 DOI: 10.1177/11779322251339698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/18/2025] [Indexed: 05/27/2025] Open
Abstract
Sirtuin 6 (SIRT6), a member of the class III histone deacetylase (HDAC) family, is crucial for the maintenance of general health and is associated with increased life expectancy and resistance to age-related diseases such as cancer and metabolic disorders. A comparative analysis of the SIRT6 gene in Ashkenazi Jewish (AJ) centenarians and noncentenarian controls found a distinct allele, centSIRT6, enriched in the centenarian group. This allele features 2 linked substitutions, N308K and A313S, and exhibits enhanced functions, including more efficient suppression of LINE1 retrotransposons, improved repair of DNA double-strand breaks, and increased efficiency in cancer cell killing. Notably, centSIRT6 shows lower deacetylase activity but higher mono-adenosine diphosphate (ADP) ribosyl transferase activity compared with the wild-type enzyme. This study used several bioinformatics tools to explore the structural changes caused by the N308K and A313S substitutions in centSIRT6 and to elucidate how these alterations contribute to changes in the enzymatic activities of SIRT6. The results indicate that these mutations reduce the structural flexibility of centSIRT6, thus weakening its interactions with acetyl-lysine but strengthening its interactions with ADP-ribose. This research provides useful information for future experimental studies to further investigate the molecular mechanisms of centSIRT6.
Collapse
Affiliation(s)
- Francisco Alejandro Lagunas-Rangel
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| |
Collapse
|
2
|
Manickam R, Santhana S, Xuan W, Bisht KS, Tipparaju SM. Nampt: a new therapeutic target for modulating NAD + levels in metabolic, cardiovascular, and neurodegenerative diseases. Can J Physiol Pharmacol 2025. [PMID: 40203459 DOI: 10.1139/cjpp-2024-0400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
NAD+ is an important cofactor involved in regulating many biochemical processes in cells. An imbalance in NAD+/NADH ratio is linked to many diseases. NAD+ is depleted in diabetes, cardiovascular and neurodegenerative diseases, and in aging, and is increased in tumor cells. NAD+ is generated in cells via the de novo, Preiss-Handler, and salvage pathways. Most of the cellular NAD+ is generated through Nampt activation, a key rate-limiting enzyme that is involved in the salvage pathway. Restoration of NAD+/NADH balance offers therapeutic advantages for improving tissue homeostasis and function. NAD+ is known to benefit and restore the body's physiological mechanisms, including DNA replication, chromatin and epigenetic modifications, and gene expression. Recent studies elucidate the role of NAD+ in cells utilizing transgenic mouse models. Translational new therapeutics are positioned to utilize the NAD+ restoration strategies for overcoming the drawbacks that exist in the pharmacological toolkit. The present review highlights the significance of Nampt-NAD+ axis as a major player in energy metabolism and provides an overview with insights into future strategies, providing pharmacological advantages to address current and future medical needs.
Collapse
Affiliation(s)
- Ravikumar Manickam
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Sandhya Santhana
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Wanling Xuan
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Kirpal S Bisht
- Department of Chemistry, College of Arts and Sciences, University of South Florida, Tampa, FL 33620, USA
| | - Srinivas M Tipparaju
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
3
|
Peng A, Li J, Xing J, Yao Y, Niu X, Zhang K. The function of nicotinamide phosphoribosyl transferase (NAMPT) and its role in diseases. Front Mol Biosci 2024; 11:1480617. [PMID: 39513038 PMCID: PMC11540786 DOI: 10.3389/fmolb.2024.1480617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/11/2024] [Indexed: 11/15/2024] Open
Abstract
Nicotinamide phosphoribosyl transferase (NAMPT) is a rate-limiting enzyme in the mammalian nicotinamide adenine dinucleotide (NAD) salvage pathway, and plays a vital role in the regulation of cell metabolic activity, reprogramming, aging and apoptosis. NAMPT synthesizes nicotinamide mononucleotide (NMN) through enzymatic action, which is a key protein involved in host defense mechanism and plays an important role in metabolic homeostasis and cell survival. NAMPT is involved in NAD metabolism and maintains intracellular NAD levels. Sirtuins (SIRTs) are a family of nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylases (HDACs), the members are capable of sensing cellular NAD+ levels. NAMPT-NAD and SIRT constitute a powerful anti-stress defense system. In this paper, the structure, biological function and correlation with diseases of NAMPT are introduced, aiming to provide new ideas for the targeted therapy of related diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Kaiming Zhang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Center Hospital, Taiyuan, China
| |
Collapse
|
4
|
Sorge M, Savoré G, Gallo A, Acquarone D, Sbroggiò M, Velasco S, Zamporlini F, Femminò S, Moiso E, Morciano G, Balmas E, Raimondi A, Nattenberg G, Stefania R, Tacchetti C, Rizzo AM, Corsetto P, Ghigo A, Turco E, Altruda F, Silengo L, Pinton P, Raffaelli N, Sniadecki NJ, Penna C, Pagliaro P, Hirsch E, Riganti C, Tarone G, Bertero A, Brancaccio M. An intrinsic mechanism of metabolic tuning promotes cardiac resilience to stress. EMBO Mol Med 2024; 16:2450-2484. [PMID: 39271959 PMCID: PMC11473679 DOI: 10.1038/s44321-024-00132-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024] Open
Abstract
Defining the molecular mechanisms underlying cardiac resilience is crucial to find effective approaches to protect the heart. A physiologic level of ROS is produced in the heart by fatty acid oxidation, but stressful events can boost ROS and cause mitochondrial dysfunction and cardiac functional impairment. Melusin is a muscle specific chaperone required for myocardial compensatory remodeling during stress. Here we report that Melusin localizes in mitochondria where it binds the mitochondrial trifunctional protein, a key enzyme in fatty acid oxidation, and decreases it activity. Studying both mice and human induced pluripotent stem cell-derived cardiomyocytes, we found that Melusin reduces lipid oxidation in the myocardium and limits ROS generation in steady state and during pressure overload and doxorubicin treatment, preventing mitochondrial dysfunction. Accordingly, the treatment with the lipid oxidation inhibitor Trimetazidine concomitantly with stressful stimuli limits ROS accumulation and prevents long-term heart dysfunction. These findings disclose a physiologic mechanism of metabolic regulation in the heart and demonstrate that a timely restriction of lipid metabolism represents a potential therapeutic strategy to improve cardiac resilience to stress.
Collapse
Affiliation(s)
- Matteo Sorge
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy.
| | - Giulia Savoré
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Andrea Gallo
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Davide Acquarone
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Mauro Sbroggiò
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Silvia Velasco
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Federica Zamporlini
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, 60121, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043, Italy
| | - Enrico Moiso
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Giampaolo Morciano
- Department of Medical Sciences, University of Ferrara, Ferrara, 44121, Italy
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, 48033, Italy
| | - Elisa Balmas
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Andrea Raimondi
- Experimental Imaging Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Gabrielle Nattenberg
- Departments of Mechanical Engineering, Bioengineering, and Laboratory Medicine and Pathology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
| | - Rachele Stefania
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Carlo Tacchetti
- Experimental Imaging Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Angela Maria Rizzo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano, 20133, Italy
| | - Paola Corsetto
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano, 20133, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Emilia Turco
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Fiorella Altruda
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Lorenzo Silengo
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara, 44121, Italy
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, 48033, Italy
| | - Nadia Raffaelli
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, 60121, Italy
| | - Nathan J Sniadecki
- Departments of Mechanical Engineering, Bioengineering, and Laboratory Medicine and Pathology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Chiara Riganti
- Department of Oncology, University of Turin, Torino, 10126, Italy
| | - Guido Tarone
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Alessandro Bertero
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Mara Brancaccio
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy.
| |
Collapse
|
5
|
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: 12] [Impact Index Per Article: 12.0] [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
|
6
|
Wu J, Han K, Sack MN. Targeting NAD+ Metabolism to Modulate Autoimmunity and Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1043-1050. [PMID: 38498807 PMCID: PMC10954088 DOI: 10.4049/jimmunol.2300693] [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: 10/18/2023] [Accepted: 12/18/2023] [Indexed: 03/20/2024]
Abstract
NAD+ biology is involved in controlling redox balance, functioning as a coenzyme in numerous enzymatic reactions, and is a cofactor for Sirtuin enzymes and a substrate for multiple regulatory enzyme reactions within and outside the cell. At the same time, NAD+ levels are diminished with aging and are consumed during the development of inflammatory and autoimmune diseases linked to aberrant immune activation. Direct NAD+ augmentation via the NAD+ salvage and Priess-Handler pathways is being investigated as a putative therapeutic intervention to improve the healthspan in inflammation-linked diseases. In this review, we survey NAD+ biology and its pivotal roles in the regulation of immunity and inflammation. Furthermore, we discuss emerging studies evaluate NAD+ boosting in murine models and in human diseases, and we highlight areas of research that remain unresolved in understanding the mechanisms of action of these nutritional supplementation strategies.
Collapse
Affiliation(s)
- Jing Wu
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Kim Han
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Michael N Sack
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
7
|
Turner L, Van Le TN, Cross E, Queriault C, Knight M, Trihemasava K, Davis J, Schaefer P, Nguyen J, Xu J, Goldspiel B, Hall E, Rome K, Scaglione M, Eggert J, Au-Yeung B, Wallace DC, Mesaros C, Baur JA, Bailis W. Single-cell NAD(H) levels predict clonal lymphocyte expansion dynamics. Sci Immunol 2024; 9:eadj7238. [PMID: 38489349 PMCID: PMC11064129 DOI: 10.1126/sciimmunol.adj7238] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 02/22/2024] [Indexed: 03/17/2024]
Abstract
Adaptive immunity requires the expansion of high-affinity lymphocytes from a heterogeneous pool. Whereas current models explain this through signal transduction, we hypothesized that antigen affinity tunes discrete metabolic pathways to license clonal lymphocyte dynamics. Here, we identify nicotinamide adenine dinucleotide (NAD) biosynthesis as a biochemical hub for the T cell receptor affinity-dependent metabolome. Through this central anabolic role, we found that NAD biosynthesis governs a quiescence exit checkpoint, thereby pacing proliferation. Normalizing cellular NAD(H) likewise normalizes proliferation across affinities, and enhancing NAD biosynthesis permits the expansion of lower affinity clones. Furthermore, single-cell differences in NAD(H) could predict division potential for both T and B cells, before the first division, unmixing proliferative heterogeneity. We believe that this supports a broader paradigm in which complex signaling networks converge on metabolic pathways to control single-cell behavior.
Collapse
Affiliation(s)
- Lucien Turner
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Tran Ngoc Van Le
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Eric Cross
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104
| | - Clemence Queriault
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Montana Knight
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Krittin Trihemasava
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - James Davis
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, 19104
| | - Patrick Schaefer
- Center for Mitochondrial and Epigenomic Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Janet Nguyen
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Jimmy Xu
- Center of Excellence in Environmental Toxicology & Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania; Philadelphia, PA 19104
| | - Brian Goldspiel
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Elise Hall
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Kelly Rome
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Michael Scaglione
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Joel Eggert
- Division of Immunology, Lowance Center for Human Immunology, Department of Medicine, Emory University; Atlanta, GA 30322
| | - Byron Au-Yeung
- Division of Immunology, Lowance Center for Human Immunology, Department of Medicine, Emory University; Atlanta, GA 30322
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
- Department of Pediatrics, Division of Human Genetics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104
| | - Clementina Mesaros
- Center of Excellence in Environmental Toxicology & Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania; Philadelphia, PA 19104
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, 19104
| | - Will Bailis
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104
| |
Collapse
|
8
|
Panova V, Richard AC. A metabolic pacer ensures smooth running of the lymphocyte activation race. Sci Immunol 2024; 9:eadn4958. [PMID: 38489351 DOI: 10.1126/sciimmunol.adn4958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 03/17/2024]
Abstract
Upon lymphocyte stimulation, accumulation of intracellular NAD(H) reflects the strength of antigen receptor signals and controls the rate of cell cycle entry and proliferation (see related Research Article by Turner et al.).
Collapse
Affiliation(s)
- Veera Panova
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Arianne C Richard
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| |
Collapse
|
9
|
Al-kuraishy HM, Jabir MS, Al-Gareeb AI, Saad HM, Batiha GES, Klionsky DJ. The beneficial role of autophagy in multiple sclerosis: Yes or No? Autophagy 2024; 20:259-274. [PMID: 37712858 PMCID: PMC10813579 DOI: 10.1080/15548627.2023.2259281] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/16/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic progressive demyelinating disease of the central nervous system (CNS) due to an increase of abnormal peripherally auto-reactive T lymphocytes which elicit autoimmunity. The main pathophysiology of MS is myelin sheath damage by immune cells and a defect in the generation of myelin by oligodendrocytes. Macroautophagy/autophagy is a critical degradation process that eliminates dysfunctional or superfluous cellular components. Autophagy has the property of a double-edged sword in MS in that it may have both beneficial and detrimental effects on MS neuropathology. Therefore, this review illustrates the protective and harmful effects of autophagy with regard to this disease. Autophagy prevents the progression of MS by reducing oxidative stress and inflammatory disorders. In contrast, over-activated autophagy is associated with the progression of MS neuropathology and in this case the use of autophagy inhibitors may alleviate the pathogenesis of MS. Furthermore, autophagy provokes the activation of different immune and supporting cells that play an intricate role in the pathogenesis of MS. Autophagy functions in the modulation of MS neuropathology by regulating cell proliferation related to demyelination and remyelination. Autophagy enhances remyelination by increasing the activity of oligodendrocytes, and astrocytes. However, autophagy induces demyelination by activating microglia and T cells. In conclusion, specific autophagic activators of oligodendrocytes, and astrocytes, and specific autophagic inhibitors of dendritic cells (DCs), microglia and T cells induce protective effects against the pathogenesis of MS.Abbreviations: ALS: amyotrophic lateral sclerosis; APCs: antigen-presenting cells; BBB: blood-brain barrier; CSF: cerebrospinal fluid; CNS: central nervous system; DCs: dendritic cells; EAE: experimental autoimmune encephalomyelitis; ER: endoplasmic reticulum; LAP: LC3-associated phagocytosis; MS: multiple sclerosis; NCA: non-canonical autophagy; OCBs: oligoclonal bands; PBMCs: peripheral blood mononuclear cells; PD: Parkinson disease; ROS: reactive oxygen species; UPR: unfolded protein response.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Iraq, Baghdad
| | - Majid S. Jabir
- Department of Applied Science, University of Technology, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Iraq, Baghdad
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, El Beheira, Egypt
| | | |
Collapse
|
10
|
Yong J, Cai S, Zeng Z. Targeting NAD + metabolism: dual roles in cancer treatment. Front Immunol 2023; 14:1269896. [PMID: 38116009 PMCID: PMC10728650 DOI: 10.3389/fimmu.2023.1269896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is indispensable for various oxidation-reduction reactions in mammalian cells, particularly during energy production. Malignant cells increase the expression levels of NAD+ biosynthesis enzymes for rapid proliferation and biomass production. Furthermore, mounting proof has indicated that NAD-degrading enzymes (NADases) play a role in creating the immunosuppressive tumor microenvironment (TME). Interestingly, both inhibiting NAD+ synthesis and targeting NADase have positive implications for cancer treatment. Here we summarize the detrimental outcomes of increased NAD+ production, the functions of NAD+ metabolic enzymes in creating an immunosuppressive TME, and discuss the progress and clinical translational potential of inhibitors for NAD+ synthesis and therapies targeting NADase.
Collapse
Affiliation(s)
- Jiaxin Yong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Songqing Cai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Zhaolei Zeng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
11
|
Santos L, Benitez-Rosendo A, Bresque M, Camacho-Pereira J, Calliari A, Escande C. Sirtuins: The NAD +-Dependent Multifaceted Modulators of Inflammation. Antioxid Redox Signal 2023; 39:1185-1208. [PMID: 37767625 DOI: 10.1089/ars.2023.0295] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Significance: Sirtuins are NAD+-dependent histone deacetylases regulating important processes in cellular biology such as inflammation, metabolism, oxidative stress, and apoptosis. Recent Advances: Despite initially being discovered to regulate transcription and life span via histone deacetylase activities, emerging data continually uncover new targets and propose additional roles. Due to the outstanding importance of the sirtuins in the control of the inflammatory response, their roles in the pathogenesis of several inflammatory-based diseases have become an area of intense research. Although sirtuins have been traditionally regarded as anti-inflammatory players, several recent findings suggest that their role in inflammation is complex and that in some cases sirtuins can indeed promote inflammation. Critical Issues: In this article, we provide an update on the latest findings concerning the new mechanisms of action and concepts about the role of sirtuins during inflammation. We focus on the impact that inflammatory-based processes exert on the liver, adipose tissue, and nervous system as well as on macrophage function and activation. Also, we discuss available data pointing to the dual role that, in particular contexts, sirtuins may have on inflammation control. Future Directions: Since the knowledge of sirtuin impact on metabolism is continually expanding, new venues of research arise. Besides become being regarded as candidates of therapeutic targets, posttranscriptional control of sirtuin expression by means of microRNAs challenges our traditional concepts of sirtuin regulation; importantly, the emerging role of NAD+ metabolism in aging and longevity has added a new dimension to the interest in sirtuin function. Antioxid. Redox Signal. 39, 1185-1208.
Collapse
Affiliation(s)
- Leonardo Santos
- Laboratory of Metabolic Diseases and Aging, INDICYO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
| | - Andrés Benitez-Rosendo
- Laboratory of Metabolic Diseases and Aging, INDICYO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
- Department of Biosciences, Facultad de Veterinaria, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Mariana Bresque
- Laboratory of Metabolic Diseases and Aging, INDICYO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Juliana Camacho-Pereira
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aldo Calliari
- Laboratory of Metabolic Diseases and Aging, INDICYO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
- Department of Biosciences, Facultad de Veterinaria, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Carlos Escande
- Laboratory of Metabolic Diseases and Aging, INDICYO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
| |
Collapse
|
12
|
Khalifa A, Guijarro A, Ravera S, Bertola N, Adorni MP, Papotti B, Raffaghello L, Benelli R, Becherini P, Namatalla A, Verzola D, Reverberi D, Monacelli F, Cea M, Pisciotta L, Bernini F, Caffa I, Nencioni A. Cyclic fasting bolsters cholesterol biosynthesis inhibitors' anticancer activity. Nat Commun 2023; 14:6951. [PMID: 37907500 PMCID: PMC10618279 DOI: 10.1038/s41467-023-42652-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 10/17/2023] [Indexed: 11/02/2023] Open
Abstract
Identifying oncological applications for drugs that are already approved for other medical indications is considered a possible solution for the increasing costs of cancer treatment. Under the hypothesis that nutritional stress through fasting might enhance the antitumour properties of at least some non-oncological agents, by screening drug libraries, we find that cholesterol biosynthesis inhibitors (CBIs), including simvastatin, have increased activity against cancers of different histology under fasting conditions. We show fasting's ability to increase CBIs' antitumour effects to depend on the reduction in circulating insulin, insulin-like growth factor-1 and leptin, which blunts the expression of enzymes from the cholesterol biosynthesis pathway and enhances cholesterol efflux from cancer cells. Ultimately, low cholesterol levels through combined fasting and CBIs reduce AKT and STAT3 activity, oxidative phosphorylation and energy stores in the tumour. Our results support further studies of CBIs in combination with fasting-based dietary regimens in cancer treatment and highlight the value of fasting for drug repurposing in oncology.
Collapse
Affiliation(s)
- Amr Khalifa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Ana Guijarro
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Silvia Ravera
- Department of Experimental Medicine, University of Genoa, Via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Nadia Bertola
- Department of Experimental Medicine, University of Genoa, Via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Maria Pia Adorni
- Department of Medicine and Surgery, University of Parma, 43125, Parma, Italy
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Lizzia Raffaghello
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Roberto Benelli
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Pamela Becherini
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
| | - Asmaa Namatalla
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
| | - Daniela Verzola
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
| | - Daniele Reverberi
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Michele Cea
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Livia Pisciotta
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Franco Bernini
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Irene Caffa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy.
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy.
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| |
Collapse
|
13
|
Fang J, Chen W, Hou P, Liu Z, Zuo M, Liu S, Feng C, Han Y, Li P, Shi Y, Shao C. NAD + metabolism-based immunoregulation and therapeutic potential. Cell Biosci 2023; 13:81. [PMID: 37165408 PMCID: PMC10171153 DOI: 10.1186/s13578-023-01031-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/14/2023] [Indexed: 05/12/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a critical metabolite that acts as a cofactor in energy metabolism, and serves as a cosubstrate for non-redox NAD+-dependent enzymes, including sirtuins, CD38 and poly(ADP-ribose) polymerases. NAD+ metabolism can regulate functionality attributes of innate and adaptive immune cells and contribute to inflammatory responses. Thus, the manipulation of NAD+ bioavailability can reshape the courses of immunological diseases. Here, we review the basics of NAD+ biochemistry and its roles in the immune response, and discuss current challenges and the future translational potential of NAD+ research in the development of therapeutics for inflammatory diseases, such as COVID-19.
Collapse
Affiliation(s)
- Jiankai Fang
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Wangwang Chen
- Laboratory Animal Center, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Pengbo Hou
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Zhanhong Liu
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Muqiu Zuo
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Shisong Liu
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Chao Feng
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Yuyi Han
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Peishan Li
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| | - Yufang Shi
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Changshun Shao
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
14
|
Carreira ASA, Ravera S, Zucal C, Thongon N, Irene C, Astigiano C, Bertola N, Buongiorno A, Roccuzzo M, Bisio A, Pardini B, Nencioni A, Bruzzone S, Provenzani A. Mitochondrial rewiring drives metabolic adaptation to NAD(H) shortage in triple negative breast cancer cells. Neoplasia 2023; 41:100903. [PMID: 37148658 DOI: 10.1016/j.neo.2023.100903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 05/08/2023]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is a key metabolic enzyme in NAD+ synthesis pathways and is found upregulated in several tumors, depicting NAD(H) lowering agents, like the NAMPT inhibitor FK866, as an appealing approach for anticancer therapy. Like other small molecules, FK866 triggers chemoresistance, observed in several cancer cellular models, which can prevent its clinical application. The molecular mechanisms sustaining the acquired of resistance to FK866 were studied in a model of triple negative breast cancer (MDA-MB-231 parental - PAR), exposed to increasing concentrations of the small molecule (MDA-MB-231 resistant - RES). RES cells are not sensitive to verapamil or cyclosporin A, excluding a potential role of increased efflux pumps activity as a mechanism of resistance. Similarly, the silencing of the enzyme Nicotinamide Riboside Kinase 1 (NMRK1) in RES cells does not increase FK866 toxicity, excluding this pathway as a compensatory mechanism of NAD+ production. Instead, Seahorse metabolic analysis revealed an increased mitochondrial spare respiratory capacity in RES cells. These cells presented a higher mitochondrial mass compared to the FK866-sensitive counterparts, as well as an increased consumption of pyruvate and succinate for energy production. Interestingly, co-treatment of PAR cells with FK866 and the mitochondrial pyruvate carrier (MPC) inhibitors UK5099 or rosiglitazone, as well as with the transient silencing of MPC2 but not of MPC1, induces a FK866-resistant phenotype. Taken together, these results unravel novel mechanisms of cell plasticity to counteract FK866 toxicity, that, besides the previously described LDHA dependency, rely on mitochondrial rewiring at functional and energetic levels.
Collapse
Affiliation(s)
| | - Silvia Ravera
- Department of Experimental Medicine, University of Genoa, Genoa, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy; Italian Institute for Genomic Medicine (IIGM), Candiolo, Italy.
| | - Chiara Zucal
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy.
| | - Natthakan Thongon
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy; Italian Institute for Genomic Medicine (IIGM), Candiolo, Italy.
| | - Caffa Irene
- Department of Internal Medicine, University of Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Cecilia Astigiano
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.
| | - Nadia Bertola
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.
| | - Arianna Buongiorno
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy.
| | - Michela Roccuzzo
- Advanced Imaging Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy.
| | - Alessandra Bisio
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy.
| | - Barbara Pardini
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy; Italian Institute for Genomic Medicine (IIGM), Candiolo, Italy.
| | - Alessio Nencioni
- Department of Internal Medicine, University of Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Santina Bruzzone
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.
| | - Alessandro Provenzani
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy.
| |
Collapse
|
15
|
Ghosh A, Khanam A, Ray K, Mathur P, Subramanian A, Poonia B, Kottilil S. CD38: an ecto-enzyme with functional diversity in T cells. Front Immunol 2023; 14:1146791. [PMID: 37180151 PMCID: PMC10172466 DOI: 10.3389/fimmu.2023.1146791] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
CD38, a nicotinamide adenine dinucleotide (NAD)+ glycohydrolase, is considered an activation marker of T lymphocytes in humans that is highly expressed during certain chronic viral infections. T cells constitute a heterogeneous population; however, the expression and function of CD38 has been poorly defined in distinct T cell compartments. We investigated the expression and function of CD38 in naïve and effector T cell subsets in the peripheral blood mononuclear cells (PBMCs) from healthy donors and people with HIV (PWH) using flow cytometry. Further, we examined the impact of CD38 expression on intracellular NAD+ levels, mitochondrial function, and intracellular cytokine production in response to virus-specific peptide stimulation (HIV Group specific antigen; Gag). Naïve T cells from healthy donors showed remarkably higher levels of CD38 expression than those of effector cells with concomitant reduced intracellular NAD+ levels, decreased mitochondrial membrane potential and lower metabolic activity. Blockade of CD38 by a small molecule inhibitor, 78c, increased metabolic function, mitochondrial mass and mitochondrial membrane potential in the naïve T lymphocytes. PWH exhibited similar frequencies of CD38+ cells in the T cell subsets. However, CD38 expression increased on Gag-specific IFN-γ and TNF-α producing cell compartments among effector T cells. 78c treatment resulted in reduced cytokine production, indicating its distinct expression and functional profile in different T cell subsets. In summary, in naïve cells high CD38 expression reflects lower metabolic activity, while in effector cells it preferentially contributes to immunopathogenesis by increasing inflammatory cytokine production. Thus, CD38 may be considered as a therapeutic target in chronic viral infections to reduce ongoing immune activation.
Collapse
Affiliation(s)
- Alip Ghosh
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Arshi Khanam
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Krishanu Ray
- Division of Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Poonam Mathur
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ananya Subramanian
- Department of Environmental Science, Policy, and Management, University of California, Berkeley, Berkeley, CA, United States
| | - Bhawna Poonia
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Shyam Kottilil
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
16
|
Watowich MB, Gilbert MR, Larion M. T cell exhaustion in malignant gliomas. Trends Cancer 2023; 9:270-292. [PMID: 36681605 PMCID: PMC10038906 DOI: 10.1016/j.trecan.2022.12.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/21/2023]
Abstract
Despite advances in understanding tumor biology, malignant gliomas remain incurable. While immunotherapy has improved outcomes in other cancer types, comparable efficacy has not yet been demonstrated for primary cancers of the central nervous system (CNS). T cell exhaustion, defined as a progressive decrease in effector function, sustained expression of inhibitory receptors, metabolic dysfunction, and distinct epigenetic and transcriptional alterations, contributes to the failure of immunotherapy in the CNS. Herein, we describe recent advances in understanding the drivers of T cell exhaustion in the glioma microenvironment. We discuss the extrinsic and intrinsic factors that contribute to exhaustion and highlight potential avenues for reversing this phenotype. Our ability to directly target specific immunosuppressive drivers in brain cancers would be a major advance in immunotherapy.
Collapse
Affiliation(s)
- Matthew B Watowich
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mioara Larion
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
17
|
Antonelli A, Scarpa ES, Bruzzone S, Astigiano C, Piacente F, Bruschi M, Fraternale A, Di Buduo CA, Balduini A, Magnani M. Anoxia Rapidly Induces Changes in Expression of a Large and Diverse Set of Genes in Endothelial Cells. Int J Mol Sci 2023; 24:ijms24065157. [PMID: 36982232 PMCID: PMC10049254 DOI: 10.3390/ijms24065157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Sinusoidal endothelial cells are the predominant vascular surface of the bone marrow and constitute the functional hematopoietic niche where hematopoietic stem and progenitor cells receive cues for self-renewal, survival, and differentiation. In the bone marrow hematopoietic niche, the oxygen tension is usually very low, and this condition affects stem and progenitor cell proliferation and differentiation and other important functions of this region. Here, we have investigated in vitro the response of endothelial cells to a marked decrease in O2 partial pressure to understand how the basal gene expression of some relevant biological factors (i.e., chemokines and interleukins) that are fundamental for the intercellular communication could change in anoxic conditions. Interestingly, mRNA levels of CXCL3, CXCL5, and IL-34 genes are upregulated after anoxia exposure but become downmodulated by sirtuin 6 (SIRT6) overexpression. Indeed, the expression levels of some other genes (such as Leukemia Inhibitory Factor (LIF)) that were not significantly affected by 8 h anoxia exposure become upregulated in the presence of SIRT6. Therefore, SIRT6 mediates also the endothelial cellular response through the modulation of selected genes in an extreme hypoxic condition.
Collapse
Affiliation(s)
- Antonella Antonelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | | | - Santina Bruzzone
- Department of Experimental Medicine, Section of Biochemistry, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Cecilia Astigiano
- Department of Experimental Medicine, Section of Biochemistry, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Francesco Piacente
- Department of Experimental Medicine, Section of Biochemistry, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Michela Bruschi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Alessandra Fraternale
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | | | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- Department of Biomedical Engineering, Tufts University in Boston, Boston, MA 02111, USA
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
- Correspondence:
| |
Collapse
|
18
|
Chen ZA, Ma HH, Wang Y, Tian H, Mi JW, Yao DM, Yang CJ. Integrated multiple microarray studies by robust rank aggregation to identify immune-associated biomarkers in Crohn's disease based on three machine learning methods. Sci Rep 2023; 13:2694. [PMID: 36792688 PMCID: PMC9931764 DOI: 10.1038/s41598-022-26345-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/13/2022] [Indexed: 02/17/2023] Open
Abstract
Crohn's disease (CD) is a complex autoimmune disorder presumed to be driven by complex interactions of genetic, immune, microbial and even environmental factors. Intrinsic molecular mechanisms in CD, however, remain poorly understood. The identification of novel biomarkers in CD cases based on larger samples through machine learning approaches may inform the diagnosis and treatment of diseases. A comprehensive analysis was conducted on all CD datasets of Gene Expression Omnibus (GEO); our team then used the robust rank aggregation (RRA) method to identify differentially expressed genes (DEGs) between controls and CD patients. PPI (protein‒protein interaction) network and functional enrichment analyses were performed to investigate the potential functions of the DEGs, with molecular complex detection (MCODE) identifying some important functional modules from the PPI network. Three machine learning algorithms, support vector machine-recursive feature elimination (SVM-RFE), random forest (RF), and least absolute shrinkage and selection operator (LASSO), were applied to determine characteristic genes, which were verified by ROC curve analysis and immunohistochemistry (IHC) using clinical samples. Univariable and multivariable logistic regression were used to establish a machine learning score for diagnosis. Single-sample GSEA (ssGSEA) was performed to examine the correlation between immune infiltration and biomarkers. In total, 5 datasets met the inclusion criteria: GSE75214, GSE95095, GSE126124, GSE179285, and GSE186582. Based on RRA integrated analysis, 203 significant DEGs were identified (120 upregulated genes and 83 downregulated genes), and MCODE revealed some important functional modules in the PPI network. Machine learning identified LCN2, REG1A, AQP9, CCL2, GIP, PROK2, DEFA5, CXCL9, and NAMPT; AQP9, PROK2, LCN2, and NAMPT were further verified by ROC curves and IHC in the external cohort. The final machine learning score was defined as [Expression level of AQP9 × (2.644)] + [Expression level of LCN2 × (0.958)] + [Expression level of NAMPT × (1.115)]. ssGSEA showed markedly elevated levels of dendritic cells and innate immune cells, such as macrophages and NK cells, in CD, consistent with the gene enrichment results that the DEGs are mainly involved in the IL-17 signaling pathway and humoral immune response. The selected biomarkers analyzed by the RRA method and machine learning are highly reliable. These findings improve our understanding of the molecular mechanisms of CD pathogenesis.
Collapse
Affiliation(s)
- Zi-An Chen
- grid.452702.60000 0004 1804 3009Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei China ,Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Disease, Shijiazhuang, 050000 Hebei China
| | - Hui-hui Ma
- grid.452702.60000 0004 1804 3009Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei China ,Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Disease, Shijiazhuang, 050000 Hebei China
| | - Yan Wang
- grid.452702.60000 0004 1804 3009Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei China ,Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Disease, Shijiazhuang, 050000 Hebei China
| | - Hui Tian
- grid.452702.60000 0004 1804 3009Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei China ,Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Disease, Shijiazhuang, 050000 Hebei China
| | - Jian-wei Mi
- grid.452702.60000 0004 1804 3009Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei China ,Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Disease, Shijiazhuang, 050000 Hebei China
| | - Dong-Mei Yao
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China. .,Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Disease, Shijiazhuang, 050000, Hebei, China.
| | - Chuan-Jie Yang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China. .,Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Disease, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
19
|
CD38-Induced Metabolic Dysfunction Primes Multiple Myeloma Cells for NAD +-Lowering Agents. Antioxidants (Basel) 2023; 12:antiox12020494. [PMID: 36830052 PMCID: PMC9952390 DOI: 10.3390/antiox12020494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/18/2023] Open
Abstract
Cancer cells fuel growth and energy demands by increasing their NAD+ biosynthesis dependency, which therefore represents an exploitable vulnerability for anti-cancer strategies. CD38 is a NAD+-degrading enzyme that has become crucial for anti-MM therapies since anti-CD38 monoclonal antibodies represent the backbone for treatment of newly diagnosed and relapsed multiple myeloma patients. Nevertheless, further steps are needed to enable a full exploitation of these strategies, including deeper insights of the mechanisms by which CD38 promotes tumorigenesis and its metabolic additions that could be selectively targeted by therapeutic strategies. Here, we present evidence that CD38 upregulation produces a pervasive intracellular-NAD+ depletion, which impairs mitochondrial fitness and enhances oxidative stress; as result, genetic or pharmacologic approaches that aim to modify CD38 surface-level prime MM cells to NAD+-lowering agents. The molecular mechanism underlying this event is an alteration in mitochondrial dynamics, which decreases mitochondria efficiency and triggers energetic remodeling. Overall, we found that CD38 handling represents an innovative strategy to improve the outcomes of NAD+-lowering agents and provides the rationale for testing these very promising agents in clinical studies involving MM patients.
Collapse
|
20
|
Holay N, Kennedy BE, Murphy JP, Konda P, Giacomantonio M, Brauer-Chapin T, Paulo JA, Kumar V, Kim Y, Elaghil M, Sisson G, Clements D, Richardson C, Gygi SP, Gujar S. After virus exposure, early bystander naïve CD8 T cell activation relies on NAD + salvage metabolism. Front Immunol 2023; 13:1047661. [PMID: 36818473 PMCID: PMC9932030 DOI: 10.3389/fimmu.2022.1047661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/20/2022] [Indexed: 02/04/2023] Open
Abstract
CD8 T cells play a central role in antiviral immunity. Type I interferons are among the earliest responders after virus exposure and can cause extensive reprogramming and antigen-independent bystander activation of CD8 T cells. Although bystander activation of pre-existing memory CD8 T cells is known to play an important role in host defense and immunopathology, its impact on naïve CD8 T cells remains underappreciated. Here we report that exposure to reovirus, both in vitro or in vivo, promotes bystander activation of naïve CD8 T cells within 24 hours and that this distinct subtype of CD8 T cell displays an innate, antiviral, type I interferon sensitized signature. The induction of bystander naïve CD8 T cells is STAT1 dependent and regulated through nicotinamide phosphoribosyl transferase (NAMPT)-mediated enzymatic actions within NAD+ salvage metabolic biosynthesis. These findings identify a novel aspect of CD8 T cell activation following virus infection with implications for human health and physiology.
Collapse
Affiliation(s)
- Namit Holay
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Barry E. Kennedy
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- IMV Inc, Halifax, NS, Canada
| | - J. Patrick Murphy
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Biology, University of Prince Edward Island, Charlottetown, PEI, Canada
| | - Prathyusha Konda
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | | | - Tatjana Brauer-Chapin
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Cell Biology, Harvard Medical School, Harvard University, Boston, MA, United States
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Harvard University, Boston, MA, United States
| | | | - Youra Kim
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Mariam Elaghil
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- IMV Inc, Halifax, NS, Canada
| | - Gary Sisson
- Department of Biology, University of Prince Edward Island, Charlottetown, PEI, Canada
| | - Derek Clements
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Christopher Richardson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Canadian Centre for Vaccinology, IWK Health Centre, Goldbloom Pavilion, Halifax, NS, Canada
- Department of Pediatrics, Dalhousie University, Halifax, NS, Canada
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Harvard University, Boston, MA, United States
| | - Shashi Gujar
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Biology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
- Cancer Immunotherapy: Innovation & Global Partnerships, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
21
|
Correale J, Marrodan M. Multiple sclerosis and obesity: The role of adipokines. Front Immunol 2022; 13:1038393. [PMID: 36457996 PMCID: PMC9705772 DOI: 10.3389/fimmu.2022.1038393] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/28/2022] [Indexed: 11/24/2023] Open
Abstract
Multiple Sclerosis (MS), a chronic inflammatory disease of the central nervous system that leads to demyelination and neurodegeneration has been associated with various environmental and lifestyle factors. Population-based studies have provided evidence showing the prevalence of MS is increasing worldwide. Because a similar trend has been observed for obesity and metabolic syndrome, interest has grown in possible underlying biological mechanisms shared by both conditions. Adipokines, a family of soluble factors produced by adipose tissue that participate in a wide range of biological functions, contribute to a low state of chronic inflammation observed in obesity, and influence immune function, metabolism, and nutritional state. In this review, we aim to describe epidemiological and biological factors common to MS and obesity, as well as provide an update on current knowledge of how different pro- and anti-inflammatory adipokines participate as immune response mediators in MS, as well as in the animal model for MS, namely, experimental autoimmune encephalomyelitis (EAE). Multiple Sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) leading to demyelination, and neurodegeneration. Although its pathogenesis is not yet fully understood, there is considerable evidence to suggest MS arises from complex interactions between individual genetic susceptibility and external environmental factors. In recent decades, population-based studies have provided evidence indicating the prevalence of MS is increasing worldwide, in parallel with the rise in obesity and metabolic syndrome. This synchronous increment in the incidence of both MS and obesity has led to a search for potential biological mechanisms linking both conditions. Notably, a large number of studies have established significant correlation between obesity and higher prevalence, or worse prognosis, of several immune-mediated conditions. Fat tissue has been found to produce a variety of soluble factors named adipokines. These mediators, secreted by both adipocytes as well as diverse immune cells, participate in a wide range of biological functions, further strengthening the concept of a link between immune function, metabolism, and nutritional state. Because obesity causes overproduction of pro-inflammatory adipokines (namely leptin, resistin and visfatin) and reduction of anti-inflammatory adipokines (adiponectin and apelin), adipose tissue dysregulation would appear to contribute to a state of chronic, low-grade inflammation favoring the development of disease. In this review, we present a summary of current knowledge related to the pathological effects of different adipokines, prevalent in obese MS patients.
Collapse
Affiliation(s)
- Jorge Correale
- Departamento de Neurología, Fleni, Buenos Aires, Argentina
- Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Universidad de Buenos Aires/Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | |
Collapse
|
22
|
Lin C, Xu JQ, Zhong GC, Chen H, Xue HM, Yang M, Chen C. Integrating RNA-seq and scRNA-seq to explore the biological significance of NAD + metabolism-related genes in the initial diagnosis and relapse of childhood B-cell acute lymphoblastic leukemia. Front Immunol 2022; 13:1043111. [PMID: 36439178 PMCID: PMC9691973 DOI: 10.3389/fimmu.2022.1043111] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/27/2022] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Nicotinamide Adenine Dinucleotide (NAD) depletion is reported to be a potential treatment for B-cell Acute Lymphoblastic Leukemia (B-ALL), but the mechanism of NAD metabolism-related genes (NMRGs) in B-ALL relapse remains unclear. METHODS Transcriptome data (GSE3912), and single-cell sequencing data (GSE130116) of B-ALL patients were downloaded from Gene Expression Omnibus (GEO) database. NMRGs were sourced from Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome databases. Further, the differentially expressed NMRGs (DE-NMRGs) were selected from the analysis between initial diagnosis and relapse B-ALL samples, which further performed functional enrichment analyses. The biomarkers were obtained through random forest (RF) algorithm and repeated cross validation. Additionally, cell type identification by estimating relative subsets of RNA transcripts (CIBERSORT) algorithm was used to evaluate the immune cell differences between the initial diagnosis and relapse samples, and the correlations between biomarkers and gene markers of differential immune cells were analyzed. Furthermore, single cell RNA sequencing was conducted in the GSE130116 dataset to find key cell clusters. In addition, according to biomarkers expressions, cell clusters were categorized into high and low biomarker expression groups, and Gene Set Enrichment Analysis (GSEA) analysis was performed on them. Finally, the cell clusters with the highest expression of biomarkers were selected to explore the roles of biomarkers in different cell clusters and identify transcription factors (TFs) influencing biological markers. RESULTS 23 DE-NMRGs were screened out, which were mainly enriched in nucleoside phosphate metabolic process, nucleotide metabolic process, and Nicotinate and nicotinamide metabolism. Moreover, 3 biomarkers (NADSYN1, SIRT3, and PARP6) were identified from the machine learning. CIBERSORT results demonstrated that four types of immune cells (B Cells naive, Monocyte, Neutrophils, and T cells CD4 memory Activated) were significantly different between the initial diagnosis and the relapse B-ALL samples, and there were strong correlations between biomarkers and differential immune cells such as positive correlation between NADSYN1 and B Cells naive. The single cell analyses showed that the biomarkers were highly expressed in common myeloid progenitors (CMP), granulocyte-macrophage progenitor (GMP), and megakaryocyte-erythroid progenitor (MEP) cell clusters. Gene set enrichment analysis (GSEA) results indicated that 55 GO terms and 3 KEGG pathways were enriched by the genes in high and low biomarker expression groups. It was found that TF CREB3L2(+) was significantly reduced in the high expression group, which may be the TF affecting biomarkers in the high expression group. CONCLUSION This study identified NADSYN1, SIRT3, and PARP6 as the biomarkers of B-ALL, explored biological significance of NMRGs in the initial diagnosis and relapse of B-ALL, and revealed mechanism of biomarkers at the level of the single cell.
Collapse
Affiliation(s)
- Chao Lin
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Jia-Qi Xu
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Gui-Chao Zhong
- Department of Pediatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Hui Chen
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Hong-Man Xue
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Mo Yang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Chun Chen
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
23
|
Visfatin Amplifies Cardiac Inflammation and Aggravates Cardiac Injury via the NF-κB p65 Signaling Pathway in LPS-Treated Mice. Mediators Inflamm 2022; 2022:3306559. [PMID: 36262545 PMCID: PMC9576419 DOI: 10.1155/2022/3306559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/09/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
Background Visfatin is an adipocytokine that has been demonstrated to be involved in cardiovascular diseases. This study aims at determining the role of visfatin in sepsis-induced cardiac injury and identify its possible mechanisms. Methods Dynamic changes in visfatin expression in mice with lipopolysaccharide- (LPS-) induced septicemia were measured. Additionally, mice were pretreated with visfatin and further administered LPS to observe the effects of visfatin on cardiac injury. Finally, septic mice were also pretreated with JSH-23 to investigate whether visfatin regulates cardiac injury via the NF-κB p65 pathway. Results Visfatin expression levels in both the heart and serum were increased in LPS-treated mice and peaked at 6 hours, and visfatin was derived from cardiac macrophages. In septic mice, pretreatment with visfatin reduced the survival rate, worsened cardiac dysfunction, and increased the expression of cardiac injury markers, including creatine kinase myocardial bound (CK-MB) and lactate dehydrogenase (LDH). Treatment with visfatin also increased the infiltration of CD3+ cells and F4/80+ cells, amplified the cardiac inflammatory response, and elevated myocardial cell apoptosis. Treatment with JSH-23 reversed the effects of visfatin in septic mice. Conclusions This study showed that visfatin amplifies the cardiac inflammatory response and aggravates cardiac injury through the p65 signaling pathway. Visfatin may be a clinical target for preventing cardiac injury in sepsis.
Collapse
|
24
|
Spinelli S, Guida L, Vigliarolo T, Passalacqua M, Begani G, Magnone M, Sturla L, Benzi A, Ameri P, Lazzarini E, Bearzi C, Rizzi R, Zocchi E. The ABA-LANCL1/2 Hormone-Receptors System Protects H9c2 Cardiomyocytes from Hypoxia-Induced Mitochondrial Injury via an AMPK- and NO-Mediated Mechanism. Cells 2022; 11:cells11182888. [PMID: 36139463 PMCID: PMC9496903 DOI: 10.3390/cells11182888] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Abscisic acid (ABA) regulates plant responses to stress, partly via NO. In mammals, ABA stimulates NO production by innate immune cells and keratinocytes, glucose uptake and mitochondrial respiration by skeletal myocytes and improves blood glucose homeostasis through its receptors LANCL1 and LANCL2. We hypothesized a role for the ABA-LANCL1/2 system in cardiomyocyte protection from hypoxia via NO. The effect of ABA and of the silencing or overexpression of LANCL1 and LANCL2 were investigated in H9c2 rat cardiomyoblasts under normoxia or hypoxia/reoxygenation. In H9c2, hypoxia induced ABA release, and ABA stimulated NO production. ABA increased the survival of H9c2 to hypoxia, and L-NAME, an inhibitor of NO synthase (NOS), abrogated this effect. ABA also increased glucose uptake and NADPH levels and increased phosphorylation of Akt, AMPK and eNOS. Overexpression or silencing of LANCL1/2 significantly increased or decreased, respectively, transcription, expression and phosphorylation of AMPK, Akt and eNOS; transcription of NAMPT, Sirt1 and the arginine transporter. The mitochondrial proton gradient and cell vitality increased in LANCL1/2-overexpressing vs. -silenced cells after hypoxia/reoxygenation, and L-NAME abrogated this difference. These results implicate the ABA-LANCL1/2 hormone-receptor system in NO-mediated cardiomyocyte protection against hypoxia.
Collapse
Affiliation(s)
- Sonia Spinelli
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Lucrezia Guida
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Tiziana Vigliarolo
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Mario Passalacqua
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Giulia Begani
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Mirko Magnone
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Laura Sturla
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Andrea Benzi
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Pietro Ameri
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Viale Benedetto XV 6, 16132 Genova, Italy
| | - Edoardo Lazzarini
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Tesserete 48, 6500 Bellinzona, Switzerland
| | - Claudia Bearzi
- Institute of Biomedical Technologies, National Research Council of Italy (ITB-CNR), Via Fratelli Cervi 93, 20054 Milan, Italy
- Fondazione Istituto Nazionale di Genetica Molecolare, Via F. Sforza 35, 20122 Milan, Italy
| | - Roberto Rizzi
- Fondazione Istituto Nazionale di Genetica Molecolare, Via F. Sforza 35, 20122 Milan, Italy
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, C.so della Repubblica 79, 04100 Latina, Italy
| | - Elena Zocchi
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
- Correspondence:
| |
Collapse
|
25
|
Bai JF, Majjigapu SR, Sordat B, Poty S, Vogel P, Elías-Rodríguez P, Moreno-Vargas AJ, Carmona AT, Caffa I, Ghanem M, Khalifa A, Monacelli F, Cea M, Robina I, Gajate C, Mollinedo F, Bellotti A, Nahimana A, Duchosal M, Nencioni A. Identification of new FK866 analogues with potent anticancer activity against pancreatic cancer. Eur J Med Chem 2022; 239:114504. [PMID: 35724566 DOI: 10.1016/j.ejmech.2022.114504] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 11/22/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal diseases for which chemotherapy has not been very successful yet. FK866 ((E)-N-(4-(1-benzoylpiperidin-4-yl)butyl)-3-(pyridin-3-yl)acrylamide) is a well-known NAMPT (nicotinamide phosphoribosyltransferase) inhibitor with anti-cancer activities, but it failed in phase II clinical trials. We found that FK866 shows anti-proliferative activity in three PDAC cell lines, as well as in Jurkat T-cell leukemia cells. More than 50 FK866 analogues were synthesized that introduce substituents on the phenyl ring of the piperidine benzamide group of FK866 and exchange its buta-1,4-diyl tether for 1-oxyprop-3-yl, (E)-but-2-en-1,4-diyl and 2- and 3-carbon tethers. The pyridin-3-yl moiety of FK866 was exchanged for chlorinated and fluorinated analogues and for pyrazin-2-yl and pyridazin-4-yl groups. Several compounds showed low nanomolar or sub-nanomolar cell growth inhibitory activity. Our best cell anti-proliferative compounds were the 2,4,6-trimethoxybenzamide analogue of FK866 ((E)-N-(4-(1-(2,4,6-trimethoxybenzoyl)piperidin-4-yl)butyl)-3-(pyridin-3-yl)acrylamide) (9), the 2,6-dimethoxybenzamide (8) and 2-methoxybenzamide (4), which exhibited an IC50 of 0.16 nM, 0.004 nM and 0.08 nM toward PDAC cells, respectively.
Collapse
Affiliation(s)
- Jian-Fei Bai
- Laboratory of Glycochemistry and Asymmetric Synthesis, Swiss Institute of Technology (EPFL), 1015, Lausanne, Switzerland
| | - Somi Reddy Majjigapu
- Laboratory of Glycochemistry and Asymmetric Synthesis, Swiss Institute of Technology (EPFL), 1015, Lausanne, Switzerland
| | - Bernard Sordat
- Laboratory of Glycochemistry and Asymmetric Synthesis, Swiss Institute of Technology (EPFL), 1015, Lausanne, Switzerland
| | - Sophie Poty
- Laboratory of Glycochemistry and Asymmetric Synthesis, Swiss Institute of Technology (EPFL), 1015, Lausanne, Switzerland
| | - Pierre Vogel
- Laboratory of Glycochemistry and Asymmetric Synthesis, Swiss Institute of Technology (EPFL), 1015, Lausanne, Switzerland
| | - Pilar Elías-Rodríguez
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, 41012, Spain
| | - Antonio J Moreno-Vargas
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, 41012, Spain
| | - Ana T Carmona
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, 41012, Spain
| | - Irene Caffa
- Department of Internal Medicine and Medical Specialties, University of Genoa, 16132, Genoa, Italy
| | - Moustafa Ghanem
- Department of Internal Medicine and Medical Specialties, University of Genoa, 16132, Genoa, Italy
| | - Amr Khalifa
- Department of Internal Medicine and Medical Specialties, University of Genoa, 16132, Genoa, Italy; Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties, University of Genoa, 16132, Genoa, Italy; Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Michele Cea
- Department of Internal Medicine and Medical Specialties, University of Genoa, 16132, Genoa, Italy; Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Inmaculada Robina
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, 41012, Spain
| | - Consuelo Gajate
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Faustino Mollinedo
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Axel Bellotti
- Central Laboratory of Hematology, Medical Laboratory and Pathology Department, Lausanne University Hospital, 1011, Lausanne, Switzerland
| | - Aimable Nahimana
- Central Laboratory of Hematology, Medical Laboratory and Pathology Department, Lausanne University Hospital, 1011, Lausanne, Switzerland
| | - Michel Duchosal
- Central Laboratory of Hematology, Medical Laboratory and Pathology Department, Lausanne University Hospital, 1011, Lausanne, Switzerland; Service of Hematology, Oncology Department, Lausanne University Hospital, 1011, Lausanne, Switzerland
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, 16132, Genoa, Italy; Ospedale Policlinico San Martino IRCCS, Genoa, Italy.
| |
Collapse
|
26
|
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: 12] [Impact Index Per Article: 4.0] [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
|
27
|
Ruszkiewicz JA, Bürkle A, Mangerich A. Fueling genome maintenance: On the versatile roles of NAD + in preserving DNA integrity. J Biol Chem 2022; 298:102037. [PMID: 35595095 PMCID: PMC9194868 DOI: 10.1016/j.jbc.2022.102037] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
NAD+ is a versatile biomolecule acting as a master regulator and substrate in various cellular processes, including redox regulation, metabolism, and various signaling pathways. In this article, we concisely and critically review the role of NAD+ in mechanisms promoting genome maintenance. Numerous NAD+-dependent reactions are involved in the preservation of genome stability, the cellular DNA damage response, and other pathways regulating nucleic acid metabolism, such as gene expression and cell proliferation pathways. Of note, NAD+ serves as a substrate to ADP-ribosyltransferases, sirtuins, and potentially also eukaryotic DNA ligases, all of which regulate various aspects of DNA integrity, damage repair, and gene expression. Finally, we critically analyze recent developments in the field as well as discuss challenges associated with therapeutic actions intended to raise NAD+ levels.
Collapse
Affiliation(s)
- Joanna A Ruszkiewicz
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany.
| | - Alexander Bürkle
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany.
| | - Aswin Mangerich
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
28
|
Piacente F, Bottero M, Benzi A, Vigo T, Uccelli A, Bruzzone S, Ferrara G. Neuroprotective Potential of Dendritic Cells and Sirtuins in Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23084352. [PMID: 35457169 PMCID: PMC9025744 DOI: 10.3390/ijms23084352] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 12/04/2022] Open
Abstract
Myeloid cells, including parenchymal microglia, perivascular and meningeal macrophages, and dendritic cells (DCs), are present in the central nervous system (CNS) and establish an intricate relationship with other cells, playing a crucial role both in health and in neurological diseases. In this context, DCs are critical to orchestrating the immune response linking the innate and adaptive immune systems. Under steady-state conditions, DCs patrol the CNS, sampling their local environment and acting as sentinels. During neuroinflammation, the resulting activation of DCs is a critical step that drives the inflammatory response or the resolution of inflammation with the participation of different cell types of the immune system (macrophages, mast cells, T and B lymphocytes), resident cells of the CNS and soluble factors. Although the importance of DCs is clearly recognized, their exact function in CNS disease is still debated. In this review, we will discuss modern concepts of DC biology in steady-state and during autoimmune neuroinflammation. Here, we will also address some key aspects involving DCs in CNS patrolling, highlighting the neuroprotective nature of DCs and emphasizing their therapeutic potential for the treatment of neurological conditions. Recently, inhibition of the NAD+-dependent deac(et)ylase sirtuin 6 was demonstrated to delay the onset of experimental autoimmune encephalomyelitis, by dampening DC trafficking towards inflamed LNs. Thus, a special focus will be dedicated to sirtuins’ role in DCs functions.
Collapse
Affiliation(s)
- Francesco Piacente
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
| | - Marta Bottero
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Andrea Benzi
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
| | - Tiziana Vigo
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Antonio Uccelli
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Santina Bruzzone
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
- Correspondence: ; Tel.: +39-(0)10-353-8150
| | - Giovanni Ferrara
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| |
Collapse
|
29
|
Sirtuins are crucial regulators of T cell metabolism and functions. Exp Mol Med 2022; 54:207-215. [PMID: 35296782 PMCID: PMC8979958 DOI: 10.1038/s12276-022-00739-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/23/2021] [Indexed: 01/01/2023] Open
Abstract
It is well known that metabolism underlies T cell differentiation and functions. The pathways regulating T cell metabolism and function are interconnected, and changes in T cell metabolic activity directly impact the effector functions and fate of T cells. Thus, understanding how metabolic pathways influence immune responses and ultimately affect disease progression is paramount. Epigenetic and posttranslational modification mechanisms have been found to control immune responses and metabolic reprogramming. Sirtuins are NAD+-dependent histone deacetylases that play key roles during cellular responses to a variety of stresses and have recently been reported to have potential roles in immune responses. Therefore, sirtuins are of significant interest as therapeutic targets to treat immune-related diseases and enhance antitumor immunity. This review aims to illustrate the potential roles of sirtuins in different subtypes of T cells during the adaptive immune response. Sirtuins, enzymes that regulate how cells respond to stress, regulate T cell metabolism and functions, and therefore blocking or boosting sirtuins influences immune responses. As part of the immune system, some types of T cells attack specific targets; others keep the immune response in check. Imene Hamaidi and Sungjune Kim at H. Lee Moffitt Cancer Center, Tampa, USA, have reviewed how sirtuins affect different subsets of T cells to either promote or suppress immune responses. Boosting sirtuins that increase the function of inflammation-suppressing T cells can improve outcomes for transplant recipients or help treat autoimmune diseases. Conversely, stimulating immune-activating sirtuins can help re-energize exhausted antitumor T cells. Understanding the complex web of sirtuin–T cell interactions may help in developing therapeutic strategies for improving transplant outcomes, and for treating autoimmune diseases and cancer.
Collapse
|
30
|
Shen D, Liu K, Wang H, Wang H. Autophagy modulation in multiple sclerosis and experimental autoimmune encephalomyelitis. Clin Exp Immunol 2022; 209:140-150. [PMID: 35641229 PMCID: PMC9390842 DOI: 10.1093/cei/uxac017] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/01/2022] [Accepted: 02/14/2022] [Indexed: 11/14/2022] Open
Abstract
Multiple sclerosis (MS), a white matter demyelinating disease of the central nervous system (CNS), is characterized by neuroinflammatory and neurodegenerative. Experimental autoimmune encephalomyelitis (EAE) is a commonly used animal model for investigating pathogenic mechanisms of MS, representing the destruction of the blood-brain barrier (BBB), the activation of T cells, and the infiltration of myeloid cells. An increasing number of studies have documented that autophagy plays a critical role in the pathogenesis of both MS and EAE. Autophagy maintains CNS homeostasis by degrading the damaged organelles and abnormal proteins. Furthermore, autophagy is involved in inflammatory responses by regulating the activation of immune cells and the secretion of inflammatory factors. However, the specific mechanisms of autophagy involved in MS and EAE are not completely understood. In this review, we will summarize the complex mechanisms of autophagy in MS and EAE, providing potential therapeutic approaches for the management of MS.
Collapse
Affiliation(s)
- Donghui Shen
- Department of Neurology, Qingdao Municipal Hospital, Qingdao 266000, Shan Dong Province, China
| | - Kang Liu
- Department of Stomatology, Qingdao Municipal Hospital, Qingdao 266000, Shan Dong Province, China
| | - Hongyan Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao 266000, Shan Dong Province, China
| | - Haifeng Wang
- Correspondence: Haifeng Wang, Department of Neurology, Qingdao Municipal Hospital, Qingdao, Shan Dong Province, China.
| |
Collapse
|
31
|
Adipokines as Immune Cell Modulators in Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms221910845. [PMID: 34639186 PMCID: PMC8509121 DOI: 10.3390/ijms221910845] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS), a chronic inflammatory and demyelinating disease of the central nervous system (CNS), is a major clinical and societal problem, which has a tremendous impact on the life of patients and their proxies. Current immunomodulatory and anti-inflammatory therapies prove to be relatively effective; however, they fail to concomitantly stop ongoing neurological deterioration and do not reverse acquired disability. The proportion to which genetic and environmental factors contribute to the etiology of MS is still incompletely understood; however, a recent association between MS etiology and obesity was shown, with obesity greatly increasing the risk of developing MS. An altered balance of adipokines, which are white adipose tissue (WAT) hormones, plays an important role in the low-grade chronic inflammation during obesity by their pervasive modification of local and systemic inflammation. Vice versa, inflammatory factors secreted by immune cells affect adipokine function. To explore the role of adipokines in MS pathology, we will here review the reciprocal effects of adipokines and immune cells and summarize alterations in adipokine levels in MS patient cohorts. Finally, we will discuss proof-of-concept studies demonstrating the therapeutic potential of adipokines to target both neuroinflammation and neurodegeneration processes in MS.
Collapse
|
32
|
Qu SD, Liu GX. Daporinad in vitro metabolite profiling via rat, dog, monkey and human liver microsomes by liquid chromatography/quadrupole-orbitrap mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2021; 35:e9150. [PMID: 34159659 DOI: 10.1002/rcm.9150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 06/13/2023]
Abstract
RATIONALE Daporinad is a novel and potent inhibitor of nicotinamide phosphoribosyl transferase with potential antineoplastic and antiangiogenic activities. We aimed to explore the metabolites of daporinad generated from liver microsomes and to propose metabolic pathways. METHODS The metabolites were generated by individually incubating daporinad (10 μM) with liver microsomes at 37°C for 60 min. The metabolites were identified by ultra-high-performance liquid chromatography/quadrupole-orbitrap mass spectrometry (UPLC/Q-Orbitrap-MS) using electrospray ionization in positive ion mode. They were deduced by accurate MS and MS/MS data. RESULTS In total, 16 metabolites were found and their identities were characterized. In rat, dog and human, they were minor; in monkey, M11 was the most abundant. Daporinad was metabolized mainly through N-dealkylation, amide hydrolysis, hydrogenation, oxygenation and dehydrogenation. There was no human-specific metabolite. CONCLUSIONS The current study provided an overview of the metabolism of daporinad, which is helpful in predicting in vivo metabolites and in selecting animal species for toxicology studies.
Collapse
Affiliation(s)
- Shan-Dan Qu
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Guang-Xuan Liu
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
33
|
Tan A, Doig CL. NAD + Degrading Enzymes, Evidence for Roles During Infection. Front Mol Biosci 2021; 8:697359. [PMID: 34485381 PMCID: PMC8415550 DOI: 10.3389/fmolb.2021.697359] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Declines in cellular nicotinamide adenine dinucleotide (NAD) contribute to metabolic dysfunction, increase susceptibility to disease, and occur as a result of pathogenic infection. The enzymatic cleavage of NAD+ transfers ADP-ribose (ADPr) to substrate proteins generating mono-ADP-ribose (MAR), poly-ADP-ribose (PAR) or O-acetyl-ADP-ribose (OAADPr). These important post-translational modifications have roles in both immune response activation and the advancement of infection. In particular, emergent data show viral infection stimulates activation of poly (ADP-ribose) polymerase (PARP) mediated NAD+ depletion and stimulates hydrolysis of existing ADP-ribosylation modifications. These studies are important for us to better understand the value of NAD+ maintenance upon the biology of infection. This review focuses specifically upon the NAD+ utilising enzymes, discusses existing knowledge surrounding their roles in infection, their NAD+ depletion capability and their influence within pathogenic infection.
Collapse
Affiliation(s)
- Arnold Tan
- Interdisciplinary Science and Technology Centre, Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Craig L Doig
- Interdisciplinary Science and Technology Centre, Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
34
|
Daniels VW, Zoeller JJ, van Gastel N, McQueeney KE, Parvin S, Potter DS, Fell GG, Ferreira VG, Yilma B, Gupta R, Spetz J, Bhola PD, Endress JE, Harris IS, Carrilho E, Sarosiek KA, Scadden DT, Brugge JS, Letai A. Metabolic perturbations sensitize triple-negative breast cancers to apoptosis induced by BH3 mimetics. Sci Signal 2021; 14:14/686/eabc7405. [PMID: 34103421 DOI: 10.1126/scisignal.abc7405] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer cells have differential metabolic dependencies compared to their nonmalignant counterparts. However, few metabolism-targeting compounds have been successful in clinical trials. Here, we investigated the metabolic vulnerabilities of triple-negative breast cancer (TNBC), particularly those metabolic perturbations that increased mitochondrial apoptotic priming and sensitivity to BH3 mimetics (drugs that antagonize antiapoptotic proteins). We used high-throughput dynamic BH3 profiling (HT-DBP) to screen a library of metabolism-perturbing small molecules, which revealed inhibitors of the enzyme nicotinamide phosphoribosyltransferase (NAMPT) as top candidates. In some TNBC cells but not in nonmalignant cells, NAMPT inhibitors increased overall apoptotic priming and induced dependencies on specific antiapoptotic BCL-2 family members. Treatment of TNBC cells with NAMPT inhibitors sensitized them to subsequent treatment with BH3 mimetics. The combination of a NAMPT inhibitor (FK866) and an MCL-1 antagonist (S63845) reduced tumor growth in a TNBC patient-derived xenograft model in vivo. We found that NAMPT inhibition reduced NAD+ concentrations below a critical threshold that resulted in depletion of adenine, which was the metabolic trigger that primed TNBC cells for apoptosis. These findings demonstrate a close interaction between metabolic and mitochondrial apoptotic signaling pathways and reveal that exploitation of a tumor-specific metabolic vulnerability can sensitize some TNBC to BH3 mimetics.
Collapse
Affiliation(s)
- Veerle W Daniels
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Ludwig Center at Harvard, Boston, MA 02215, USA
| | - Jason J Zoeller
- Ludwig Center at Harvard, Boston, MA 02215, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| | - Nick van Gastel
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Kelley E McQueeney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Salma Parvin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Danielle S Potter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Geoffrey G Fell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Vinícius G Ferreira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos, SP 13568-250, Brazil.,Instituto Nacional de Ciência e Tecnologia de Bioanalítica, INCTBio, Campinas, SP 13083-970, Brazil
| | - Binyam Yilma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Rajat Gupta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Johan Spetz
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02215, USA
| | - Patrick D Bhola
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jennifer E Endress
- Ludwig Center at Harvard, Boston, MA 02215, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| | - Isaac S Harris
- Ludwig Center at Harvard, Boston, MA 02215, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA.,Department of Biomedical Genetics and Wilmot Cancer Institute, University of Rochester Medical Center, 601 Elmwood Ave., Rochester, NY 14642, USA
| | - Emanuel Carrilho
- Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos, SP 13568-250, Brazil.,Instituto Nacional de Ciência e Tecnologia de Bioanalítica, INCTBio, Campinas, SP 13083-970, Brazil
| | - Kristopher A Sarosiek
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02215, USA
| | - David T Scadden
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Joan S Brugge
- Ludwig Center at Harvard, Boston, MA 02215, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. .,Ludwig Center at Harvard, Boston, MA 02215, USA
| |
Collapse
|
35
|
Ghanem MS, Monacelli F, Nencioni A. Advances in NAD-Lowering Agents for Cancer Treatment. Nutrients 2021; 13:1665. [PMID: 34068917 PMCID: PMC8156468 DOI: 10.3390/nu13051665] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is an essential redox cofactor, but it also acts as a substrate for NAD-consuming enzymes, regulating cellular events such as DNA repair and gene expression. Since such processes are fundamental to support cancer cell survival and proliferation, sustained NAD production is a hallmark of many types of neoplasms. Depleting intratumor NAD levels, mainly through interference with the NAD-biosynthetic machinery, has emerged as a promising anti-cancer strategy. NAD can be generated from tryptophan or nicotinic acid. In addition, the "salvage pathway" of NAD production, which uses nicotinamide, a byproduct of NAD degradation, as a substrate, is also widely active in mammalian cells and appears to be highly exploited by a subset of human cancers. In fact, research has mainly focused on inhibiting the key enzyme of the latter NAD production route, nicotinamide phosphoribosyltransferase (NAMPT), leading to the identification of numerous inhibitors, including FK866 and CHS-828. Unfortunately, the clinical activity of these agents proved limited, suggesting that the approaches for targeting NAD production in tumors need to be refined. In this contribution, we highlight the recent advancements in this field, including an overview of the NAD-lowering compounds that have been reported so far and the related in vitro and in vivo studies. We also describe the key NAD-producing pathways and their regulation in cancer cells. Finally, we summarize the approaches that have been explored to optimize the therapeutic response to NAMPT inhibitors in cancer.
Collapse
Affiliation(s)
- Moustafa S. Ghanem
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
36
|
Huffaker TB, Ekiz HA, Barba C, Lee SH, Runtsch MC, Nelson MC, Bauer KM, Tang WW, Mosbruger TL, Cox JE, Round JL, Voth WP, O'Connell RM. A Stat1 bound enhancer promotes Nampt expression and function within tumor associated macrophages. Nat Commun 2021; 12:2620. [PMID: 33976173 PMCID: PMC8113251 DOI: 10.1038/s41467-021-22923-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 03/30/2021] [Indexed: 02/03/2023] Open
Abstract
Tumor associated macrophage responses are regulated by distinct metabolic states that affect their function. However, the ability of specific signals in the local tumor microenvironment to program macrophage metabolism remains under investigation. Here, we identify NAMPT, the rate limiting enzyme in NAD salvage synthesis, as a target of STAT1 during cellular activation by interferon gamma, an important driver of macrophage polarization and antitumor responses. We demonstrate that STAT1 occupies a conserved element within the first intron of Nampt, termed Nampt-Regulatory Element-1 (NRE1). Through disruption of NRE1 or pharmacological inhibition, a subset of M1 genes is sensitive to NAMPT activity through its impact on glycolytic processes. scRNAseq is used to profile in vivo responses by NRE1-deficient, tumor-associated leukocytes in melanoma tumors through the creation of a unique mouse strain. Reduced Nampt and inflammatory gene expression are present in specific myeloid and APC populations; moreover, targeted ablation of NRE1 in macrophage lineages results in greater tumor burden. Finally, elevated NAMPT expression correlates with IFNγ responses and melanoma patient survival. This study identifies IFN and STAT1-inducible Nampt as an important factor that shapes the metabolic program and function of tumor associated macrophages.
Collapse
Affiliation(s)
- Thomas B Huffaker
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - H Atakan Ekiz
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Cindy Barba
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Soh-Hyun Lee
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Marah C Runtsch
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Morgan C Nelson
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Kaylyn M Bauer
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - William W Tang
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | | | - James E Cox
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
| | - June L Round
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Warren P Voth
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA.
| | - Ryan M O'Connell
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA.
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
37
|
Navarro MN, Gómez de Las Heras MM, Mittelbrunn M. Nicotinamide adenine dinucleotide metabolism in the immune response, autoimmunity and inflammageing. Br J Pharmacol 2021; 179:1839-1856. [PMID: 33817782 PMCID: PMC9292562 DOI: 10.1111/bph.15477] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Metabolism is dynamically regulated to accompany immune cell function, and altered immunometabolism can result in impaired immune responses. Concomitantly, the pharmacological manipulation of metabolic processes offers an opportunity for therapeutic intervention in inflammatory disorders. The nicotinamide adenine dinucleotide (NAD+) is a critical metabolic intermediate that serves as enzyme cofactor in redox reactions, and is also used as a co‐substrate by many enzymes such as sirtuins, adenosine diphosphate ribose transferases and synthases. Through these activities, NAD+ metabolism regulates a broad spectrum of cellular functions such as energy metabolism, DNA repair, regulation of the epigenetic landscape and inflammation. Thus, the manipulation of NAD+ availability using pharmacological compounds such as NAD+ precursors can have immune‐modulatory properties in inflammation. Here, we discuss how the NAD+ metabolism contributes to the immune response and inflammatory conditions, with a special focus on multiple sclerosis, inflammatory bowel diseases and inflammageing.
Collapse
Affiliation(s)
- Maria N Navarro
- Interactions With The Environment Program, Immune System Development and Function Unit, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Manuel M Gómez de Las Heras
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Maria Mittelbrunn
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital 12 de Octubre (i+12), Madrid, Spain
| |
Collapse
|
38
|
Pillai VB, Gupta MP. Is nuclear sirtuin SIRT6 a master regulator of immune function? Am J Physiol Endocrinol Metab 2021; 320:E399-E414. [PMID: 33308014 PMCID: PMC7988780 DOI: 10.1152/ajpendo.00483.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 12/29/2022]
Abstract
The ability to ward off pathogens with minimal damage to the host determines the immune system's robustness. Multiple factors, including pathogen processing, identification, secretion of mediator and effector molecules, and immune cell proliferation and differentiation into various subsets, constitute the success of mounting an effective immune response. Cellular metabolism controls all of these intricate processes. Cells utilize diverse fuel sources and switch back and forth between different metabolic pathways depending on their energy needs. The three most critical metabolic pathways on which immune cells depend to meet their energy needs are oxidative metabolism, glycolysis, and glutaminolysis. Dynamic switching between these metabolic pathways is needed for optimal function of the immune cells. Moreover, switching between these metabolic pathways needs to be tightly regulated to achieve the best results. Immune cells depend on the Warburg effect for their growth, proliferation, secretory, and effector functions. Here, we hypothesize that the sirtuin, SIRT6, could be a negative regulator of the Warburg effect. We also postulate that SIRT6 could act as a master regulator of immune cell metabolism and function by regulating critical signaling pathways.
Collapse
Affiliation(s)
- Vinodkumar B Pillai
- Department of Surgery (Division of Cardiothoracic Surgery), Pritzker School of Medicine, Basic Science Division, University of Chicago, Chicago, Illinois
| | - Mahesh P Gupta
- Department of Surgery (Division of Cardiothoracic Surgery), Pritzker School of Medicine, Basic Science Division, University of Chicago, Chicago, Illinois
| |
Collapse
|
39
|
Liu H, Zhang J, Xu X, Lu S, Yang D, Xie C, Jia M, Zhang W, Jin L, Wang X, Shen X, Li F, Wang W, Bao X, Li S, Zhu M, Wang W, Wang Y, Huang Z, Teng H. SARM1 promotes neuroinflammation and inhibits neural regeneration after spinal cord injury through NF-κB signaling. Am J Cancer Res 2021; 11:4187-4206. [PMID: 33754056 PMCID: PMC7977471 DOI: 10.7150/thno.49054] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 01/17/2021] [Indexed: 12/18/2022] Open
Abstract
Axonal degeneration is a common pathological feature in many acute and chronic neurological diseases such as spinal cord injury (SCI). SARM1 (sterile alpha and TIR motif-containing 1), the fifth TLR (Toll-like receptor) adaptor, has diverse functions in the immune and nervous systems, and recently has been identified as a key mediator of Wallerian degeneration (WD). However, the detailed functions of SARM1 after SCI still remain unclear. Methods: Modified Allen's method was used to establish a contusion model of SCI in mice. Furthermore, to address the function of SARM1 after SCI, conditional knockout (CKO) mice in the central nervous system (CNS), SARM1Nestin-CKO mice, and SARM1GFAP-CKO mice were successfully generated by Nestin-Cre and GFAP-Cre transgenic mice crossed with SARM1flox/flox mice, respectively. Immunostaining, Hematoxylin-Eosin (HE) staining, Nissl staining and behavioral test assays such as footprint and Basso Mouse Scale (BMS) scoring were used to examine the roles of SARM1 pathway in SCI based on these conditional knockout mice. Drugs such as FK866, an inhibitor of SARM1, and apoptozole, an inhibitor of heat shock protein 70 (HSP70), were used to further explore the molecular mechanism of SARM1 in neural regeneration after SCI. Results: We found that SARM1 was upregulated in neurons and astrocytes at early stage after SCI. SARM1Nestin-CKO and SARM1GFAP-CKO mice displayed normal development of the spinal cords and motor function. Interestingly, conditional deletion of SARM1 in neurons and astrocytes promoted the functional recovery of behavior performance after SCI. Mechanistically, conditional deletion of SARM1 in neurons and astrocytes promoted neuronal regeneration at intermediate phase after SCI, and reduced neuroinflammation at SCI early phase through downregulation of NF-κB signaling after SCI, which may be due to upregulation of HSP70. Finally, FK866, an inhibitor of SARM1, reduced the neuroinflammation and promoted the neuronal regeneration after SCI. Conclusion: Our results indicate that SARM1-mediated prodegenerative pathway and neuroinflammation promotes the pathological progress of SCI and anti-SARM1 therapeutics are viable and promising approaches for preserving neuronal function after SCI.
Collapse
|
40
|
Covarrubias AJ, Perrone R, Grozio A, Verdin E. NAD + metabolism and its roles in cellular processes during ageing. Nat Rev Mol Cell Biol 2021; 22:119-141. [PMID: 33353981 PMCID: PMC7963035 DOI: 10.1038/s41580-020-00313-x] [Citation(s) in RCA: 801] [Impact Index Per Article: 200.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a coenzyme for redox reactions, making it central to energy metabolism. NAD+ is also an essential cofactor for non-redox NAD+-dependent enzymes, including sirtuins, CD38 and poly(ADP-ribose) polymerases. NAD+ can directly and indirectly influence many key cellular functions, including metabolic pathways, DNA repair, chromatin remodelling, cellular senescence and immune cell function. These cellular processes and functions are critical for maintaining tissue and metabolic homeostasis and for healthy ageing. Remarkably, ageing is accompanied by a gradual decline in tissue and cellular NAD+ levels in multiple model organisms, including rodents and humans. This decline in NAD+ levels is linked causally to numerous ageing-associated diseases, including cognitive decline, cancer, metabolic disease, sarcopenia and frailty. Many of these ageing-associated diseases can be slowed down and even reversed by restoring NAD+ levels. Therefore, targeting NAD+ metabolism has emerged as a potential therapeutic approach to ameliorate ageing-related disease, and extend the human healthspan and lifespan. However, much remains to be learnt about how NAD+ influences human health and ageing biology. This includes a deeper understanding of the molecular mechanisms that regulate NAD+ levels, how to effectively restore NAD+ levels during ageing, whether doing so is safe and whether NAD+ repletion will have beneficial effects in ageing humans.
Collapse
Affiliation(s)
- Anthony J Covarrubias
- Buck Institute for Research on Aging, Novato, CA, USA
- UCSF Department of Medicine, San Francisco, CA, USA
| | | | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA.
- UCSF Department of Medicine, San Francisco, CA, USA.
| |
Collapse
|
41
|
Drijvers JM, Gillis JE, Muijlwijk T, Nguyen TH, Gaudiano EF, Harris IS, LaFleur MW, Ringel AE, Yao CH, Kurmi K, Juneja VR, Trombley JD, Haigis MC, Sharpe AH. Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8 + T Cells. Cancer Immunol Res 2021; 9:184-199. [PMID: 33277233 PMCID: PMC7864883 DOI: 10.1158/2326-6066.cir-20-0384] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/19/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022]
Abstract
Metabolic constraints in the tumor microenvironment constitute a barrier to effective antitumor immunity and similarities in the metabolic properties of T cells and cancer cells impede the specific therapeutic targeting of metabolism in either population. To identify distinct metabolic vulnerabilities of CD8+ T cells and cancer cells, we developed a high-throughput in vitro pharmacologic screening platform and used it to measure the cell type-specific sensitivities of activated CD8+ T cells and B16 melanoma cells to a wide array of metabolic perturbations during antigen-specific killing of cancer cells by CD8+ T cells. We illustrated the applicability of this screening platform by showing that CD8+ T cells were more sensitive to ferroptosis induction by inhibitors of glutathione peroxidase 4 (GPX4) than B16 and MC38 cancer cells. Overexpression of ferroptosis suppressor protein 1 (FSP1) or cytosolic GPX4 yielded ferroptosis-resistant CD8+ T cells without compromising their function, while genetic deletion of the ferroptosis sensitivity-promoting enzyme acyl-CoA synthetase long-chain family member 4 (ACSL4) protected CD8+ T cells from ferroptosis but impaired antitumor CD8+ T-cell responses. Our screen also revealed high T cell-specific vulnerabilities for compounds targeting NAD+ metabolism or autophagy and endoplasmic reticulum (ER) stress pathways. We focused the current screening effort on metabolic agents. However, this in vitro screening platform may also be valuable for rapid testing of other types of compounds to identify regulators of antitumor CD8+ T-cell function and potential therapeutic targets.
Collapse
Affiliation(s)
- Jefte M Drijvers
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Jacob E Gillis
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Tara Muijlwijk
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Thao H Nguyen
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Emily F Gaudiano
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Isaac S Harris
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Martin W LaFleur
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Alison E Ringel
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Cong-Hui Yao
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Kiran Kurmi
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Vikram R Juneja
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Justin D Trombley
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts.
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts.
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
42
|
Integrative computational approach identifies drug targets in CD4 + T-cell-mediated immune disorders. NPJ Syst Biol Appl 2021; 7:4. [PMID: 33483502 PMCID: PMC7822845 DOI: 10.1038/s41540-020-00165-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
CD4+ T cells provide adaptive immunity against pathogens and abnormal cells, and they are also associated with various immune-related diseases. CD4+ T cells’ metabolism is dysregulated in these pathologies and represents an opportunity for drug discovery and development. Genome-scale metabolic modeling offers an opportunity to accelerate drug discovery by providing high-quality information about possible target space in the context of a modeled disease. Here, we develop genome-scale models of naïve, Th1, Th2, and Th17 CD4+ T-cell subtypes to map metabolic perturbations in rheumatoid arthritis, multiple sclerosis, and primary biliary cholangitis. We subjected these models to in silico simulations for drug response analysis of existing FDA-approved drugs and compounds. Integration of disease-specific differentially expressed genes with altered reactions in response to metabolic perturbations identified 68 drug targets for the three autoimmune diseases. In vitro experimental validation, together with literature-based evidence, showed that modulation of fifty percent of identified drug targets suppressed CD4+ T cells, further increasing their potential impact as therapeutic interventions. Our approach can be generalized in the context of other diseases, and the metabolic models can be further used to dissect CD4+ T-cell metabolism.
Collapse
|
43
|
Benzi A, Sturla L, Heine M, Fischer AW, Spinelli S, Magnone M, Sociali G, Parodi A, Fenoglio D, Emionite L, Koch-Nolte F, Mittrücker HW, Guse AH, De Flora A, Zocchi E, Heeren J, Bruzzone S. CD38 downregulation modulates NAD + and NADP(H) levels in thermogenic adipose tissues. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1866:158819. [PMID: 33010451 DOI: 10.1016/j.bbalip.2020.158819] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/05/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
Different strategies to boost NAD+ levels are considered promising means to promote healthy aging and ameliorate dysfunctional metabolism. CD38 is a NAD+-dependent enzyme involved in the regulation of different cell functions. In the context of systemic energy metabolism, it has been demonstrated that brown adipocytes, the parenchymal cells of brown adipose tissue (BAT) as well as beige adipocytes that emerge in white adipose tissue (WAT) depots in response to catabolic conditions, are important to maintain metabolic homeostasis. In this study we aim to understand the functional relevance of CD38 for NAD+ and energy metabolism in BAT and WAT, also using a CD38-/- mouse model. During cold exposure, an increase in NAD+ levels occurred in BAT of wild type mice, together with a marked downregulation of CD38, as detected at the mRNA and protein level. CD38 downregulation was observed also in WAT of cold-exposed mice, where it was accompanied by a strong increase in NADP(H) levels. Accordingly, NAD kinase and glucose-6-phosphate dehydrogenase activities were enhanced in WAT (but not in BAT). Increased NAD+ levels were observed in BAT/WAT from CD38-/- compared with wild type mice, in line with CD38 being a major NAD+-consumer in AT. CD38-/- mice kept at 6 °C had higher levels of Ucp1 and Pgc-1α in BAT and WAT, and increased levels of phosphorylated hormone-sensitive lipase in BAT, compared with wild type mice. These results demonstrate that CD38, by modulating cellular NAD(P)+ levels, is involved in the regulation of thermogenic responses in cold-activated BAT and WAT.
Collapse
Affiliation(s)
- Andrea Benzi
- DIMES-Section of Biochemistry, University of Genova, Italy
| | - Laura Sturla
- DIMES-Section of Biochemistry, University of Genova, Italy.
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander W Fischer
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonia Spinelli
- DIMES-Section of Biochemistry, University of Genova, Italy
| | - Mirko Magnone
- DIMES-Section of Biochemistry, University of Genova, Italy
| | | | | | - Daniela Fenoglio
- IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine, University of Genova, Italy
| | - Laura Emionite
- Animal Facility, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132 Genova, Italy
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Willi Mittrücker
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H Guse
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Elena Zocchi
- DIMES-Section of Biochemistry, University of Genova, Italy
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|
44
|
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: 38] [Impact Index Per Article: 7.6] [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
|
45
|
Soncini D, Minetto P, Martinuzzi C, Becherini P, Fenu V, Guolo F, Todoerti K, Calice G, Contini P, Miglino M, Rivoli G, Aquino S, Dominietto A, Cagnetta A, Passalacqua M, Bruzzone S, Nencioni A, Zucchetti M, Ceruti T, Neri A, Lemoli RM, Cea M. Amino acid depletion triggered by ʟ-asparaginase sensitizes MM cells to carfilzomib by inducing mitochondria ROS-mediated cell death. Blood Adv 2020; 4:4312-4326. [PMID: 32915979 PMCID: PMC7509874 DOI: 10.1182/bloodadvances.2020001639] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/26/2020] [Indexed: 02/07/2023] Open
Abstract
Metabolic reprogramming is emerging as a cancer vulnerability that could be therapeutically exploitable using different approaches, including amino acid depletion for those tumors that rely on exogenous amino acids for their maintenance. ʟ-Asparaginase (ASNase) has contributed to a significant improvement in acute lymphoblastic leukemia outcomes; however, toxicity and resistance limit its clinical use in other tumors. Here, we report that, in multiple myeloma (MM) cells, the DNA methylation status is significantly associated with reduced expression of ASNase-related gene signatures, thus suggesting ASNase sensitivity for this tumor. Therefore, we tested the effects of ASNase purified from Erwinia chrysanthemi (Erw-ASNase), combined with the next-generation proteasome inhibitor (PI) carfilzomib. We observed an impressive synergistic effect on MM cells, whereas normal peripheral blood mononuclear cells were not affected. Importantly, this effect was associated with increased reactive oxygen species (ROS) generation, compounded mitochondrial damage, and Nrf2 upregulation, regardless of the c-Myc oncogenic-specific program. Furthermore, the cotreatment resulted in genomic instability and DNA repair mechanism impairment via increased mitochondrial oxidative stress, which further enhanced its antitumor activity. Interestingly, carfilzomib-resistant cells were found to be highly dependent on amino acid starvation, as reflected by their higher sensitivity to Erw-ASNase treatment compared with isogenic cells. Overall, by affecting several cellular programs, Erw-ASNase makes MM cells more vulnerable to carfilzomib, providing proof of concept for clinical use of this combination as a novel strategy to enhance PI sensitivity in MM patients.
Collapse
Affiliation(s)
- Debora Soncini
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Paola Minetto
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Division of Hematology and Hematopoietic Stem Cell Transplantation Unit, Ospedale Policlinico San Martino, Genoa, Italy
| | - Claudia Martinuzzi
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Pamela Becherini
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Valeria Fenu
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fabio Guolo
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Katia Todoerti
- Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan, Italy
| | - Giovanni Calice
- IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | | | - Maurizio Miglino
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giulia Rivoli
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Sara Aquino
- Division of Hematology and Hematopoietic Stem Cell Transplantation Unit, Ospedale Policlinico San Martino, Genoa, Italy
| | - Alida Dominietto
- Division of Hematology and Hematopoietic Stem Cell Transplantation Unit, Ospedale Policlinico San Martino, Genoa, Italy
| | - Antonia Cagnetta
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Mario Passalacqua
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Alessio Nencioni
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and
| | - Massimo Zucchetti
- Clinical Cancer Pharmacology Unit, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; and
| | - Tommaso Ceruti
- Clinical Cancer Pharmacology Unit, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; and
| | - Antonino Neri
- Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Roberto M Lemoli
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Michele Cea
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
46
|
Ferrara G, Benzi A, Sturla L, Marubbi D, Frumento D, Spinelli S, Abbotto E, Ivaldi F, von Holtey M, Murone M, Nencioni A, Uccelli A, Bruzzone S. Sirt6 inhibition delays the onset of experimental autoimmune encephalomyelitis by reducing dendritic cell migration. J Neuroinflammation 2020; 17:228. [PMID: 32736564 PMCID: PMC7393881 DOI: 10.1186/s12974-020-01906-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/20/2020] [Indexed: 01/14/2023] Open
Abstract
Background Experimental autoimmune encephalomyelitis (EAE) is the most common animal model of multiple sclerosis (MS), a neuroinflammatory and demyelinating disease characterized by multifocal perivascular infiltrates of immune cells. Although EAE is predominantly considered a T helper 1-driven autoimmune disease, mounting evidence suggests that activated dendritic cells (DC), which are the bridge between innate and adaptive immunity, also contribute to its pathogenesis. Sirtuin 6 (SIRT6), a NAD+-dependent deacetylase involved in genome maintenance and in metabolic homeostasis, regulates DC activation, and its pharmacological inhibition could, therefore, play a role in EAE development. Methods EAE was induced in female C57bl/6 mice by MOG35-55 injection. The effect of treatment with a small compound SIRT6 inhibitor, administered according to therapeutic and preventive protocols, was assessed by evaluating the clinical EAE score. SIRT6 inhibition was confirmed by Western blot analysis by assessing the acetylation of histone 3 lysine 9, a known SIRT6 substrate. The expression of DC activation and migration markers was evaluated by FACS in mouse lymph nodes. In addition, the expression of inflammatory and anti-inflammatory cytokines in the spinal cord were assessed by qPCR. T cell infiltration in spinal cords was evaluated by immunofluorescence imaging. The effect of Sirt6 inhibition on the migration of resting and activated bone marrow-derived dendritic cells was investigated in in vitro chemotaxis assays. Results Preventive pharmacological Sirt6 inhibition effectively delayed EAE disease onset through a novel regulatory mechanism, i.e., by reducing the representation of CXCR4-positive and of CXCR4/CCR7-double-positive DC in lymph nodes. The delay in EAE onset correlated with the early downregulation in the expression of CD40 on activated lymph node DC, with increased level of the anti-inflammatory cytokine IL-10, and with a reduced encephalitogenic T cell infiltration in the central nervous system. Consistent with the in vivo data, in vitro pharmacological Sirt6 inhibition in LPS-stimulated, bone marrow-derived DC reduced CCL19/CCL21- and SDF-1-induced DC migration. Conclusions Our findings indicate the ability of Sirt6 inhibition to impair DC migration, to downregulate pathogenic T cell inflammatory responses and to delay EAE onset. Therefore, Sirt6 might represent a valuable target for developing novel therapeutic agents for the treatment of early stages of MS, or of other autoimmune disorders.
Collapse
Affiliation(s)
- Giovanni Ferrara
- Ospedale Policlinico San Martino, IRCCS, Largo R. Benzi, 10, 16132, Genova, Italy.
| | - Andrea Benzi
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Laura Sturla
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Daniela Marubbi
- Ospedale Policlinico San Martino, IRCCS, Largo R. Benzi, 10, 16132, Genova, Italy.,Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Davide Frumento
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Sonia Spinelli
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Elena Abbotto
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Federico Ivaldi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy
| | | | | | - Alessio Nencioni
- Ospedale Policlinico San Martino, IRCCS, Largo R. Benzi, 10, 16132, Genova, Italy.,Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genova, Italy
| | - Antonio Uccelli
- Ospedale Policlinico San Martino, IRCCS, Largo R. Benzi, 10, 16132, Genova, Italy.,Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| |
Collapse
|
47
|
Sundaram G, Lim CK, Brew BJ, Guillemin GJ. Kynurenine pathway modulation reverses the experimental autoimmune encephalomyelitis mouse disease progression. J Neuroinflammation 2020; 17:176. [PMID: 32505212 PMCID: PMC7276083 DOI: 10.1186/s12974-020-01844-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/14/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic immune-mediated disorder of the central nervous system characterized by demyelination, neuroinflammation, and neurodegeneration. Activation of the kynurenine pathway (KP) results from acute and chronic neuroinflammation leading to both immune suppression and neurotoxicity. However, the exact effects of KP metabolites and changes in neurodegenerative diseases over time are not fully understood. Studies, including those in MS models, have reported that short-term KP activation is beneficial through immune tolerance. However, the effects of long-term KP activation are poorly understood. We hypothesized that such chronic activation is responsible for the neurodegeneration in MS, and further, modulating the KP in EAE-induced mice could significantly decrease the EAE disease severity. METHODS We biochemically altered the KP at different stages of the disease in experimental allergic encephalomyelitis (EAE) mouse model of MS and at two different enzymatic levels of the KP (IDO-1 (indoleamine 2,3 dioxygenase)) and KMO (kynurenine monooxygenase). CNS tissue and blood samples were analyzed longitudinally using GCMS, HPLC, IHC, and RT-PCR. RESULTS We showed that the KP was steadily upregulated correlating with disease severity and associated with a shift towards increasing concentrations of the KP metabolite quinolinic acid, a neuro- and gliotoxin. KP modulation by inhibition of IDO-1 with 1-methyl tryptophan (1-MT) was dependent on the timing of treatment at various stages of EAE. IDO-1 inhibition at EAE score 2 led to significantly higher numbers of FoxP3 cells (p < 0.001) in the spleen than earlier IDO-1 inhibition (prophylactic 1-MT treatment group (p < 0.001)), 1-MT treatment after EAE induction (EAE score 0; p < 0.001), and 1-MT treatment at EAE score of 1 (p < 0.05). Significant improvement of disease severity was observed in EAE mice treated with 1-MT at EAE score 2 compared to the untreated group (p < 0.05). KP modulation by KMO inhibition with Ro 61-8048 led to significantly greater numbers of Foxp3 cells (p < 0.05) in Ro 61-8048 treated mice and even more significant amelioration of EAE disease compared to the 1-MT treatment groups. CONCLUSIONS These results provide a new mechanistic link between neuroinflammation and neurodegeneration and point to KP modulation at the KMO level to preserve immune tolerance and limit neurodegeneration in EAE. They provide the foundation for new clinical trials for MS.
Collapse
Affiliation(s)
- Gayathri Sundaram
- Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Chai K Lim
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Bruce J Brew
- Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, NSW, 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia.
- Department of Neurology, St Vincent's Hospital, Sydney, NSW, 2010, Australia.
| | - Gilles J Guillemin
- Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, NSW, 2010, Australia.
- Neuroinflammation Group, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
48
|
Schultz MD, Dadali T, Jacques SA, Muller-Steffner H, Foote JB, Sorci L, Kellenberger E, Botta D, Lund FE. Inhibition of the NAD salvage pathway in schistosomes impairs metabolism, reproduction, and parasite survival. PLoS Pathog 2020; 16:e1008539. [PMID: 32459815 PMCID: PMC7252647 DOI: 10.1371/journal.ppat.1008539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
NAD, a key co-enzyme required for cell metabolism, is synthesized via two pathways in most organisms. Since schistosomes apparently lack enzymes required for de novo NAD biosynthesis, we evaluated whether these parasites, which infect >200 million people worldwide, maintain NAD homeostasis via the NAD salvage biosynthetic pathway. We found that intracellular NAD levels decline in schistosomes treated with drugs that block production of nicotinamide or nicotinamide mononucleotide–known NAD precursors in the non-deamidating salvage pathway. Moreover, in vitro inhibition of the NAD salvage pathway in schistosomes impaired egg production, disrupted the outer membranes of both immature and mature parasites and caused loss of mobility and death. Inhibiting the NAD salvage pathway in schistosome-infected mice significantly decreased NAD levels in adult parasites, which correlated with reduced egg production, fewer liver granulomas and parasite death. Thus, schistosomes, unlike their mammalian hosts, appear limited to one metabolic pathway to maintain NAD-dependent metabolic processes. Schistosomiasis (snail fever) is a deadly parasitic disease that affects more than 200 million people worldwide and, if not treated, can lead to death. This disease is caused by parasitic worms called schistosomes that feed on the host blood and lay hundreds of eggs each day that damage the liver and kidneys. Therapies to treat schistosomiasis are limited. The most widely-used anti-schistosomal drug, praziquantel, is not effective against immature parasites and adult worms can, in some cases, become resistant to this drug. It is therefore important to find new therapies to treat this deadly disease. In this study, we observed that schistosomes cannot use amino acids to make Nicotinamide Adenine Dinucleotide (NAD)–a key cellular metabolite found in all living organisms. Instead, these parasites salvage NAD by scavenging vitamins from the host. We observed that disruption of this NAD salvage pathway negatively impacts metabolism, reproduction and survival of both adult and immature worms. As such, targeting the parasite’s NAD salvage pathway is a promising therapeutic approach for the treatment of snail fever.
Collapse
Affiliation(s)
- Michael D. Schultz
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Tulin Dadali
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Sylvain A. Jacques
- Laboratoire d’Innovation Thérapeutique, LIT UMR 7200 CNRS-Université de Strasbourg, MEDALIS Drug Discovery Center, Faculté de Pharmacie, Illkirch, France
| | - Hélène Muller-Steffner
- Laboratoire des Systèmes Chimiques Fonctionnels, CAMB UMR 7199 CNRS-Université de Strasbourg, MEDALIS Drug Discovery Center, Faculté de Pharmacie, Illkirch, France
| | - Jeremy B. Foote
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Leonardo Sorci
- Department of Materials, Environmental Sciences and Urban Planning, Division of Bioinformatics and Biochemistry, Polytechnic University of Marche, Ancona, Italy
| | - Esther Kellenberger
- Laboratoire d’Innovation Thérapeutique, LIT UMR 7200 CNRS-Université de Strasbourg, MEDALIS Drug Discovery Center, Faculté de Pharmacie, Illkirch, France
| | - Davide Botta
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Frances E. Lund
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
49
|
Galli U, Colombo G, Travelli C, Tron GC, Genazzani AA, Grolla AA. Recent Advances in NAMPT Inhibitors: A Novel Immunotherapic Strategy. Front Pharmacol 2020; 11:656. [PMID: 32477131 PMCID: PMC7235340 DOI: 10.3389/fphar.2020.00656] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is a cofactor of many enzymatic reactions as well as being a substrate for a number of NAD-consuming enzymes (e.g., PARPS, sirtuins, etc). NAD can be synthesized de novo starting from tryptophan, nicotinamide, nicotinic acid, or nicotinamide riboside from the diet. On the other hand, the nicotinamide that is liberated by NAD-consuming enzymes can be salvaged to re-form NAD. In this former instance, nicotinamide phosphoribosyltransferase (NAMPT) is the bottleneck enzyme. In the many cells in which the salvage pathway is predominant, NAMPT, therefore, represents an important controller of intracellular NAD concentrations, and as a consequence of energy metabolism. It is, therefore, not surprising that NAMPT is over expressed by tumoral cells, which take advantage from this to sustain growth rate and tumor progression. This has led to the initiation of numerous medicinal chemistry programs to develop NAMPT inhibitors in the context of oncology. More recently, however, it has been shown that NAMPT inhibitors do not solely target the tumor but also have an effect on the immune system. To add complexity, this enzyme can also be secreted by cells, and in the extracellular space it acts as a cytokine mainly through the activation of Toll like Receptor 4 (TLR4), although it has not been clarified yet if this is the only receptor responsible for its actions. While specific small molecules have been developed only against the intracellular form of NAMPT, growing evidences sustain the possibility to target the extracellular form. In this contribution, the most recent evidences on the medicinal chemistry of NAMPT will be reviewed, together with the key elements that sustain the hypothesis of NAMPT targeting and the drawbacks so far encountered.
Collapse
Affiliation(s)
- Ubaldina Galli
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Giorgia Colombo
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Cristina Travelli
- Department of Pharmaceutical Sciences, University of Pavia, Pavia, Italy
| | - Gian Cesare Tron
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Ambra A Grolla
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
50
|
Tang JZ, Xu WQ, Wei FJ, Jiang YZ, Zheng XX. Role of Nampt overexpression in a rat model of Hashimoto's thyroiditis and its mechanism of action. Exp Ther Med 2020; 19:2895-2900. [PMID: 32256774 PMCID: PMC7086292 DOI: 10.3892/etm.2020.8539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022] Open
Abstract
The present study was designed to investigate the role of nicotinamide phosphoribosyltransferase (Nampt) overexpression in a rat model of Hashimoto's thyroiditis (HT) and its mechanism of action. A rat model of HT was constructed, and the HT rats were injected with an adenoviral expression vector carrying the Nampt gene. The expression of Nampt and Toll-like receptor 4 (TLR4) in thyroid tissues was examined using immunohistochemistry (IHC), RT-qPCR and western blot analyses. Serum anti-thyroglobulin antibodies (TGAb) and anti-thyroid peroxidase antibodies (TPOAb) were measured using chemiluminescence method. Hematoxylin and eosin (H&E) and IHC staining of the rat thyroid tissues showed destroyed thyroid follicles and monocyte infiltration, as well as increased Nampt expression in the thyroid tissues of rats with HT. Furthermore, it was found that Nampt overexpression led to increased severity of inflammatory infiltration in thyroid tissues and increased levels of TPOAb in the serum of HT rats; however, the serum TGAb level was not affected by Nampt overexpression. In addition, Nampt overexpression promoted TLR4 expression in HT rats. In conclusion, it was demonstrated that Nampt was strongly expressed in the capillary region of HT rats thyroid tissues. The Nampt mRNA level was increased but the Nampt protein level was decreased in the thyroid tissues of rats with HT. Nampt overexpression has a promotive effect on HT progression, and this effect was related to TLR4. This study suggests that inhibition of Nampt activity may be valuable in the treatment of HT.
Collapse
Affiliation(s)
- Jia-Zhen Tang
- Department of Endocrinology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wen-Qiong Xu
- Department of Endocrinology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fu-Juan Wei
- Department of Endocrinology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yang-Zhen Jiang
- Department of Endocrinology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiao-Xue Zheng
- Department of Endocrinology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|