1
|
Nasuhidehnavi A, Zarzycka W, Górecki I, Chiao YA, Lee CF. Emerging interactions between mitochondria and NAD + metabolism in cardiometabolic diseases. Trends Endocrinol Metab 2024:S1043-2760(24)00191-7. [PMID: 39198117 DOI: 10.1016/j.tem.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 09/01/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential coenzyme for redox reactions and regulates cellular catabolic pathways. An intertwined relationship exists between NAD+ and mitochondria, with consequences for mitochondrial function. Dysregulation in NAD+ homeostasis can lead to impaired energetics and increased oxidative stress, contributing to the pathogenesis of cardiometabolic diseases. In this review, we explore how disruptions in NAD+ homeostasis impact mitochondrial function in various cardiometabolic diseases. We discuss emerging studies demonstrating that enhancing NAD+ synthesis or inhibiting its consumption can ameliorate complications of this family of pathological conditions. Additionally, we highlight the potential role and therapeutic promise of mitochondrial NAD+ transporters in regulating cellular and mitochondrial NAD+ homeostasis.
Collapse
Affiliation(s)
- Azadeh Nasuhidehnavi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13790, USA
| | - Weronika Zarzycka
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ignacy Górecki
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Ying Ann Chiao
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Chi Fung Lee
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
2
|
Choi S, Lee Y, Park S, Jang SY, Park J, Oh DW, Kim SM, Kim TH, Lee GS, Cho C, Kim BS, Lee D, Kim EH, Cheong HK, Moon JH, Song JJ, Hwang J, Kim MH. Dissemination of pathogenic bacteria is reinforced by a MARTX toxin effector duet. Nat Commun 2024; 15:6218. [PMID: 39043696 PMCID: PMC11266601 DOI: 10.1038/s41467-024-50650-0] [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: 02/02/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024] Open
Abstract
Multiple bacterial genera take advantage of the multifunctional autoprocessing repeats-in-toxin (MARTX) toxin to invade host cells. Secretion of the MARTX toxin by Vibrio vulnificus, a deadly opportunistic pathogen that causes primary septicemia, the precursor of sepsis, is a major driver of infection; however, the molecular mechanism via which the toxin contributes to septicemia remains unclear. Here, we report the crystal and cryo-electron microscopy (EM) structures of a toxin effector duet comprising the domain of unknown function in the first position (DUF1)/Rho inactivation domain (RID) complexed with human targets. These structures reveal how the duet is used by bacteria as a potent weapon. The data show that DUF1 acts as a RID-dependent transforming NADase domain (RDTND) that disrupts NAD+ homeostasis by hijacking calmodulin. The cryo-EM structure of the RDTND-RID duet complexed with calmodulin and Rac1, together with immunological analyses in vitro and in mice, provide mechanistic insight into how V. vulnificus uses the duet to suppress ROS generation by depleting NAD(P)+ and modifying Rac1 in a mutually-reinforcing manner that ultimately paralyzes first line immune responses, promotes dissemination of invaders, and induces sepsis. These data may allow development of tools or strategies to combat MARTX toxin-related human diseases.
Collapse
Affiliation(s)
- Sanghyeon Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Korea
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Youngjin Lee
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Shinhye Park
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, 34134, Korea
| | - Song Yee Jang
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Core Research Facility & Analysis Center, KRIBB, Daejeon, 34141, Korea
| | - Jongbin Park
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Do Won Oh
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Korea
| | - Su-Man Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Biology Education, Chonnam National University, Gwangju, 61186, Korea
| | - Tae-Hwan Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea
| | - Ga Seul Lee
- Core Research Facility & Analysis Center, KRIBB, Daejeon, 34141, Korea
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, 28644, Korea
| | - Changyi Cho
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul, 03760, Korea
| | - Byoung Sik Kim
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul, 03760, Korea
| | - Donghan Lee
- Korea Basic Science Institute, Cheongju, Chungbuk, 28119, Korea
| | - Eun-Hee Kim
- Korea Basic Science Institute, Cheongju, Chungbuk, 28119, Korea
| | - Hae-Kap Cheong
- Korea Basic Science Institute, Cheongju, Chungbuk, 28119, Korea
| | - Jeong Hee Moon
- Core Research Facility & Analysis Center, KRIBB, Daejeon, 34141, Korea
| | - Ji-Joon Song
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Jungwon Hwang
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.
| | - Myung Hee Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.
| |
Collapse
|
3
|
Mogol AN, Kaminsky AZ, Dutton DJ, Madak Erdogan Z. Targeting NAD+ Metabolism: Preclinical Insights into Potential Cancer Therapy Strategies. Endocrinology 2024; 165:bqae043. [PMID: 38565429 DOI: 10.1210/endocr/bqae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 03/17/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
NAD+ is one of the most important metabolites for cellular activities, and its biosynthesis mainly occurs through the salvage pathway using the nicotinamide phosphoribosyl transferase (NAMPT) enzyme. The main nicotinamide adenine dinucleotide (NAD) consumers, poly-ADP-ribose-polymerases and sirtuins enzymes, are heavily involved in DNA repair and chromatin remodeling. Since cancer cells shift their energy production pathway, NAD levels are significantly affected. NAD's roles in cell survival led to the use of NAD depletion in cancer therapies. NAMPT inhibition (alone or in combination with other cancer therapies, including endocrine therapy and chemotherapy) results in decreased cell viability and tumor burden for many cancer types. Many NAMPT inhibitors (NAMPTi) tested before were discontinued due to toxicity; however, a novel NAMPTi, KPT-9274, is a promising, low-toxicity option currently in clinical trials.
Collapse
Affiliation(s)
- Ayça N Mogol
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Champaign, IL 61801, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Champaign, IL 61801, USA
| | - Alanna Z Kaminsky
- Food Science and Human Nutrition Department, University of Illinois Urbana-Champaign, Champaign, IL 6180161801, USA
| | - David J Dutton
- Molecular Cell Biology Department, University of Illinois Urbana-Champaign, Champaign, IL 61801, USA
| | - Zeynep Madak Erdogan
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Champaign, IL 61801, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Champaign, IL 61801, USA
- Food Science and Human Nutrition Department, University of Illinois Urbana-Champaign, Champaign, IL 6180161801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Champaign, IL 61801, USA
| |
Collapse
|
4
|
Chini CCS, Cordeiro HS, Tran NLK, Chini EN. NAD metabolism: Role in senescence regulation and aging. Aging Cell 2024; 23:e13920. [PMID: 37424179 PMCID: PMC10776128 DOI: 10.1111/acel.13920] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/11/2023] Open
Abstract
The geroscience hypothesis proposes that addressing the biology of aging could directly prevent the onset or mitigate the severity of multiple chronic diseases. Understanding the interplay between key aspects of the biological hallmarks of aging is essential in delivering the promises of the geroscience hypothesis. Notably, the nucleotide nicotinamide adenine dinucleotide (NAD) interfaces with several biological hallmarks of aging, including cellular senescence, and changes in NAD metabolism have been shown to be involved in the aging process. The relationship between NAD metabolism and cellular senescence appears to be complex. On the one hand, the accumulation of DNA damage and mitochondrial dysfunction induced by low NAD+ can promote the development of senescence. On the other hand, the low NAD+ state that occurs during aging may inhibit SASP development as this secretory phenotype and the development of cellular senescence are both highly metabolically demanding. However, to date, the impact of NAD+ metabolism on the progression of the cellular senescence phenotype has not been fully characterized. Therefore, to explore the implications of NAD metabolism and NAD replacement therapies, it is essential to consider their interactions with other hallmarks of aging, including cellular senescence. We propose that a comprehensive understanding of the interplay between NAD boosting strategies and senolytic agents is necessary to advance the field.
Collapse
Affiliation(s)
- Claudia Christiano Silva Chini
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Heidi Soares Cordeiro
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Ngan Le Kim Tran
- Center for Clinical and Translational Science and Mayo Clinic Graduate School of Biomedical SciencesMayo ClinicJacksonvilleFloridaUSA
| | - Eduardo Nunes Chini
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| |
Collapse
|
5
|
Waddell J, Khatoon R, Kristian T. Cellular and Mitochondrial NAD Homeostasis in Health and Disease. Cells 2023; 12:1329. [PMID: 37174729 PMCID: PMC10177113 DOI: 10.3390/cells12091329] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/26/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
The mitochondrion has a unique position among other cellular organelles due to its dynamic properties and symbiotic nature, which is reflected in an active exchange of metabolites and cofactors between the rest of the intracellular compartments. The mitochondrial energy metabolism is greatly dependent on nicotinamide adenine dinucleotide (NAD) as a cofactor that is essential for both the activity of respiratory and TCA cycle enzymes. The NAD level is determined by the rate of NAD synthesis, the activity of NAD-consuming enzymes, and the exchange rate between the individual subcellular compartments. In this review, we discuss the NAD synthesis pathways, the NAD degradation enzymes, and NAD subcellular localization, as well as NAD transport mechanisms with a focus on mitochondria. Finally, the effect of the pathologic depletion of mitochondrial NAD pools on mitochondrial proteins' post-translational modifications and its role in neurodegeneration will be reviewed. Understanding the physiological constraints and mechanisms of NAD maintenance and the exchange between subcellular compartments is critical given NAD's broad effects and roles in health and disease.
Collapse
Affiliation(s)
- Jaylyn Waddell
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Rehana Khatoon
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Tibor Kristian
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Veterans Affairs Maryland Health Center System, 10 North Greene Street, Baltimore, MD 21201, USA
| |
Collapse
|
6
|
He L, Xu F, Li Y, Jin H, Lo PC. Cupric-ion-promoted fabrication of oxygen-replenishing nanotherapeutics for synergistic chemo and photodynamic therapy against tumor hypoxia. Acta Biomater 2023; 162:57-71. [PMID: 36944404 DOI: 10.1016/j.actbio.2023.03.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/16/2023] [Accepted: 03/13/2023] [Indexed: 03/23/2023]
Abstract
Mixing a glutathione (GSH)-responsive carboxy zinc(II) phthalocyanine (ZnPc*) and CuSO4·5H2O in water with or without the presence of the anticancer drug SN38 resulted in the formation of self-assembled nanotherapeutics labeled as ZnPc*/Cu/SN38@NP and ZnPc*/Cu@NP, respectively. The Cu2+ ions not only promoted the self-assembly of the carboxy phthalocyanine through metal complexation, but also catalyzed the transformation of H2O2 to oxygen via a catalase-like reaction, rendering an oxygen-replenishing property to the nanosystems. Both nanosystems exhibited high stability in aqueous media, but the nanoparticles disassembled gradually in an acidic or GSH-enriched environment and inside human colorectal adenocarcinoma HT29 cells, releasing the encapsulated therapeutic components. The disassembly process together with the activation by the intracellular GSH led to relaxation of the intrinsic quenching of the nanophotosensitizers and restoration of the photoactivities of ZnPc*. Under a hypoxic condition, ZnPc*/Cu/SN38@NP could attenuate the intracellular hypoxia level and maintain the photodynamic activity due to its Cu2+-promoted oxygen-replenishing ability. The photodynamic effect of ZnPc* and the anticancer effect of SN38 worked cooperatively, causing substantial apoptotic cell death. The dual therapeutic actions could also effectively inhibit the tumor growth in HT29 tumor-bearing nude mice without initiating notable adverse effects to the mice. STATEMENT OF SIGNIFICANCE: The oxygen-dependent nature of photodynamic therapy generally reduces its efficacy against tumor hypoxia, which is a common characteristic of advanced solid tumors and usually leads to resistance toward various anticancer therapies. We report herein a facile approach to assemble a glutathione-responsive carboxy phthalocyanine-based photosensitizer and an anticancer drug in aqueous media, in which Cu(II) ions were used to promote the self-assembly through metal complexation and catalyze the conversion of H2O2 to oxygen through a catalase-like reaction, making the resulting nanoparticles possessing an oxygen-replenishing property that could promote the photodynamic effect against hypoxic cancer cells and tumors. The use of Cu(II) ions to achieve the aforementioned dual functions in the fabrication of advanced nano-photosensitizing systems has not been reported.
Collapse
Affiliation(s)
- Lin He
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Feijie Xu
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Yongxin Li
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Honglin Jin
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Pui-Chi Lo
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China.
| |
Collapse
|
7
|
Mills CM, Benton TZ, Piña I, Francis MJ, Reyes L, Dolloff NG, Peterson YK, Woster PM. Stimulation of natural killer cells with small molecule inhibitors of CD38 for the treatment of neuroblastoma. Chem Sci 2023; 14:2168-2182. [PMID: 36845935 PMCID: PMC9945084 DOI: 10.1039/d2sc05749b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/28/2023] [Indexed: 02/02/2023] Open
Abstract
High-risk neuroblastoma (NB) accounts for 15% of all pediatric cancer deaths. Refractory disease for high-risk NB patients is attributed to chemotherapy resistance and immunotherapy failure. The poor prognosis for high-risk NB patients demonstrates an unmet medical need for the development of new, more efficacious therapeutics. CD38 is an immunomodulating protein that is expressed constitutively on natural killer (NK) cells and other immune cells in the tumor microenvironment (TME). Furthermore, CD38 over expression is implicated in propagating an immunosuppressive milieu within the TME. Through virtual and physical screening, we have identified drug-like small molecule inhibitors of CD38 with low micromolar IC50 values. We have begun to explore structure activity relationships for CD38 inhibition through derivatization of our most effective hit molecule to develop a new compound with lead-like physicochemical properties and improved potency. We have demonstrated that our derivatized inhibitor, compound 2, elicits immunomodulatory effects in NK cells by increasing cell viability by 190 ± 36% in multiple donors and by significantly increasing interferon gamma. Additionally, we have illustrated that NK cells exhibited enhanced cytotoxicity toward NB cells (14% reduction of NB cells over 90 minutes) when given a combination treatment of our inhibitor and the immunocytokine ch14.18-IL2. Herein we describe the synthesis and biological evaluation of small molecule CD38 inhibitors and demonstrate their potential utility as a novel approach to NB immunotherapy. These compounds represent the first examples of small molecules that stimulate immune function for the treatment of cancer.
Collapse
Affiliation(s)
- Catherine M Mills
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Thomas Z Benton
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Ivett Piña
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Megan J Francis
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Leticia Reyes
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Nathan G Dolloff
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Yuri K Peterson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Patrick M Woster
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| |
Collapse
|
8
|
The Role of CD38 in the Pathogenesis of Cardiorenal Metabolic Disease and Aging, an Approach from Basic Research. Cells 2023; 12:cells12040595. [PMID: 36831262 PMCID: PMC9954496 DOI: 10.3390/cells12040595] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Aging is a major risk factor for the leading causes of mortality, and the incidence of age-related diseases including cardiovascular disease, kidney disease and metabolic disease increases with age. NAD+ is a classic coenzyme that exists in all species, and that plays a crucial role in oxidation-reduction reactions. It is also involved in the regulation of many cellular functions including inflammation, oxidative stress and differentiation. NAD+ declines with aging in various organs, and the reduction in NAD+ is possibly involved in the development of age-related cellular dysfunction in cardiorenal metabolic organs through the accumulation of inflammation and oxidative stress. Levels of NAD+ are regulated by the balance between its synthesis and degradation. CD38 is the main NAD+-degrading enzyme, and CD38 is activated in response to inflammation with aging, which is associated with the reduction in NAD+ levels. In this review, focusing on CD38, we discuss the role of CD38 in aging and the pathogenesis of age-related diseases, including cardiorenal metabolic disease.
Collapse
|
9
|
Gao L, Du X, Li J, Qin FXF. Evolving roles of CD38 metabolism in solid tumour microenvironment. Br J Cancer 2023; 128:492-504. [PMID: 36396822 PMCID: PMC9938187 DOI: 10.1038/s41416-022-02052-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 11/19/2022] Open
Abstract
Given that plenty of clinical findings and reviews have already explained in detail on the progression of CD38 in multiple myeloma and haematological system tumours, here we no longer give unnecessary discussion on the above progression. Though therapeutic antibodies have been regarded as a greatest breakthrough in multiple myeloma immunotherapies due to the durable anti-tumour responses in the clinic, but the role of CD38 in the immunologic regulation and evasion of non-hematopoietic solid tumours are just initiated and controversial. Therefore, we will focus on the bio-function of CD38 enzymatic substrates or metabolites in the variety of non-hematopoietic malignancies and the potential therapeutic value of targeting the CD38-NAD+ or CD38-cADPR/ADPR signal axis. Though limited, we review some ongoing researches and clinical trials on therapeutic approaches in solid tumour as well.
Collapse
Affiliation(s)
- Long Gao
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China
| | - Xiaohong Du
- Institute of Clinical Medicine Research, Suzhou Science and Technology Town Hospital, Suzhou, China
| | - Jiabin Li
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China.
| | - F Xiao-Feng Qin
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 100005, Beijing, China.
- Suzhou Institute of Systems Medicine, 215123, Suzhou, China.
| |
Collapse
|
10
|
Nizami HL, Minor KE, Chiao YA, Light CM, Lee CF. Sexually dimorphic effects of SARM1 deletion on cardiac NAD + metabolism and function. Am J Physiol Heart Circ Physiol 2022; 323:H774-H781. [PMID: 36053750 PMCID: PMC9529255 DOI: 10.1152/ajpheart.00370.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) decline is repeatedly observed in heart disease and its risk factors. Although strategies promoting NAD+ synthesis to elevate NAD+ levels improve cardiac function, whether inhibition of NAD+ consumption can be therapeutic is less investigated. In this study, we examined the role of sterile-α and TIR motif containing 1 (SARM1) NAD+ hydrolase in mouse hearts, using global SARM1-knockout mice (KO). Cardiac function was assessed by echocardiography in male and female KO mice and wild-type (WT) controls. Hearts were collected for biochemical, histological, and molecular analyses. We found that the cardiac NAD+ pool was elevated in female KO mice, but only trended to increase in male KO mice. SARM1 deletion induced changes to a greater number of NAD+ metabolism transcripts in male mice than in female mice. Body weights, cardiac systolic and diastolic function, and geometry showed no changes in both male and female KO mice compared with WT counterparts. Male KO mice showed a small, but significant, elevation in cardiac collagen levels compared with WT counterparts, but no difference in collagen levels was detected in female mice. The increased collagen levels were associated with greater number of altered profibrotic and senescence-associated inflammatory genes in male KO mice, but not in female KO mice.NEW & NOTEWORTHY We examined the effects of SARM1 deletion on NAD+ pool, transcripts of NAD+ metabolism, and fibrotic pathway for the first time in mouse hearts. We observed the sexually dimorphic effects of SARM1 deletion. How these sex-dependent effects influence the outcomes of SARM1 deficiency in male and female mice in responses to cardiac stresses warrant further investigation. The elevation of cardiac NAD+ pool by SARM1 deletion provides evidence that targeting SARM1 may reverse disease-related NAD+ decline.
Collapse
Affiliation(s)
- Hina Lateef Nizami
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Keaton E Minor
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Ying Ann Chiao
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Christine M Light
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Chi Fung Lee
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
11
|
Targeting CD38 in Neoplasms and Non-Cancer Diseases. Cancers (Basel) 2022; 14:cancers14174169. [PMID: 36077708 PMCID: PMC9454480 DOI: 10.3390/cancers14174169] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/21/2022] [Accepted: 08/25/2022] [Indexed: 01/12/2023] Open
Abstract
Simple Summary CD38 remains an interesting target for anticancer therapy. Its relatively high abundance in neoplasms and crucial impact on NAD+/cADPR metabolism and the activity of T cells allows for changing the immune response in autoimmune diseases, neoplasms, and finally the induction of cell death. Antibody-dependent cell cytotoxicity is responsible for cell death induced by targeting the tumor with anti-CD38 antibodies, such as daratumumab. A wide range of laboratory experiments and clinical trials show an especially promising role of anti-CD38 therapy against multiple myeloma, NK cell lymphomas, and CD19- B-cell malignancies. More studies are required to include more diseases in the therapeutic protocols involving the modulation of CD38 activity. Abstract CD38 is a myeloid antigen present both on the cell membrane and in the intracellular compartment of the cell. Its occurrence is often enhanced in cancer cells, thus making it a potential target in anticancer therapy. Daratumumab and isatuximab already received FDA approval, and novel agents such as MOR202, TAK079 and TNB-738 undergo clinical trials. Also, novel therapeutics such as SAR442085 aim to outrank the older antibodies against CD38. Multiple myeloma and immunoglobulin light-chain amyloidosis may be effectively treated with anti-CD38 immunotherapy. Its role in other hematological malignancies is also important concerning both diagnostic process and potential treatment in the future. Aside from the hematological malignancies, CD38 remains a potential target in gastrointestinal, neurological and pulmonary system disorders. Due to the strong interaction of CD38 with TCR and CD16 on T cells, it may also serve as the biomarker in transplant rejection in renal transplant patients. Besides, CD38 finds its role outside oncology in systemic lupus erythematosus and collagen-induced arthritis. CD38 plays an important role in viral infections, including AIDS and COVID-19. Most of the undergoing clinical trials focus on the use of anti-CD38 antibodies in the therapy of multiple myeloma, CD19- B-cell malignancies, and NK cell lymphomas. This review focuses on targeting CD38 in cancer and non-cancerous diseases using antibodies, cell-based therapies and CD38 inhibitors. We also provide a summary of current clinical trials targeting CD38.
Collapse
|
12
|
CD38: An important regulator of T cell function. Biomed Pharmacother 2022; 153:113395. [PMID: 35834988 DOI: 10.1016/j.biopha.2022.113395] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 11/23/2022] Open
Abstract
Cluster of differentiation 38 (CD38) is a multifunctional extracellular enzyme on the cell surface with NADase and cyclase activities. CD38 is not only expressed in human immune cells, such as lymphocytes and plasma cells, but also is abnormally expressed in a variety of tumor cells, which is closely related to the occurrence and development of tumors. T cells are one of the important immune cells in the body. As NAD consuming enzymes, CD38, ART2, SIRT1 and PARP1 are closely related to the number and function of T cells. CD38 may also influence the activity of ART2, SIRT1 and PARP1 through the CD38-NAD+ axis to indirectly affect the number and function of T cells. Thus, CD38-NAD+ axis has a profound effect on T cell activity. In this paper, we reviewed the role and mechanism of CD38+ CD4+ T cells / CD38+ CD8+ T cells in cellular immunity and the effects of the CD38-NAD+ axis on T cell activity. We also summarized the relationship between the CD38 expression level on T cell surface and disease prediction and prognosis, the effects of anti-CD38 monoclonal antibodies on T cell activity and function, and the role of anti-CD38 chimeric antigen receptor (CAR) T cell therapy in tumor immunity. This will provide an important theoretical basis for a comprehensive understanding of the relationship between CD38 and T cells.
Collapse
|
13
|
Novak Kujundžić R. COVID-19: Are We Facing Secondary Pellagra Which Cannot Simply Be Cured by Vitamin B3? Int J Mol Sci 2022; 23:ijms23084309. [PMID: 35457123 PMCID: PMC9032523 DOI: 10.3390/ijms23084309] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 02/07/2023] Open
Abstract
Immune response to SARS-CoV-2 and ensuing inflammation pose a huge challenge to the host’s nicotinamide adenine dinucleotide (NAD+) metabolism. Humans depend on vitamin B3 for biosynthesis of NAD+, indispensable for many metabolic and NAD+-consuming signaling reactions. The balance between its utilization and resynthesis is vitally important. Many extra-pulmonary symptoms of COVID-19 strikingly resemble those of pellagra, vitamin B3 deficiency (e.g., diarrhoea, dermatitis, oral cavity and tongue manifestations, loss of smell and taste, mental confusion). In most developed countries, pellagra is successfully eradicated by vitamin B3 fortification programs. Thus, conceivably, it has not been suspected as a cause of COVID-19 symptoms. Here, the deregulation of the NAD+ metabolism in response to the SARS-CoV-2 infection is reviewed, with special emphasis on the differences in the NAD+ biosynthetic pathway’s efficiency in conditions predisposing for the development of serious COVID-19. SARS-CoV-2 infection-induced NAD+ depletion and the elevated levels of its metabolites contribute to the development of a systemic disease. Acute liberation of nicotinamide (NAM) in antiviral NAD+-consuming reactions potentiates “NAM drain”, cooperatively mediated by nicotinamide N-methyltransferase and aldehyde oxidase. “NAM drain” compromises the NAD+ salvage pathway’s fail-safe function. The robustness of the host’s NAD+ salvage pathway, prior to the SARS-CoV-2 infection, is an important determinant of COVID-19 severity and persistence of certain symptoms upon resolution of infection.
Collapse
Affiliation(s)
- Renata Novak Kujundžić
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
14
|
Zeidler JD, Hogan KA, Agorrody G, Peclat TR, Kashyap S, Kanamori KS, Gomez LS, Mazdeh DZ, Warner GM, Thompson KL, Chini CCS, Chini EN. The CD38 glycohydrolase and the NAD sink: implications for pathological conditions. Am J Physiol Cell Physiol 2022; 322:C521-C545. [PMID: 35138178 PMCID: PMC8917930 DOI: 10.1152/ajpcell.00451.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 02/07/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD) acts as a cofactor in several oxidation-reduction (redox) reactions and is a substrate for a number of nonredox enzymes. NAD is fundamental to a variety of cellular processes including energy metabolism, cell signaling, and epigenetics. NAD homeostasis appears to be of paramount importance to health span and longevity, and its dysregulation is associated with multiple diseases. NAD metabolism is dynamic and maintained by synthesis and degradation. The enzyme CD38, one of the main NAD-consuming enzymes, is a key component of NAD homeostasis. The majority of CD38 is localized in the plasma membrane with its catalytic domain facing the extracellular environment, likely for the purpose of controlling systemic levels of NAD. Several cell types express CD38, but its expression predominates on endothelial cells and immune cells capable of infiltrating organs and tissues. Here we review potential roles of CD38 in health and disease and postulate ways in which CD38 dysregulation causes changes in NAD homeostasis and contributes to the pathophysiology of multiple conditions. Indeed, in animal models the development of infectious diseases, autoimmune disorders, fibrosis, metabolic diseases, and age-associated diseases including cancer, heart disease, and neurodegeneration are associated with altered CD38 enzymatic activity. Many of these conditions are modified in CD38-deficient mice or by blocking CD38 NADase activity. In diseases in which CD38 appears to play a role, CD38-dependent NAD decline is often a common denominator of pathophysiology. Thus, understanding dysregulation of NAD homeostasis by CD38 may open new avenues for the treatment of human diseases.
Collapse
Affiliation(s)
- Julianna D Zeidler
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Kelly A Hogan
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Guillermo Agorrody
- Departamento de Fisiopatología, Hospital de Clínicas, Montevideo, Uruguay
- Laboratorio de Patologías del Metabolismo y el Envejecimiento, Instituto Pasteur de Montevideo, Montevideo, Uruguay
| | - Thais R Peclat
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Sonu Kashyap
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida
| | - Karina S Kanamori
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Lilian Sales Gomez
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Delaram Z Mazdeh
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Gina M Warner
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Katie L Thompson
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Claudia C S Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida
| | - Eduardo Nunes Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
15
|
Yan Y, Huang C, Shu Y, Wen H, Shan C, Wang X, Liu J, Li W. An HDAC8-selective fluorescent probe for imaging in living tumor cell lines and tissue slices. Org Biomol Chem 2021; 19:8352-8366. [PMID: 34528053 DOI: 10.1039/d1ob01367j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Histone deacetylase 8 (HDAC8) has been used as a therapeutic target for many cancers as it is highly expressed in neuroblastoma cells and breast cancer cells. HDAC8-selective fluorescent probes need to be urgently developed. Herein, two novel fluorescent probes, namely NP-C6-PCI and AM-C6-PCI, based on the conjugation of 1,8-naphthalimide with a highly selective inhibitor of HDAC8 (PCI-34051) were reported. Compared with PCI-34051 (KD = 6.25 × 10-5 M), NP-C6-PCI (KD = 8.05 × 10-6 M) and AM-C6-PCI (KD = 7.42 × 10-6 M) showed great selectivity toward HDAC8. Two fluorescent probes exhibited high fluorescence intensity under λex = 450 nm and a large Stokes shift (100 nm). NP-C6-PCI was selected for cell and tissue imaging due to the similarity in the bioactivity of NP-C6-PCI with PCI-34051. The ability of NP-C6-PCI to target imaging HDAC8 in SH-SY5Y and MDA-MB-231 tumor cells was demonstrated. Furthermore, NP-C6-PCI was applied to imaging SH-SY5Y tumor tissue slices to indicate the relative expression level of HDAC8. Therefore, this HDAC8-selective fluorescent probe can be expected for applications in HDAC8-targeted drug screening as well as in pathologic diagnoses.
Collapse
Affiliation(s)
- Yinyu Yan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Chaoqun Huang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Yi Shu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Hongmei Wen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Chenxiao Shan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Xinzhi Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Jian Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Wei Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| |
Collapse
|
16
|
Audrito V, Messana VG, Brandimarte L, Deaglio S. The Extracellular NADome Modulates Immune Responses. Front Immunol 2021; 12:704779. [PMID: 34421911 PMCID: PMC8371318 DOI: 10.3389/fimmu.2021.704779] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/21/2021] [Indexed: 12/30/2022] Open
Abstract
The term NADome refers to the intricate network of intracellular and extracellular enzymes that regulate the synthesis or degradation of nicotinamide adenine dinucleotide (NAD) and to the receptors that engage it. Traditionally, NAD was linked to intracellular energy production through shuffling electrons between oxidized and reduced forms. However, recent data indicate that NAD, along with its biosynthetic and degrading enzymes, has a life outside of cells, possibly linked to immuno-modulating non-enzymatic activities. Extracellular NAD can engage puriginergic receptors triggering an inflammatory response, similar - to a certain extent - to what described for adenosine triphosphate (ATP). Likewise, NAD biosynthetic and degrading enzymes have been amply reported in the extracellular space, where they possess both enzymatic and non-enzymatic functions. Modulation of these enzymes has been described in several acute and chronic conditions, including obesity, cancer, inflammatory bowel diseases and sepsis. In this review, the role of the extracellular NADome will be discussed, focusing on its proposed role in immunomodulation, together with the different strategies for its targeting and their potential therapeutic impact.
Collapse
Affiliation(s)
- Valentina Audrito
- Laboratory of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Vincenzo Gianluca Messana
- Laboratory of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Lorenzo Brandimarte
- Laboratory of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Deaglio
- Laboratory of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
17
|
Shi B, Wang W, Korman B, Kai L, Wang Q, Wei J, Bale S, Marangoni RG, Bhattacharyya S, Miller S, Xu D, Akbarpour M, Cheresh P, Proccissi D, Gursel D, Espindola-Netto JM, Chini CCS, de Oliveira GC, Gudjonsson JE, Chini EN, Varga J. Targeting CD38-dependent NAD + metabolism to mitigate multiple organ fibrosis. iScience 2020; 24:101902. [PMID: 33385109 PMCID: PMC7770554 DOI: 10.1016/j.isci.2020.101902] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/20/2020] [Accepted: 12/03/2020] [Indexed: 11/26/2022] Open
Abstract
The processes underlying synchronous multiple organ fibrosis in systemic sclerosis (SSc) remain poorly understood. Age-related pathologies are associated with organismal decline in nicotinamide adenine dinucleotide (NAD+) that is due to dysregulation of NAD+ homeostasis and involves the NADase CD38. We now show that CD38 is upregulated in patients with diffuse cutaneous SSc, and CD38 levels in the skin associate with molecular fibrosis signatures, as well as clinical fibrosis scores, while expression of key NAD+-synthesizing enzymes is unaltered. Boosting NAD+ via genetic or pharmacological CD38 targeting or NAD+ precursor supplementation protected mice from skin, lung, and peritoneal fibrosis. In mechanistic experiments, CD38 was found to reduce NAD+ levels and sirtuin activity to augment cellular fibrotic responses, while inhibiting CD38 had the opposite effect. Thus, we identify CD38 upregulation and resulting disrupted NAD+ homeostasis as a fundamental mechanism driving fibrosis in SSc, suggesting that CD38 might represent a novel therapeutic target. CD38 shows elevated expression in skin biopsies of patients with systemic sclerosis Elevated CD38 is associated with reduced NAD+ and augmented fibrotic responses Genetic loss of CD38 is associated with increased NAD+ levels and attenuated fibrosis NAD+ boosting via CD38 inhibition or NR supplementation prevents multi-organ fibrosis
Collapse
Affiliation(s)
- Bo Shi
- Northwestern Scleroderma Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Wenxia Wang
- Northwestern Scleroderma Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Benjamin Korman
- Northwestern Scleroderma Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Li Kai
- Northwestern Scleroderma Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qianqian Wang
- Northwestern Scleroderma Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jun Wei
- Northwestern Scleroderma Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Swarna Bale
- Northwestern Scleroderma Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Roberta Goncalves Marangoni
- Northwestern Scleroderma Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Swati Bhattacharyya
- Northwestern Scleroderma Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Stephen Miller
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Dan Xu
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mahzad Akbarpour
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Paul Cheresh
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Daniele Proccissi
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Demirkan Gursel
- Pathology Core Facility, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | - Claudia C S Chini
- Department of Anesthesiology and Kogod Center on Aging, Mayo Clinic, Rochester 55905 MN, USA
| | - Guilherme C de Oliveira
- Department of Anesthesiology and Kogod Center on Aging, Mayo Clinic, Rochester 55905 MN, USA
| | | | - Eduardo N Chini
- Department of Anesthesiology and Kogod Center on Aging, Mayo Clinic, Rochester 55905 MN, USA
| | - John Varga
- Northwestern Scleroderma Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
18
|
Affiliation(s)
- Shuai Wu
- Immunology, Microenvironment & Metastasis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Rugang Zhang
- Immunology, Microenvironment & Metastasis Program, The Wistar Institute, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Covarrubias AJ, Kale A, Perrone R, Lopez-Dominguez JA, Pisco AO, Kasler HG, Schmidt MS, Heckenbach I, Kwok R, Wiley CD, Wong HS, Gibbs E, Iyer SS, Basisty N, Wu Q, Kim IJ, Silva E, Vitangcol K, Shin KO, Lee YM, Riley R, Ben-Sahra I, Ott M, Schilling B, Scheibye-Knudsen M, Ishihara K, Quake SR, Newman J, Brenner C, Campisi J, Verdin E. Senescent cells promote tissue NAD + decline during ageing via the activation of CD38 + macrophages. Nat Metab 2020; 2:1265-1283. [PMID: 33199924 PMCID: PMC7908681 DOI: 10.1038/s42255-020-00305-3] [Citation(s) in RCA: 203] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/25/2020] [Indexed: 11/08/2022]
Abstract
Declining tissue nicotinamide adenine dinucleotide (NAD) levels are linked to ageing and its associated diseases. However, the mechanism for this decline is unclear. Here, we show that pro-inflammatory M1-like macrophages, but not naive or M2 macrophages, accumulate in metabolic tissues, including visceral white adipose tissue and liver, during ageing and acute responses to inflammation. These M1-like macrophages express high levels of the NAD-consuming enzyme CD38 and have enhanced CD38-dependent NADase activity, thereby reducing tissue NAD levels. We also find that senescent cells progressively accumulate in visceral white adipose tissue and liver during ageing and that inflammatory cytokines secreted by senescent cells (the senescence-associated secretory phenotype, SASP) induce macrophages to proliferate and express CD38. These results uncover a new causal link among resident tissue macrophages, cellular senescence and tissue NAD decline during ageing and offer novel therapeutic opportunities to maintain NAD levels during ageing.
Collapse
Affiliation(s)
- Anthony J Covarrubias
- Buck Institute for Research on Aging, Novato, CA, USA
- UCSF Department of Medicine, San Francisco, CA, USA
| | - Abhijit Kale
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | | | | | | | - Mark S Schmidt
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Indra Heckenbach
- Buck Institute for Research on Aging, Novato, CA, USA
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ryan Kwok
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Hoi-Shan Wong
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Eddy Gibbs
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Shankar S Iyer
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Qiuxia Wu
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Ik-Jung Kim
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Elena Silva
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Kyong-Oh Shin
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea
| | - Yong-Moon Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | | | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
| | - Melanie Ott
- Gladstone Institutes, Virology and Immunology, San Francisco, CA, USA
| | | | - Morten Scheibye-Knudsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Katsuhiko Ishihara
- Immunology and Molecular Genetics, Kawasaki Medical School, Kurashiki, Japan
| | - Stephen R Quake
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - John Newman
- Buck Institute for Research on Aging, Novato, CA, USA
- UCSF Department of Medicine, San Francisco, CA, USA
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Diabetes & Cancer Metabolism, City of Hope National Medical Center, Duarte, CA, USA
| | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA.
- UCSF Department of Medicine, San Francisco, CA, USA.
| |
Collapse
|
20
|
Dai Z, Zhang XN, Nasertorabi F, Cheng Q, Li J, Katz BB, Smbatyan G, Pei H, Louie SG, Lenz HJ, Stevens RC, Zhang Y. Synthesis of site-specific antibody-drug conjugates by ADP-ribosyl cyclases. SCIENCE ADVANCES 2020; 6:eaba6752. [PMID: 32537509 PMCID: PMC7269645 DOI: 10.1126/sciadv.aba6752] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/02/2020] [Indexed: 05/02/2023]
Abstract
Most of the current antibody-drug conjugates (ADCs) in clinic are heterogeneous mixtures. To produce homogeneous ADCs, established procedures often require multiple steps or long reaction times. The introduced mutations or foreign sequences may cause high immunogenicity. Here, we explore a new concept of transforming CD38 enzymatic activity into a facile approach for generating site-specific ADCs. This was achieved through coupling bifunctional antibody-CD38 fusion proteins with designer dinucleotide-based covalent inhibitors with stably attached payloads. The resulting adenosine diphosphate-ribosyl cyclase-enabled ADC (ARC-ADC) with a drug-to-antibody ratio of 2 could be rapidly generated through single-step conjugation. The generated ARC-ADC targeting human epidermal growth factor receptor 2 (HER2) displays excellent stability and potency against HER2-positive breast cancer both in vitro and in vivo. This proof-of-concept study demonstrates a new strategy for production of site-specific ADCs. It may provide a general approach for the development of a novel class of ADCs with potentially enhanced properties.
Collapse
Affiliation(s)
- Zhefu Dai
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Xiao-Nan Zhang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Fariborz Nasertorabi
- Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA
| | - Qinqin Cheng
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Jiawei Li
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Benjamin B. Katz
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Goar Smbatyan
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Hua Pei
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Stan G. Louie
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Raymond C. Stevens
- Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA
| | - Yong Zhang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
- Department of Chemistry, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA 90089, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
- Research Center for Liver Diseases, University of Southern California, Los Angeles, CA 90089, USA
- Corresponding author.
| |
Collapse
|
21
|
Zuo W, Liu N, Zeng Y, Liu Y, Li B, Wu K, Xiao Y, Liu Q. CD38: A Potential Therapeutic Target in Cardiovascular Disease. Cardiovasc Drugs Ther 2020; 35:815-828. [PMID: 32472237 DOI: 10.1007/s10557-020-07007-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Substantial research has demonstrated the association between cardiovascular disease and the dysregulation of intracellular calcium, ageing, reduction in nicotinamide adenine dinucleotide NAD+ content, and decrease in sirtuin activity. CD38, which comprises the soluble type, type II, and type III, is the main NADase in mammals. This molecule catalyses the production of cyclic adenosine diphosphate ribose (cADPR), nicotinic acid adenine dinucleotide phosphate (NAADP), and adenosine diphosphate ribose (ADPR), which stimulate the release of Ca2+, accompanied by NAD+ consumption and decreased sirtuin activity. Therefore, the relationship between cardiovascular disease and CD38 has been attracting increased attention. In this review, we summarize the structure, regulation, function, targeted drug development, and current research on CD38 in the cardiac context. More importantly, we provide original views about the as yet elusive mechanisms of CD38 action in certain cardiovascular disease models. Based on our review, we predict that CD38 may serve as a novel therapeutic target in cardiovascular disease in the future.
Collapse
Affiliation(s)
- Wanyun Zuo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Na Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Yunhong Zeng
- Department of Cardiology, Hunan Children's Hospital, No. 86 Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, China
| | - Yaozhong Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Biao Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Keke Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Yunbin Xiao
- Department of Cardiology, Hunan Children's Hospital, No. 86 Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, China.
| | - Qiming Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China.
| |
Collapse
|
22
|
McReynolds MR, Chellappa K, Baur JA. Age-related NAD + decline. Exp Gerontol 2020; 134:110888. [PMID: 32097708 PMCID: PMC7442590 DOI: 10.1016/j.exger.2020.110888] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/11/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential metabolite that is reported to decline in concentration in tissues of aged animals. Strategies to increase NAD+ availability have shown promise in treating many conditions in rodents, including age-related degeneration, which has in turn driven intense interest in the effects of supplements on human health. However, many aspects of NAD+ metabolism remain poorly understood, and human data are limited. Here, we discuss the state of the evidence for an age-related decline in NAD+, along with potential mechanistic explanations, including increased consumption or decreased synthesis of NAD+ and changes in the composition of cells or tissues with age. Key challenges for the field involve the development of better tools to resolve information on the NAD+ content of specific cells and subcellular compartments as well as determining the threshold levels at which NAD+ depletion triggers physiological consequences in different tissues. Understanding how NAD+ metabolism changes with age in humans may ultimately allow the design of more targeted strategies to maintain its availability, such as inhibition of key consumers in specific tissues or direct delivery of precursors to sites of deficiency. In the meantime, human clinical trials with oral supplements are poised to provide some of the first direct evidence as to whether increasing NAD+ availability can impact human physiology. Thus, it is an exciting time for NAD+ research, with much remaining to be learned in terms of both basic biology and potential therapeutic applications.
Collapse
Affiliation(s)
- Melanie R McReynolds
- Lewis-Sigler Institute for Integrative Genomics, Department of Chemistry, Princeton University, Princeton, NJ, United States of America
| | - Karthikeyani Chellappa
- Department of Physiology, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Joseph A Baur
- Department of Physiology, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America.
| |
Collapse
|
23
|
Affiliation(s)
- Yu Jiang
- Physical Sciences and Engineering Division, KAUST Catalysis Center, Polymer Synthesis Laboratory, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Nikos Hadjichristidis
- Physical Sciences and Engineering Division, KAUST Catalysis Center, Polymer Synthesis Laboratory, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| |
Collapse
|
24
|
Depaix A, Kowalska J. NAD Analogs in Aid of Chemical Biology and Medicinal Chemistry. Molecules 2019; 24:molecules24224187. [PMID: 31752261 PMCID: PMC6891637 DOI: 10.3390/molecules24224187] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/21/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) serves as an essential redox co-factor and mediator of multiple biological processes. Besides its well-established role in electron transfer reactions, NAD serves as a substrate for other biotransformations, which, at the molecular level, can be classified as protein post-translational modifications (protein deacylation, mono-, and polyADP-ribosylation) and formation of signaling molecules (e.g., cyclic ADP ribose). These biochemical reactions control many crucial biological processes, such as cellular signaling and recognition, DNA repair and epigenetic modifications, stress response, immune response, aging and senescence, and many others. However, the links between the biological effects and underlying molecular processes are often poorly understood. Moreover, NAD has recently been found to tag the 5′-ends of some cellular RNAs, but the function of these NAD-capped RNAs remains largely unrevealed. Synthetic NAD analogs are invaluable molecular tools to detect, monitor, structurally investigate, and modulate activity of NAD-related enzymes and biological processes in order to aid their deeper understanding. Here, we review the recent advances in the design and development of NAD analogs as probes for various cellular NAD-related enzymes, enzymatic inhibitors with anticancer or antimicrobial therapeutic potential, and other NAD-related chemical biology tools. We focus on research papers published within the last 10 years.
Collapse
|
25
|
Hogan KA, Chini CCS, Chini EN. The Multi-faceted Ecto-enzyme CD38: Roles in Immunomodulation, Cancer, Aging, and Metabolic Diseases. Front Immunol 2019; 10:1187. [PMID: 31214171 PMCID: PMC6555258 DOI: 10.3389/fimmu.2019.01187] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/10/2019] [Indexed: 11/23/2022] Open
Abstract
CD38 (Cluster of Differentiation 38) is a multifunctional ecto-enzyme that metabolizes NAD+ and mediates nicotinamide dinucleotide (NAD+) and extracellular nucleotide homeostasis as well as intracellular calcium. CD38 is also an emerging therapeutic target under conditions in which metabolism is altered including infection, aging, and tumorigenesis. We describe multiple enzymatic activities of CD38, which may explain the breadth of biological roles observed for this enzyme. Of greatest significance is the role of CD38 as an ecto-enzyme capable of modulating extracellular NAD+ precursor availability: 1 to bacteria unable to perform de novo synthesis of NAD+; and 2 in aged parenchyma impacted by the accumulation of immune cells during the process of ‘inflammaging’. We also discuss the paradoxical role of CD38 as a modulator of intracellular NAD+, particularly in tumor immunity. Finally, we provide a summary of therapeutic approaches to CD38 inhibition and ‘NAD+ boosting’ for treatment of metabolic dysfunction observed during aging and in tumor immunity. The present review summarizes the role of CD38 in nicotinamide nucleotide homeostasis with special emphasis on the role of CD38 as an immunomodulator and druggable target.
Collapse
Affiliation(s)
- Kelly A Hogan
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Claudia C S Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Eduardo N Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| |
Collapse
|
26
|
Zhang XN, Dai Z, Cheng Q, Zhang Y. Chemoenzymatic Preparation of 4'-Thioribose NAD .. ACTA ACUST UNITED AC 2019; 77:e83. [PMID: 30951610 DOI: 10.1002/cpnc.83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This chemoenzymatic procedure describes a strategy for the preparation of 4'-thioribose nicotinamide adenine dinucleotide (S-NAD+ ), including chemical synthesis of nicotinamide 4'-riboside (S-NR), recombinant expression and purification of two NAD+ biosynthesis enzymes nicotinamide riboside kinase (NRK) and nicotinamide mononucleotide adenylyltransferase (NMNAT), and enzymatic synthesis of S-NAD+ . The first basic protocol describes the procedures for introduction of nicotinamide onto 4'-thioribose and subsequent deprotection to generate S-NR as the key intermediate for enzymatically synthesizing S-NAD+ . In the second basic protocol, experimental methods are detailed for the production of recombinant human NRK1 and NMNAT1 to catalyze conversion of S-NR to S-NAD+ . The third basic protocol presents the enzymatic approach for the generation of S-NAD+ from S-NR precursor. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Xiao-Nan Zhang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Zhefu Dai
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Qinqin Cheng
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Yong Zhang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California.,Department of Chemistry, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California.,Research Center for Liver Diseases, University of Southern California, Los Angeles, California
| |
Collapse
|
27
|
Dai Z, Zhang XN, Nasertorabi F, Cheng Q, Pei H, Louie SG, Stevens RC, Zhang Y. Facile chemoenzymatic synthesis of a novel stable mimic of NAD . Chem Sci 2018; 9:8337-8342. [PMID: 30568770 PMCID: PMC6256357 DOI: 10.1039/c8sc03899f] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/14/2018] [Indexed: 01/01/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential cofactor participating in a variety of important enzyme-catalyzed physiological and pathophysiological processes. Analogues of NAD+ provide key and valuable agents for investigating NAD+-dependent enzymes. In this study, we report the preparation of a novel stable NAD+ mimic, 4'-thioribose NAD+ (S-NAD+), using a facile and efficient chemoenzymatic approach. Substrate activity assays indicated the resulting S-NAD+ is chemically inert to human CD38 and sirtuin 2 enzymes, but capable of participating in redox reactions in a manner similar to NAD+. X-ray crystallographic analysis revealed binding of S-NAD+ to the active site of human CD38 and critical residues involved in leaving group activation and catalysis. By more closely mimicking NAD+ in geometry and electrostatics, the generated S-NAD+ offers a unique and important tool that can be extended to study enzymes utilizing NAD+.
Collapse
Affiliation(s)
- Zhefu Dai
- Department of Pharmacology and Pharmaceutical Sciences , School of Pharmacy , University of Southern California , 1985 Zonal Ave , Los Angeles , CA 90089 , USA .
| | - Xiao-Nan Zhang
- Department of Pharmacology and Pharmaceutical Sciences , School of Pharmacy , University of Southern California , 1985 Zonal Ave , Los Angeles , CA 90089 , USA .
| | - Fariborz Nasertorabi
- Departments of Biological Sciences and Chemistry , Bridge Institute , Michelson Center for Convergent Bioscience , University of Southern California , Los Angeles , CA 90089 , USA .
| | - Qinqin Cheng
- Department of Pharmacology and Pharmaceutical Sciences , School of Pharmacy , University of Southern California , 1985 Zonal Ave , Los Angeles , CA 90089 , USA .
| | - Hua Pei
- Titus Family Department of Clinical Pharmacy , School of Pharmacy , University of Southern California , 1985 Zonal Ave , Los Angeles , CA 90089 , USA
| | - Stan G Louie
- Titus Family Department of Clinical Pharmacy , School of Pharmacy , University of Southern California , 1985 Zonal Ave , Los Angeles , CA 90089 , USA
| | - Raymond C Stevens
- Departments of Biological Sciences and Chemistry , Bridge Institute , Michelson Center for Convergent Bioscience , University of Southern California , Los Angeles , CA 90089 , USA .
| | - Yong Zhang
- Department of Pharmacology and Pharmaceutical Sciences , School of Pharmacy , University of Southern California , 1985 Zonal Ave , Los Angeles , CA 90089 , USA .
- Department of Chemistry , Dornsife College of Letters, Arts and Sciences , University of Southern California , Los Angeles , CA 90089 , USA
- Norris Comprehensive Cancer Center , University of Southern California , Los Angeles , CA 90089 , USA
- Research Center for Liver Diseases , University of Southern California , Los Angeles , CA 90089 , USA
| |
Collapse
|
28
|
Kelu JJ, Webb SE, Galione A, Miller AL. Characterization of ADP-ribosyl cyclase 1-like (ARC1-like) activity and NAADP signaling during slow muscle cell development in zebrafish embryos. Dev Biol 2018; 445:211-225. [PMID: 30447180 DOI: 10.1016/j.ydbio.2018.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/09/2018] [Accepted: 11/09/2018] [Indexed: 10/27/2022]
Abstract
We recently demonstrated the requirement of two-pore channel type 2 (TPC2)-mediated Ca2+ release during slow muscle cell differentiation and motor circuit maturation in intact zebrafish embryos. However, the upstream trigger(s) of TPC2/Ca2+ signaling during these developmental processes remains unclear. Nicotinic acid adenine dinucleotide phosphate (NAADP) is a potent Ca2+ mobilizing messenger, which is suggested to target TPC2 in mediating the release of Ca2+ from acidic vesicles. Here, we report the molecular cloning of the zebrafish ADP ribosyl cyclase (ARC) homolog (i.e., ARC1-like), which is a putative enzyme for generating NAADP. We characterized the expression of the arc1-like transcript and the NAADP levels between ~ 16 h post-fertilization (hpf) and ~ 48 hpf in whole zebrafish embryos. We showed that if ARC1-like (when fused with either EGFP or tdTomato) was overexpressed it localized in the plasma membrane, and associated with intracellular organelles, such as the acidic vesicles, Golgi complex and sarcoplasmic reticulum, in primary muscle cell cultures. Morpholino (MO)-mediated knockdown of arc1-like or pharmacological inhibition of ARC1-like (via treatment with nicotinamide), led to an attenuation of Ca2+ signaling and disruption of slow muscle cell development. In addition, the injection of arc1-like mRNA into ARC1-like morphants partially rescued the Ca2+ signals and slow muscle cell development. Together, our data might suggest a link between ARC1-like, NAADP, TPC2 and Ca2+ signaling during zebrafish myogenesis.
Collapse
Affiliation(s)
- Jeffrey J Kelu
- Division of Life Science&State Key Laboratory of Molecular Neuroscience, HKUST, Hong Kong
| | - Sarah E Webb
- Division of Life Science&State Key Laboratory of Molecular Neuroscience, HKUST, Hong Kong
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Andrew L Miller
- Division of Life Science&State Key Laboratory of Molecular Neuroscience, HKUST, Hong Kong.
| |
Collapse
|
29
|
Mottahedeh J, Haffner MC, Grogan TR, Hashimoto T, Crowell PD, Beltran H, Sboner A, Bareja R, Esopi D, Isaacs WB, Yegnasubramanian S, Rettig MB, Elashoff DA, Platz EA, De Marzo AM, Teitell MA, Goldstein AS. CD38 is methylated in prostate cancer and regulates extracellular NAD . Cancer Metab 2018; 6:13. [PMID: 30258629 PMCID: PMC6150989 DOI: 10.1186/s40170-018-0186-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 09/11/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Cancer cell metabolism requires sustained pools of intracellular nicotinamide adenine dinucleotide (NAD+) which is maintained by a balance of NAD+ hydrolase activity and NAD+ salvage activity. We recently reported that human prostate cancer can be initiated following oncogene expression in progenitor-like luminal cells marked by low expression of the NAD+-consuming enzyme CD38. CD38 expression is reduced in prostate cancer compared to benign prostate, suggesting that tumor cells may reduce CD38 expression in order to enhance pools of NAD+. However, little is known about how CD38 expression is repressed in advanced prostate cancer and whether CD38 plays a role in regulating NAD+ levels in prostate epithelial cells. METHODS CD38 expression, its association with recurrence after prostatectomy for clinically localized prostate cancer, and DNA methylation of the CD38 promoter were evaluated in human prostate tissues representing various stages of disease progression. CD38 was inducibly over-expressed in benign and malignant human prostate cell lines in order to determine the effects on cell proliferation and levels of NAD+ and NADH. NAD+ and NADH were also measured in urogenital tissues from wild-type and CD38 knockout mice. RESULTS CD38 mRNA expression was reduced in metastatic castration-resistant prostate cancer compared to localized prostate cancer. In a large cohort of men undergoing radical prostatectomy, CD38 protein expression was inversely correlated with recurrence. We identified methylation of the CD38 promoter in primary and metastatic prostate cancer. Over-expression of wild-type CD38, but not an NAD+ hydrolase-deficient mutant, depleted extracellular NAD+ levels in benign and malignant prostate cell lines. However, expression of CD38 did not significantly alter intracellular NAD+ levels in human prostate cell lines grown in vitro and in urogenital tissues isolated from wild-type and CD38 knockout mice. CONCLUSIONS CD38 protein expression in prostate cancer is associated with risk of recurrence. Methylation results suggest that CD38 is epigenetically regulated in localized and metastatic prostate cancer tissues. Our study provides support for CD38 as a regulator of extracellular, but not intracellular, NAD+ in epithelial cells. These findings suggest that repression of CD38 by methylation may serve to increase the availability of extracellular NAD+ in prostate cancer tissues.
Collapse
Affiliation(s)
- Jack Mottahedeh
- Department of Molecular, Cell & Developmental Biology, University of California Los Angeles, Los Angeles, CA USA
| | - Michael C. Haffner
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Tristan R. Grogan
- Department of Medicine Statistics Core, University of California Los Angeles, Los Angeles, CA USA
| | - Takao Hashimoto
- Department of Molecular, Cell & Developmental Biology, University of California Los Angeles, Los Angeles, CA USA
| | - Preston D. Crowell
- Molecular Biology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA USA
| | - Himisha Beltran
- Department of Medicine, Division of Medical Oncology, Weill Cornell Medicine, New York, NY USA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY USA
| | - Andrea Sboner
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY USA
| | - Rohan Bareja
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY USA
| | - David Esopi
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD USA
| | - William B. Isaacs
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD USA
- James Buchanan Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD USA
| | - Srinivasan Yegnasubramanian
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD USA
- Departments of Oncology, Pathology, and Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Matthew B. Rettig
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
- Department of Urology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA USA
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA USA
| | - David A. Elashoff
- Department of Medicine Statistics Core, University of California Los Angeles, Los Angeles, CA USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA USA
| | - Elizabeth A. Platz
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Urology and the James Buchanan Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD USA
| | - Angelo M. De Marzo
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD USA
- James Buchanan Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Michael A. Teitell
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA USA
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
- Broad Stem Cell Research Center, University of California Los Angeles, Los Angeles, CA USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA USA
| | - Andrew S. Goldstein
- Department of Molecular, Cell & Developmental Biology, University of California Los Angeles, Los Angeles, CA USA
- Department of Urology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA USA
- Broad Stem Cell Research Center, University of California Los Angeles, Los Angeles, CA USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA USA
| |
Collapse
|
30
|
Chmielewski JP, Bowlby SC, Wheeler FB, Shi L, Sui G, Davis AL, Howard TD, D'Agostino RB, Miller LD, Sirintrapun SJ, Cramer SD, Kridel SJ. CD38 Inhibits Prostate Cancer Metabolism and Proliferation by Reducing Cellular NAD + Pools. Mol Cancer Res 2018; 16:1687-1700. [PMID: 30076241 DOI: 10.1158/1541-7786.mcr-17-0526] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 06/01/2018] [Accepted: 07/13/2018] [Indexed: 12/19/2022]
Abstract
Tumor cells require increased rates of cell metabolism to generate the macromolecules necessary to sustain proliferation. They rely heavily on NAD+ as a cofactor for multiple metabolic enzymes in anabolic and catabolic reactions. NAD+ also serves as a substrate for PARPs, sirtuins, and cyclic ADP-ribose synthases. Dysregulation of the cyclic ADP-ribose synthase CD38, the main NAD'ase in cells, is reported in multiple cancer types. This study demonstrates a novel connection between CD38, modulation of NAD+, and tumor cell metabolism in prostate cancer. CD38 expression inversely correlates with prostate cancer progression. Expressing CD38 in prostate cancer cells lowered intracellular NAD+, resulting in cell-cycle arrest and expression of p21Cip1 (CDKNA1). In parallel, CD38 diminishes glycolytic and mitochondrial metabolism, activates AMP-activated protein kinase (AMPK), and inhibits fatty acid and lipid synthesis. Pharmacologic inhibition of nicotinamide phosphoribosyltransferase (NAMPT) mimicked the metabolic consequences of CD38 expression, demonstrating similarity between CD38 expression and NAMPT inhibition. Modulation of NAD+ by CD38 also induces significant differential expression of the transcriptome, producing a gene expression signature indicative of a nonproliferative phenotype. Altogether, in the context of prostate cancer, the data establish a novel role for the CD38-NAD+ axis in the regulation of cell metabolism and development.Implications: This research establishes a mechanistic connection between CD38 and metabolic control. It also provides the foundation for the translation of agents that modulate NAD+ levels in cancer cells as therapeutics. Mol Cancer Res; 16(11); 1687-700. ©2018 AACR.
Collapse
Affiliation(s)
- Jeffrey P Chmielewski
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Sarah C Bowlby
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Frances B Wheeler
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Lihong Shi
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Guangchao Sui
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Amanda L Davis
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Timothy D Howard
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ralph B D'Agostino
- Comprehensive Cancer Center at Wake Forest Baptist Medical Center, Winston-Salem, North Carolina.,Public Health Sciences-Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Lance D Miller
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina.,Comprehensive Cancer Center at Wake Forest Baptist Medical Center, Winston-Salem, North Carolina
| | - S Joseph Sirintrapun
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Scott D Cramer
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| | - Steven J Kridel
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina. .,Comprehensive Cancer Center at Wake Forest Baptist Medical Center, Winston-Salem, North Carolina
| |
Collapse
|
31
|
Chini EN, Chini CCS, Espindola Netto JM, de Oliveira GC, van Schooten W. The Pharmacology of CD38/NADase: An Emerging Target in Cancer and Diseases of Aging. Trends Pharmacol Sci 2018; 39:424-436. [PMID: 29482842 DOI: 10.1016/j.tips.2018.02.001] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/23/2018] [Accepted: 02/03/2018] [Indexed: 10/18/2022]
Abstract
Recent reports indicate that intracellular NAD levels decline in tissues during chronological aging, and that therapies aimed at increasing cellular NAD levels could have beneficial effects in many age-related diseases. The protein CD38 (cluster of differentiation 38) is a multifunctional enzyme that degrades NAD and modulates cellular NAD homeostasis. At the physiological level, CD38 has been implicated in the regulation of metabolism and in the pathogenesis of multiple conditions including aging, obesity, diabetes, heart disease, asthma, and inflammation. Interestingly, many of these functions are mediated by CD38 enzymatic activity. In addition, CD38 has also been identified as a cell-surface marker in hematologic cancers such as multiple myeloma, and a cytotoxic anti-CD38 antibody has been approved by the FDA for use in this disease. Although this is a remarkable development, killing CD38-positive tumor cells with cytotoxic anti-CD38 antibodies is only one of the potential pharmacological uses of targeting CD38. The present review discusses the biology of the CD38 enzyme and the current state of development of pharmacological tools aimed at CD38, and explores how these agents may represent a novel approach for treating human conditions including cancer, metabolic disease, and diseases of aging.
Collapse
Affiliation(s)
- Eduardo N Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | - Claudia C S Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Jair Machado Espindola Netto
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Guilherme C de Oliveira
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | |
Collapse
|
32
|
Boslett J, Hemann C, Christofi FL, Zweier JL. Characterization of CD38 in the major cell types of the heart: endothelial cells highly express CD38 with activation by hypoxia-reoxygenation triggering NAD(P)H depletion. Am J Physiol Cell Physiol 2017; 314:C297-C309. [PMID: 29187364 DOI: 10.1152/ajpcell.00139.2017] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The NAD(P)+-hydrolyzing enzyme CD38 is activated in the heart during the process of ischemia and reperfusion, triggering NAD(P)(H) depletion. However, the presence and role of CD38 in the major cell types of the heart are unknown. Therefore, we characterize the presence and function of CD38 in cardiac myocytes, endothelial cells, and fibroblasts. To comprehensively evaluate CD38 in these cells, we measured gene transcription via mRNA, as well as protein expression and enzymatic activity. Endothelial cells strongly expressed CD38, while only low expression was present in cardiac myocytes with intermediate levels in fibroblasts. In view of this high level expression in endothelial cells and the proposed role of CD38 in the pathogenesis of endothelial dysfunction, endothelial cells were subjected to hypoxia-reoxygenation to characterize the effect of this stress on CD38 expression and activity. An activity-based CD38 imaging method and CD38 activity assays were used to characterize CD38 activity in normoxic and hypoxic-reoxygenated endothelial cells, with marked CD38 activation seen following hypoxia-reoxygenation. To test the impact of hypoxia-reoxygenation-induced CD38 activation on endothelial cells, NAD(P)(H) levels and endothelial nitric oxide synthase (eNOS)-derived NO production were measured. Marked NADP(H) depletion with loss of NO and increase in superoxide production occurred following hypoxia-reoxygenation that was prevented by CD38 inhibition or knockdown. Thus, endothelial cells have high expression of CD38 which is activated by hypoxia-reoxygenation triggering CD38-mediated NADP(H) depletion with loss of eNOS-mediated NO generation and increased eNOS uncoupling. This demonstrates the importance of CD38 in the endothelium and explains the basis by which CD38 triggers post-ischemic endothelial dysfunction.
Collapse
Affiliation(s)
- James Boslett
- Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University , Columbus, Ohio
| | - Craig Hemann
- Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University , Columbus, Ohio
| | - Fedias L Christofi
- Department of Anesthesiology, Wexner Medical Center at Ohio State University, Columbus, Ohio
| | - Jay L Zweier
- Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University , Columbus, Ohio
| |
Collapse
|
33
|
Scully SS, Minden ZJ, Mukerji R, Andrianova E, Kaberna J, Lentini S, Tassa C, Wang Z, Low S, McDonnell KA. Synthesis and Evaluation of Thiazoloquinolinones with Linkers To Enable Targeting of CD38. ACS Med Chem Lett 2017; 8:196-200. [PMID: 28197311 DOI: 10.1021/acsmedchemlett.6b00409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/22/2016] [Indexed: 11/28/2022] Open
Abstract
Several monoclonal antibodies and inhibitors targeting CD38, an ectoenzyme overexpressed on malignant plasma cells, have previously been discovered. Herein, we expand structure-activity relationships of reported small-molecule thiazoloquinolinones and show that several 4-cyclohexylamino analogues have potent binding affinity for CD38 using surface plasmon resonance. Moreover, active amine analogues could be acylated and functionalized with alkyne and fluorescein groups. Fluorescein analogue 21 bound selectively to CD38 overexpressing cells, demonstrating the potential utility of thiazoloquinolinones as small-molecule conjugates for the delivery of therapeutic and imaging agents.
Collapse
Affiliation(s)
- Stephen S. Scully
- BIND Therapeutics, 325 Vassar Street, Cambridge, Massachusetts 02139, United States
| | - Zachary J. Minden
- BIND Therapeutics, 325 Vassar Street, Cambridge, Massachusetts 02139, United States
| | - Ratul Mukerji
- BIND Therapeutics, 325 Vassar Street, Cambridge, Massachusetts 02139, United States
| | - Elizaveta Andrianova
- BIND Therapeutics, 325 Vassar Street, Cambridge, Massachusetts 02139, United States
| | - James Kaberna
- BIND Therapeutics, 325 Vassar Street, Cambridge, Massachusetts 02139, United States
| | - Scott Lentini
- BIND Therapeutics, 325 Vassar Street, Cambridge, Massachusetts 02139, United States
| | - Carlos Tassa
- BIND Therapeutics, 325 Vassar Street, Cambridge, Massachusetts 02139, United States
| | - Zhaolin Wang
- BIND Therapeutics, 325 Vassar Street, Cambridge, Massachusetts 02139, United States
| | - Susan Low
- BIND Therapeutics, 325 Vassar Street, Cambridge, Massachusetts 02139, United States
| | - Kevin A. McDonnell
- BIND Therapeutics, 325 Vassar Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
34
|
Faidallah HM, Panda SS, Serrano JC, Girgis AS, Khan KA, Alamry KA, Therathanakorn T, Meyers MJ, Sverdrup FM, Eickhoff CS, Getchell SG, Katritzky AR. Synthesis, antimalarial properties and 2D-QSAR studies of novel triazole-quinine conjugates. Bioorg Med Chem 2016; 24:3527-39. [DOI: 10.1016/j.bmc.2016.05.060] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 05/23/2016] [Accepted: 05/28/2016] [Indexed: 10/24/2022]
|
35
|
Chen Y, Ren JQ, Zhang XG, Wu DY, Shen AG, Hu JM. Alkyne-Modulated Surface-Enhanced Raman Scattering-Palette for Optical Interference-Free and Multiplex Cellular Imaging. Anal Chem 2016; 88:6115-9. [DOI: 10.1021/acs.analchem.6b01374] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Yong Chen
- Key
Laboratory of Analytical Chemistry for Biology and Medicine (Ministry
of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Jia-Qiang Ren
- Key
Laboratory of Analytical Chemistry for Biology and Medicine (Ministry
of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Xia-Guang Zhang
- State
Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative
Innovation Center of Chemistry for Energy Materials, and Department
of Chemistry, and College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - De-Yin Wu
- State
Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative
Innovation Center of Chemistry for Energy Materials, and Department
of Chemistry, and College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Ai-Guo Shen
- Key
Laboratory of Analytical Chemistry for Biology and Medicine (Ministry
of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Ji-Ming Hu
- Key
Laboratory of Analytical Chemistry for Biology and Medicine (Ministry
of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
36
|
Salvador CEM, Pieber B, Neu PM, Torvisco A, Kleber Z Andrade C, Kappe CO. A sequential Ugi multicomponent/Cu-catalyzed azide-alkyne cycloaddition approach for the continuous flow generation of cyclic peptoids. J Org Chem 2015; 80:4590-602. [PMID: 25842982 DOI: 10.1021/acs.joc.5b00445] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The development of a continuous flow multistep strategy for the synthesis of linear peptoids and their subsequent macrocyclization via Click chemistry is described. The central transformation of this process is an Ugi four-component reaction generating the peptidomimetic core structure. In order to avoid exposure to the often toxic and malodorous isocyanide building blocks, the continuous approach was telescoped by the dehydration of the corresponding formamide. In a concurrent operation, the highly energetic azide moiety required for the subsequent intramolecular copper-catalyzed azide-alkyne cycloaddition (Click reaction) was installed by nucleophilic substitution from a bromide precursor. All steps yielding to the linear core structures can be conveniently coupled without the need for purification steps resulting in a single process generating the desired peptidomimetics in good to excellent yields within a 25 min reaction time. The following macrocyclization was realized in a coil reactor made of copper without any additional additive. A careful process intensification study demonstrated that this transformation occurs quantitatively within 25 min at 140 °C. Depending on the resulting ring strain, either a dimeric or a monomeric form of the cyclic product was obtained.
Collapse
Affiliation(s)
- Carlos Eduardo M Salvador
- †Institute of Chemistry, University of Graz, Heinrichstrasse 28, A-8010 Graz, Austria.,‡Laboratório de Química Metodológica e Orgânica Sintética, Instituto de Química, Universidade de Brasília, Campus Universitário Darcy Ribeiro, C.P. 4478, 70904-970, Brasília-DF, Brazil
| | - Bartholomäus Pieber
- †Institute of Chemistry, University of Graz, Heinrichstrasse 28, A-8010 Graz, Austria
| | - Philipp M Neu
- †Institute of Chemistry, University of Graz, Heinrichstrasse 28, A-8010 Graz, Austria
| | - Ana Torvisco
- §Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria
| | - Carlos Kleber Z Andrade
- ‡Laboratório de Química Metodológica e Orgânica Sintética, Instituto de Química, Universidade de Brasília, Campus Universitário Darcy Ribeiro, C.P. 4478, 70904-970, Brasília-DF, Brazil
| | - C Oliver Kappe
- †Institute of Chemistry, University of Graz, Heinrichstrasse 28, A-8010 Graz, Austria
| |
Collapse
|
37
|
Ruggieri S, Orsomando G, Sorci L, Raffaelli N. Regulation of NAD biosynthetic enzymes modulates NAD-sensing processes to shape mammalian cell physiology under varying biological cues. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1138-49. [PMID: 25770681 DOI: 10.1016/j.bbapap.2015.02.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 12/25/2022]
Abstract
In addition to its role as a redox coenzyme, NAD is a substrate of various enzymes that split the molecule to either catalyze covalent modifications of target proteins or convert NAD into biologically active metabolites. The coenzyme bioavailability may be significantly affected by these reactions, with ensuing major impact on energy metabolism, cell survival, and aging. Moreover, through the activity of the NAD-dependent deacetylating sirtuins, NAD behaves as a beacon molecule that reports the cell metabolic state, and accordingly modulates transcriptional responses and metabolic adaptations. In this view, NAD biosynthesis emerges as a highly regulated process: it enables cells to preserve NAD homeostasis in response to significant NAD-consuming events and it can be modulated by various stimuli to induce, via NAD level changes, suitable NAD-mediated metabolic responses. Here we review the current knowledge on the regulation of mammalian NAD biosynthesis, with focus on the relevant rate-limiting enzymes. This article is part of a Special Issue entitled: Cofactor-dependent proteins: evolution, chemical diversity and bio-applications.
Collapse
Affiliation(s)
- Silverio Ruggieri
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Giuseppe Orsomando
- Department of Clinical Sciences, Section of Biochemistry, Polytechnic University of Marche, Ancona, Italy
| | - Leonardo Sorci
- Department of Clinical Sciences, Section of Biochemistry, Polytechnic University of Marche, Ancona, Italy
| | - Nadia Raffaelli
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy.
| |
Collapse
|
38
|
Cabezas A, Ribeiro JM, Rodrigues JR, López-Villamizar I, Fernández A, Canales J, Pinto RM, Costas MJ, Cameselle JC. Molecular bases of catalysis and ADP-ribose preference of human Mn2+-dependent ADP-ribose/CDP-alcohol diphosphatase and conversion by mutagenesis to a preferential cyclic ADP-ribose phosphohydrolase. PLoS One 2015; 10:e0118680. [PMID: 25692488 PMCID: PMC4334965 DOI: 10.1371/journal.pone.0118680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/06/2015] [Indexed: 11/19/2022] Open
Abstract
Among metallo-dependent phosphatases, ADP-ribose/CDP-alcohol diphosphatases form a protein family (ADPRibase-Mn-like) mainly restricted, in eukaryotes, to vertebrates and plants, with preferential expression, at least in rodents, in immune cells. Rat and zebrafish ADPRibase-Mn, the only biochemically studied, are phosphohydrolases of ADP-ribose and, somewhat less efficiently, of CDP-alcohols and 2´,3´-cAMP. Furthermore, the rat but not the zebrafish enzyme displays a unique phosphohydrolytic activity on cyclic ADP-ribose. The molecular basis of such specificity is unknown. Human ADPRibase-Mn showed similar activities, including cyclic ADP-ribose phosphohydrolase, which seems thus common to mammalian ADPRibase-Mn. Substrate docking on a homology model of human ADPRibase-Mn suggested possible interactions of ADP-ribose with seven residues located, with one exception (Cys253), either within the metallo-dependent phosphatases signature (Gln27, Asn110, His111), or in unique structural regions of the ADPRibase-Mn family: s2s3 (Phe37 and Arg43) and h7h8 (Phe210), around the active site entrance. Mutants were constructed, and kinetic parameters for ADP-ribose, CDP-choline, 2´,3´-cAMP and cyclic ADP-ribose were determined. Phe37 was needed for ADP-ribose preference without catalytic effect, as indicated by the increased ADP-ribose Km and unchanged kcat of F37A-ADPRibase-Mn, while the Km values for the other substrates were little affected. Arg43 was essential for catalysis as indicated by the drastic efficiency loss shown by R43A-ADPRibase-Mn. Unexpectedly, Cys253 was hindering for cADPR phosphohydrolase, as indicated by the specific tenfold gain of efficiency of C253A-ADPRibase-Mn with cyclic ADP-ribose. This allowed the design of a triple mutant (F37A+L196F+C253A) for which cyclic ADP-ribose was the best substrate, with a catalytic efficiency of 3.5´104 M-1s-1 versus 4´103 M-1s-1 of the wild type.
Collapse
Affiliation(s)
- Alicia Cabezas
- Grupo de Enzimología, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - João Meireles Ribeiro
- Grupo de Enzimología, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - Joaquim Rui Rodrigues
- Escola Superior de Tecnologia e Gestão, Instituto Politécnico de Leiria, Leiria, Portugal
| | - Iralis López-Villamizar
- Grupo de Enzimología, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - Ascensión Fernández
- Grupo de Enzimología, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - José Canales
- Grupo de Enzimología, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - Rosa María Pinto
- Grupo de Enzimología, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - María Jesús Costas
- Grupo de Enzimología, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - José Carlos Cameselle
- Grupo de Enzimología, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
- * E-mail:
| |
Collapse
|
39
|
Wang S, Zhu W, Wang X, Li J, Zhang K, Zhang L, Zhao YJ, Lee HC, Zhang L. Design, synthesis and SAR studies of NAD analogues as potent inhibitors towards CD38 NADase. Molecules 2014; 19:15754-67. [PMID: 25268725 PMCID: PMC6271716 DOI: 10.3390/molecules191015754] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/22/2014] [Accepted: 09/22/2014] [Indexed: 11/21/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD), one of the most important coenzymes in the cells, is a substrate of the signaling enzyme CD38, by which NAD is converted to a second messenger, cyclic ADP-ribose, which releases calcium from intracellular calcium stores. Starting with 2′-deoxy-2′-fluoroarabinosyl-β-nicotinamide adenine dinucleotide (ara-F NAD), a series of NAD analogues were synthesized and their activities to inhibit CD38 NAD glycohydrolase (NADase) were evaluated. The adenosine-modified analogues showed potent inhibitory activities, among which 2′-deoxy-2′-fluoroarabinosyl-β-nicotinamideguanine dinucleotide (ara-F NGD) was the most effective one. The structure-activity relationship of NAD analogues was also discussed.
Collapse
Affiliation(s)
- Shengjun Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Wenjie Zhu
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518052, China.
| | - Xuan Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Jianguo Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Kehui Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Yong-Juan Zhao
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518052, China.
| | - Hon Cheung Lee
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518052, China.
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|