1
|
Suh J, Lee YS. The multifaceted roles of mitochondria in osteoblasts: from energy production to mitochondrial-derived vesicle secretion. J Bone Miner Res 2024; 39:1205-1214. [PMID: 38907370 PMCID: PMC11371665 DOI: 10.1093/jbmr/zjae088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/03/2024] [Indexed: 06/24/2024]
Abstract
Mitochondria in osteoblasts have been demonstrated to play multiple crucial functions in bone formation from intracellular adenosine triphosphate production to extracellular secretion of mitochondrial components. The present review explores the current knowledge about mitochondrial biology in osteoblasts, including mitochondrial biogenesis, bioenergetics, oxidative stress generation, and dynamic changes in morphology. Special attention is given to recent findings, including mitochondrial donut formation in osteoblasts, which actively generates mitochondrial-derived vesicles (MDVs), followed by extracellular secretion of small mitochondria and MDVs. We also discuss the therapeutic effects of targeting osteoblast mitochondria, highlighting their potential applications in improving bone health.
Collapse
Affiliation(s)
- Joonho Suh
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Yun-Sil Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
2
|
Suh J, Lee YS. Mitochondria as secretory organelles and therapeutic cargos. Exp Mol Med 2024; 56:66-85. [PMID: 38172601 PMCID: PMC10834547 DOI: 10.1038/s12276-023-01141-7] [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: 07/28/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 01/05/2024] Open
Abstract
Mitochondria have been primarily considered intracellular organelles that are responsible for generating energy for cell survival. However, accumulating evidence suggests that mitochondria are secreted into the extracellular space under physiological and pathological conditions, and these secreted mitochondria play diverse roles by regulating metabolism, the immune response, or the differentiation/maturation in target cells. Furthermore, increasing amount of research shows the therapeutic effects of local or systemic administration of mitochondria in various disease models. These findings have led to growing interest in exploring mitochondria as potential therapeutic agents. Here, we discuss the emerging roles of mitochondria as extracellularly secreted organelles to shed light on their functions beyond energy production. Additionally, we provide information on therapeutic outcomes of mitochondrial transplantation in animal models of diseases and an update on ongoing clinical trials, underscoring the potential of using mitochondria as a novel therapeutic intervention.
Collapse
Affiliation(s)
- Joonho Suh
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Yun-Sil Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
3
|
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: 1.0] [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
|
4
|
Suh J, Kim NK, Shim W, Lee SH, Kim HJ, Moon E, Sesaki H, Jang JH, Kim JE, Lee YS. Mitochondrial fragmentation and donut formation enhance mitochondrial secretion to promote osteogenesis. Cell Metab 2023; 35:345-360.e7. [PMID: 36754021 DOI: 10.1016/j.cmet.2023.01.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/07/2022] [Accepted: 01/11/2023] [Indexed: 02/10/2023]
Abstract
Mitochondrial components have been abundantly detected in bone matrix, implying that they are somehow transported extracellularly to regulate osteogenesis. Here, we demonstrate that mitochondria and mitochondrial-derived vesicles (MDVs) are secreted from mature osteoblasts to promote differentiation of osteoprogenitors. We show that osteogenic induction stimulates mitochondrial fragmentation, donut formation, and secretion of mitochondria through CD38/cADPR signaling. Enhancing mitochondrial fission and donut formation through Opa1 knockdown or Fis1 overexpression increases mitochondrial secretion and accelerates osteogenesis. We also show that mitochondrial fusion promoter M1, which induces Opa1 expression, impedes osteogenesis, whereas osteoblast-specific Opa1 deletion increases bone mass. We further demonstrate that secreted mitochondria and MDVs enhance bone regeneration in vivo. Our findings suggest that mitochondrial morphology in mature osteoblasts is adapted for extracellular secretion, and secreted mitochondria and MDVs are critical promoters of osteogenesis.
Collapse
Affiliation(s)
- Joonho Suh
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Na-Kyung Kim
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Wonn Shim
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Seung-Hoon Lee
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyo-Jeong Kim
- Electron Microscopy Research Center, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Eunyoung Moon
- Electron Microscopy and Spectroscopy Team, Korea Basic Science Institute, Ochang, Cheongju, Chungbuk, Republic of Korea
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jae Hyuck Jang
- Electron Microscopy Research Center, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea; Electron Microscopy and Spectroscopy Team, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Jung-Eun Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Yun-Sil Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Ma JJ, Ying J, Wang JY, Xu TT, Xia HT, Jin HT, Xiao LW, Shang WJ, Wang WQ, Feng JY. CD38 Drives Progress of Osteoarthritis by Affecting Cartilage Homeostasis. Orthop Surg 2022; 14:946-954. [PMID: 35441488 PMCID: PMC9087467 DOI: 10.1111/os.13258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/19/2022] [Accepted: 02/18/2022] [Indexed: 12/01/2022] Open
Abstract
Objective To observe expression of CD38, a key modulator of nicotinamide dinucleotide (NAD+) metabolism in mice with knee osteoarthritis, and protective effect of CD38 inhibition during the osteoarthritis (OA) development. Method The destabilization of the medial meniscus (DMM) model was performed in mice to mimic the process of OA. Immunofluorescence of CD38 was performed to evaluate its response during the OA process. Limb bud‐derived mesenchymal cells were isolated for micromass culture. 100 nM or 1 μM CD38 inhibitor (78c) treatment for 14 days and CD38 sgRNA infection were then used to explore the effects of chondrogenic differentiation via Alcian blue staining. The expressions of chondrogenic markers were detected using RT‐PCR and Western blot. To explore the protective effect of CD38 inhibitor on cartilage degradation during OA in vivo, a CD38 inhibitor was injected into the knee joint after DMM operations. Micro‐CT analysis and Safranin O‐fast green staining were used to evaluate subchondral bone micro‐architecture changes and cartilage degeneration. Results Compared to the control group, the CD38 expression in superficial cartilage was obviously increased in DMM group (P < 0.05). During the normal chondrogenic differentiation, the extracellular matrix formed and expression of Sox9, Col2, aggrecan increased apparently while CD38 expression decreased, which could be reversed with ablation of CD38 in limb bud‐derived mesenchymal cells. Consistent with findings in vitro, CD38 blockage via CD38 inhibitor injection protected against osteosclerosis in medial subchondral bone and cartilage degeneration in DMM‐induced experimental mice. Compared to the Sham group, DMM mice showed significantly increased values of BV and BV/TV in subchondral bone (P < 0.05) and Mankin score, which could be rescued by 78c treatment (P < 0.05). Also the CD38 inhibitor contributed to homeostasis of anabolism and catabolism by upregulating Sox9, Col2, aggrecan and downregulating Runx2, Col10 and Mmp13. Conclusion This study primarily implicates CD38 as an important regulator of chondrogenic differentiation. Inhibition of CD38 demonstrated protection against cartilage degeneration, which suggests that CD38 could be a potential therapeutic target for OA.
Collapse
Affiliation(s)
- Jin-Jin Ma
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun Ying
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jin-Yu Wang
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tao-Tao Xu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Han-Ting Xia
- The Affliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Hong-Ting Jin
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu-Wei Xiao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | | | | | - Jian-Yin Feng
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
6
|
Messerer DAC, Schmidt H, Frick M, Huber-Lang M. Ion and Water Transport in Neutrophil Granulocytes and Its Impairment during Sepsis. Int J Mol Sci 2021; 22:1699. [PMID: 33567720 PMCID: PMC7914618 DOI: 10.3390/ijms22041699] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Neutrophil granulocytes are the vanguard of innate immunity in response to numerous pathogens. Their activity drives the clearance of microbe- and damage-associated molecular patterns, thereby contributing substantially to the resolution of inflammation. However, excessive stimulation during sepsis leads to cellular unresponsiveness, immunological dysfunction, bacterial expansion, and subsequent multiple organ dysfunction. During the short lifespan of neutrophils, they can become significantly activated by complement factors, cytokines, and other inflammatory mediators. Following stimulation, the cells respond with a defined (electro-)physiological pattern, including depolarization, calcium influx, and alkalization as well as with increased metabolic activity and polarization of the actin cytoskeleton. Activity of ion transport proteins and aquaporins is critical for multiple cellular functions of innate immune cells, including chemotaxis, generation of reactive oxygen species, and phagocytosis of both pathogens and tissue debris. In this review, we first describe the ion transport proteins and aquaporins involved in the neutrophil ion-water fluxes in response to chemoattractants. We then relate ion and water flux to cellular functions with a focus on danger sensing, chemotaxis, phagocytosis, and oxidative burst and approach the role of altered ion transport protein expression and activity in impaired cellular functions and cell death during systemic inflammation as in sepsis.
Collapse
Affiliation(s)
- David Alexander Christian Messerer
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, 89081 Ulm, Germany;
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, 89081 Ulm, Germany
| | - Hanna Schmidt
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany; (H.S.); (M.F.)
- Department of Pediatrics and Adolescent Medicine, University Hospital of Ulm, 89081 Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany; (H.S.); (M.F.)
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, 89081 Ulm, Germany;
| |
Collapse
|
7
|
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: 41] [Impact Index Per Article: 8.2] [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
|
8
|
Dakroub A, A. Nasser S, Younis N, Bhagani H, Al-Dhaheri Y, Pintus G, Eid AA, El-Yazbi AF, Eid AH. Visfatin: A Possible Role in Cardiovasculo-Metabolic Disorders. Cells 2020; 9:2444. [PMID: 33182523 PMCID: PMC7696687 DOI: 10.3390/cells9112444] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Visfatin/NAMPT (nicotinamide phosphoribosyltransferase) is an adipocytokine with several intriguing properties. It was first identified as pre-B-cell colony-enhancing factor but turned out to possess enzymatic functions in nicotinamide adenine dinucleotide biosynthesis, with ubiquitous expression in skeletal muscles, liver, cardiomyocytes, and brain cells. Visfatin exists in an intracellular (iNAMPT) and extracellular (eNAMPT) form. Intracellularly, visfatin/iNAMPT plays a regulatory role in NAD+ biosynthesis and thereby affects many NAD-dependent proteins such as sirtuins, PARPs, MARTs and CD38/157. Extracellularly, visfatin is associated with many hormone-like signaling pathways and activates some intracellular signaling cascades. Importantly, eNAMPT has been associated with several metabolic disorders including obesity and type 1 and 2 diabetes. In this review, a brief overview about visfatin is presented with special emphasis on its relevance to metabolic diseases. Visfatin/NAMPT appears to be a unique molecule with clinical significance with a prospective promising diagnostic, prognostic, and therapeutic applications in many cardiovasculo-metabolic disorders.
Collapse
Affiliation(s)
- Ali Dakroub
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (A.D.); (N.Y.); (H.B.); (A.F.E.-Y.)
| | - Suzanne A. Nasser
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut P.O. Box 11-5020, Lebanon;
| | - Nour Younis
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (A.D.); (N.Y.); (H.B.); (A.F.E.-Y.)
| | - Humna Bhagani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (A.D.); (N.Y.); (H.B.); (A.F.E.-Y.)
| | - Yusra Al-Dhaheri
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain P.O. Box 15551, UAE;
| | - Gianfranco Pintus
- Department of Medical Laboratory Sciences, University of Sharjah, Sharjah P.O. Box 27272, UAE;
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100 Sassari, Italy
| | - Assaad A. Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon;
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (A.D.); (N.Y.); (H.B.); (A.F.E.-Y.)
- Department of Pharmacology and Toxicology, Alexandria University, Alexandria 21521, El-Mesallah, Egypt
| | - Ali H. Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (A.D.); (N.Y.); (H.B.); (A.F.E.-Y.)
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
9
|
Chini CCS, Peclat TR, Warner GM, Kashyap S, Espindola-Netto JM, de Oliveira GC, Gomez LS, Hogan KA, Tarragó MG, Puranik AS, Agorrody G, Thompson KL, Dang K, Clarke S, Childs BG, Kanamori KS, Witte MA, Vidal P, Kirkland AL, De Cecco M, Chellappa K, McReynolds MR, Jankowski C, Tchkonia T, Kirkland JL, Sedivy JM, van Deursen JM, Baker DJ, van Schooten W, Rabinowitz JD, Baur JA, Chini EN. CD38 ecto-enzyme in immune cells is induced during aging and regulates NAD + and NMN levels. Nat Metab 2020; 2:1284-1304. [PMID: 33199925 PMCID: PMC8752031 DOI: 10.1038/s42255-020-00298-z] [Citation(s) in RCA: 206] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 09/10/2020] [Indexed: 11/14/2022]
Abstract
Decreased NAD+ levels have been shown to contribute to metabolic dysfunction during aging. NAD+ decline can be partially prevented by knockout of the enzyme CD38. However, it is not known how CD38 is regulated during aging, and how its ecto-enzymatic activity impacts NAD+ homeostasis. Here we show that an increase in CD38 in white adipose tissue (WAT) and the liver during aging is mediated by accumulation of CD38+ immune cells. Inflammation increases CD38 and decreases NAD+. In addition, senescent cells and their secreted signals promote accumulation of CD38+ cells in WAT, and ablation of senescent cells or their secretory phenotype decreases CD38, partially reversing NAD+ decline. Finally, blocking the ecto-enzymatic activity of CD38 can increase NAD+ through a nicotinamide mononucleotide (NMN)-dependent process. Our findings demonstrate that senescence-induced inflammation promotes accumulation of CD38 in immune cells that, through its ecto-enzymatic activity, decreases levels of NMN and NAD+.
Collapse
Affiliation(s)
- 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, USA
| | - Thais R Peclat
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Gina M Warner
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Sonu Kashyap
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, 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, 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, USA
| | - Lilian S Gomez
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Kelly A Hogan
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Mariana G Tarragó
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Amrutesh S Puranik
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
- Division of Rheumatology, Department of Medicine, NYU Langone Health, New York, NY, USA
| | - Guillermo Agorrody
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Katie L Thompson
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | | | - Bennett G Childs
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Karina S Kanamori
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Micaela A Witte
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Paola Vidal
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Anna L Kirkland
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Marco De Cecco
- Center on the Biology of Aging and Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
- Astellas Institute for Regenerative Medicine, Marlborough, MA, USA
| | - Karthikeyani Chellappa
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Melanie R McReynolds
- Lewis-Sigler Institute for Integrative Genomics, Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Connor Jankowski
- Lewis-Sigler Institute for Integrative Genomics, Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - John M Sedivy
- Center on the Biology of Aging and Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Jan M van Deursen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Darren J Baker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | | | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - 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, USA.
| |
Collapse
|
10
|
Ogura Y, Kitada M, Xu J, Monno I, Koya D. CD38 inhibition by apigenin ameliorates mitochondrial oxidative stress through restoration of the intracellular NAD +/NADH ratio and Sirt3 activity in renal tubular cells in diabetic rats. Aging (Albany NY) 2020; 12:11325-11336. [PMID: 32507768 PMCID: PMC7343471 DOI: 10.18632/aging.103410] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/01/2020] [Indexed: 12/18/2022]
Abstract
Mitochondrial oxidative stress is a significant contributor to the pathogenesis of diabetic kidney disease (DKD). We previously showed that mitochondrial oxidative stress in the kidneys of Zucker diabetic fatty rats is associated with a decreased intracellular NAD+/NADH ratio and NAD+-dependent deacetylase Sirt3 activity, and increased expression of the NAD+-degrading enzyme CD38. In this study, we used a CD38 inhibitor, apigenin, to investigate the role of CD38 in DKD. Apigenin significantly reduced renal injuries, including tubulointerstitial fibrosis, tubular cell damage, and pro-inflammatory gene expression in diabetic rats. In addition, apigenin down-regulated CD38 expression, and increased the intracellular NAD+/NADH ratio and Sirt3-mediated mitochondrial antioxidative enzyme activity in the kidneys of diabetic rats. In vitro, inhibition of CD38 activity by apigenin or CD38 knockdown increased the NAD+/NADH ratio and Sirt3 activity in renal proximal tubular HK-2 cells cultured under high-glucose conditions. Together, these results demonstrate that by inhibiting the Sirt3 activity and increasing mitochondrial oxidative stress in renal tubular cells, CD38 plays a crucial role in the pathogenesis of DKD.
Collapse
Affiliation(s)
- Yoshio Ogura
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Ishikawa, Japan
| | - Munehiro Kitada
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Ishikawa, Japan
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Jing Xu
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Ishikawa, Japan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Itaru Monno
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Ishikawa, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Ishikawa, Japan
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| |
Collapse
|
11
|
CD38 Expression by Myeloma Cells and Its Role in the Context of Bone Marrow Microenvironment: Modulation by Therapeutic Agents. Cells 2019; 8:cells8121632. [PMID: 31847204 PMCID: PMC6952797 DOI: 10.3390/cells8121632] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/28/2019] [Accepted: 12/11/2019] [Indexed: 01/18/2023] Open
Abstract
In the last decades CD38 has emerged as an attractive target for multiple myeloma (MM). CD38 is a novel multifunctional glycoprotein that acts as a receptor, adhesion molecule interacting with CD31 and as an ectoenzyme. As an ectoenzyme, CD38 functions as a metabolic sensor catalyzing the extracellular conversion of NAD+ to the immunosuppressive factor adenosine (ADO). Other ectoenzymes, CD73 and CD203a, together with CD38, are also involved in the alternative axis of extracellular production of ADO, bypassing the canonical pathway mediated by CD39. CD38 is ubiquitously expressed in the bone marrow microenvironment; however, only MM cells display a very high surface density, which lead to the development of several anti-CD38 monoclonal antibodies (mAbs). The efficacy of anti-CD38 mAbs depends from the presence of CD38 on the surface of MM and immune-microenvironment cells. Interestingly, it has been reported that several drugs like lenalidomide, panobinostat, the all-trans retinoic acid and the DNA methyltransferase inhibitors may increase the expression of CD38. Hence, the possibility to modulate CD38 by increasing its expression on MM cells is the pre-requisite to potentiate the clinical efficacy of the anti-CD38 mAbs and to design clinical trials with the combination of anti-CD38 mAbs and these drugs.
Collapse
|
12
|
NAD binding by human CD38 analyzed by Trp189 fluorescence. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:1189-1196. [PMID: 30472140 DOI: 10.1016/j.bbamcr.2018.11.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 11/23/2022]
Abstract
The NAD-glycohydrolase/ADP-ribosyl cyclase CD38 catalyzes the metabolism of nicotinamide adenine dinucleotide (NAD) to the Ca2+ mobilizing second messengers ADP-ribose (ADPR), 2'-deoxy-ADPR, and cyclic ADP-ribose (cADPR). In the present study, we investigated binding and metabolism of NAD by a soluble fragment of human CD38, sCD38, and its catalytically inactive mutant by monitoring changes in endogenous tryptophan (Trp) fluorescence. Addition of NAD resulted in a concentration-dependent decrease in sCD38 fluorescence that is mainly caused by the Trp residue W189. Amplitude of the fluorescence decrease was fitted as one-site binding curve revealing a dissociation constant for NAD of 29 μM. A comparable dissociation constant was found with the catalytically inactive sCD38 mutant (KD 37 μM NAD) indicating that binding of NAD is not significantly affected by the mutation. The NAD-induced decrease in Trp fluorescence completely recovered in case of sCD38. Kinetics of recovery was slowed down with decreasing temperature and sCD38 concentration and increasing NAD concentration demonstrating that recovery in fluorescence is proportional to the enzymatic activity of sCD38. Accordingly, recovery in fluorescence was not observed with the catalytically inactive mutant. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
|
13
|
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: 1.1] [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
|
14
|
Ogura Y, Kitada M, Monno I, Kanasaki K, Watanabe A, Koya D. Renal mitochondrial oxidative stress is enhanced by the reduction of Sirt3 activity, in Zucker diabetic fatty rats. Redox Rep 2018; 23:153-159. [PMID: 29897845 PMCID: PMC6748695 DOI: 10.1080/13510002.2018.1487174] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Objectives: Mitochondrial oxidative stress is involved in the
pathogenesis of diabetic kidney disease. The objective of our study is to
identify the mechanisms of renal mitochondrial oxidative stress, focusing on
Sirt3, which is nicotinamide adenine dinucleotide (NAD+;
oxidized NAD)-dependent deacetylase in mitochondria. Methods: Renal mitochondrial oxidative stress and Sirt3 activity,
using Zucker diabetic fatty rats (ZDFRs) and cultured proximal tubular cells
under high-glucose condition were evaluated. Results: At 28 weeks of age, ZDFRs exhibited the increased urinary
albumin/liver-type fatty acid-binding protein
(L-FABP)/8-hydroxy-2'-deoxyguanosine (8-OHdG) excretion, histological
tubular cell damage, compared to non-diabetic Zucker Lean rats. In renal
mitochondria, acetylated isocitrate dehydrogenase2 (IDH2) and superoxide
dismutase2 (SOD2), accompanied with mitochondrial oxidative stress and
mitochondrial morphologic alterations, were increased in ZDFRs, indicating
inactivation of Sirt3. Additionally, expression of the NAD-degrading enzyme,
CD38, was increased, and the NAD+/NADH (reduced NAD) ratio was
reduced in the renal cortex of ZDFRs. High-glucose stimulation in cultured
proximal tubular cells also resulted in an increase in acetylated IDH2/SOD2,
CD38 overexpression and a reduction in the NAD+/NADH ratio. Conclusions: Enhancement of mitochondrial oxidative stress in the
diabetic kidney was mediated by the reduction of Sirt3 activity. CD38
overexpression may be related to a reduction in the NAD+/NADH
ratio in the diabetic kidney.
Collapse
Affiliation(s)
- Yoshio Ogura
- a Department of Diabetology and Endocrinology , Kanazawa Medical University , Ishikawa , Japan
| | - Munehiro Kitada
- a Department of Diabetology and Endocrinology , Kanazawa Medical University , Ishikawa , Japan.,b Division of Anticipatory Molecular Food Science and Technology , Medical Research Institute, Kanazawa Medical University , Ishikawa , Japan
| | - Itaru Monno
- a Department of Diabetology and Endocrinology , Kanazawa Medical University , Ishikawa , Japan
| | - Keizo Kanasaki
- a Department of Diabetology and Endocrinology , Kanazawa Medical University , Ishikawa , Japan.,b Division of Anticipatory Molecular Food Science and Technology , Medical Research Institute, Kanazawa Medical University , Ishikawa , Japan
| | - Ai Watanabe
- a Department of Diabetology and Endocrinology , Kanazawa Medical University , Ishikawa , Japan
| | - Daisuke Koya
- a Department of Diabetology and Endocrinology , Kanazawa Medical University , Ishikawa , Japan.,b Division of Anticipatory Molecular Food Science and Technology , Medical Research Institute, Kanazawa Medical University , Ishikawa , Japan
| |
Collapse
|
15
|
Nelissen TP, Bamford RA, Tochitani S, Akkus K, Kudzinskas A, Yokoi K, Okamoto H, Yamamoto Y, Burbach JPH, Matsuzaki H, Oguro-Ando A. CD38 is Required for Dendritic Organization in Visual Cortex and Hippocampus. Neuroscience 2018; 372:114-125. [PMID: 29306053 DOI: 10.1016/j.neuroscience.2017.12.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/15/2017] [Accepted: 12/26/2017] [Indexed: 12/26/2022]
Abstract
Morphological screening of mouse brains with known behavioral deficits can give great insight into the relationship between brain regions and their behavior. Oxytocin- and CD38-deficient mice have previously been shown to have behavioral phenotypes, such as restrictions in social memory, social interactions, and maternal behavior. CD38 is reported as an autism spectrum disorder (ASD) candidate gene and its behavioral phenotypes may be linked to ASD. To address whether these behavioral phenotypes relate to brain pathology and neuronal morphology, here we investigate the morphological changes in the CD38-deficient mice brains, with focus on the pathology and neuronal morphology of the cortex and hippocampus, using Nissl staining, immunohistochemistry, and Golgi staining. No difference was found in terms of cortical layer thickness. However, we found abnormalities in the number of neurons and neuronal morphology in the visual cortex and dentate gyrus (DG). In particular, there were arborisation differences between CD38-/- and CD38+/+ mice in the apical dendrites of the visual cortex and hippocampal CA1 pyramidal neurons. The data suggest that CD38 is implicated in appropriate development of brain regions important for social behavior.
Collapse
Affiliation(s)
- Thom P Nelissen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht and Utrecht University, Stratenum 4.205, P.O. Box 85060, 3508 AB Utrecht, The Netherlands
| | - Rosemary A Bamford
- University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, United Kingdom
| | - Shiro Tochitani
- Research Center for Child Mental Development, University of Fukui, Fukui 910-1193, Japan; Department of Radiological Technology, Faculty of Health Science, Suzaka University of Medical Science, Suzaka, Mie, Japan
| | - Kamuran Akkus
- University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, United Kingdom
| | - Aurimas Kudzinskas
- University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, United Kingdom
| | - Kenichiro Yokoi
- Research Center for Child Mental Development, University of Fukui, Fukui 910-1193, Japan
| | - Hiroshi Okamoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendei 980-8575, Japan; Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan
| | - J Peter H Burbach
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht and Utrecht University, Stratenum 4.205, P.O. Box 85060, 3508 AB Utrecht, The Netherlands
| | - Hideo Matsuzaki
- Research Center for Child Mental Development, University of Fukui, Fukui 910-1193, Japan; Department of Development of Functional Brain Activities, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Fukui 910-1193, Japan.
| | - Asami Oguro-Ando
- University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, United Kingdom.
| |
Collapse
|
16
|
Costa F, Toscani D, Chillemi A, Quarona V, Bolzoni M, Marchica V, Vescovini R, Mancini C, Martella E, Campanini N, Schifano C, Bonomini S, Accardi F, Horenstein AL, Aversa F, Malavasi F, Giuliani N. Expression of CD38 in myeloma bone niche: A rational basis for the use of anti-CD38 immunotherapy to inhibit osteoclast formation. Oncotarget 2017; 8:56598-56611. [PMID: 28915615 PMCID: PMC5593586 DOI: 10.18632/oncotarget.17896] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 04/29/2017] [Indexed: 12/20/2022] Open
Abstract
It is known that multiple myeloma (MM) cells express CD38 and that a recently developed human anti-CD38 monoclonal antibody Daratumumab mediates myeloma killing. However, the expression of CD38 and other functionally related ectoenzymes within the MM bone niche and the potential effects of Daratumumab on bone cells are still unknown. This study firstly defines by flow cytometry and immunohistochemistry the expression of CD38 by bone marrow cells in a cohort of patients with MM and indolent monoclonal gammopathies. Results indicate that only plasma cells expressed CD38 at high level within the bone niche. In addition, the flow cytometry analysis shows that CD38 was also expressed by monocytes and early osteoclast progenitors but not by osteoblasts and mature osteoclasts. Indeed, CD38 was lost during in vitro osteoclastogenesis. Consistently, we found that Daratumumab reacted with CD38 expressed on monocytes and its binding inhibited in vitro osteoclastogenesis and bone resorption activity from bone marrow total mononuclear cells of MM patients, targeting early osteoclast progenitors. The inhibitory effect was not observed from purified CD14+ cells, suggesting an indirect inhibitory effect of Daratumumab. Interestingly, all-trans retinoic acid treatment increased the inhibitory effect of Daratumumab on osteoclast formation. These observations provide a rationale for the use of an anti-CD38 antibody-based approach as treatment for multiple myeloma-induced osteoclastogenesis.
Collapse
Affiliation(s)
- Federica Costa
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Denise Toscani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Antonella Chillemi
- Laboratory of Immunogenetics, Department of Medical Sciences and CeRMS, University of Torino, Torino, Italy
| | - Valeria Quarona
- Laboratory of Immunogenetics, Department of Medical Sciences and CeRMS, University of Torino, Torino, Italy
| | - Marina Bolzoni
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Valentina Marchica
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,CoreLab, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy
| | - Rosanna Vescovini
- Clinical Medicine Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Cristina Mancini
- Pathology, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy
| | - Eugenia Martella
- Pathology, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy
| | | | - Chiara Schifano
- Hematology and BMT Center, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy
| | - Sabrina Bonomini
- Hematology and BMT Center, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy
| | - Fabrizio Accardi
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Hematology and BMT Center, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy
| | - Alberto L Horenstein
- Laboratory of Immunogenetics, Department of Medical Sciences and CeRMS, University of Torino, Torino, Italy
| | - Franco Aversa
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Hematology and BMT Center, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy
| | - Fabio Malavasi
- Laboratory of Immunogenetics, Department of Medical Sciences and CeRMS, University of Torino, Torino, Italy
| | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Hematology and BMT Center, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy.,CoreLab, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy
| |
Collapse
|
17
|
Abstract
Cardiac arrhythmias can follow disruption of the normal cellular electrophysiological processes underlying excitable activity and their tissue propagation as coherent wavefronts from the primary sinoatrial node pacemaker, through the atria, conducting structures and ventricular myocardium. These physiological events are driven by interacting, voltage-dependent, processes of activation, inactivation, and recovery in the ion channels present in cardiomyocyte membranes. Generation and conduction of these events are further modulated by intracellular Ca2+ homeostasis, and metabolic and structural change. This review describes experimental studies on murine models for known clinical arrhythmic conditions in which these mechanisms were modified by genetic, physiological, or pharmacological manipulation. These exemplars yielded molecular, physiological, and structural phenotypes often directly translatable to their corresponding clinical conditions, which could be investigated at the molecular, cellular, tissue, organ, and whole animal levels. Arrhythmogenesis could be explored during normal pacing activity, regular stimulation, following imposed extra-stimuli, or during progressively incremented steady pacing frequencies. Arrhythmic substrate was identified with temporal and spatial functional heterogeneities predisposing to reentrant excitation phenomena. These could arise from abnormalities in cardiac pacing function, tissue electrical connectivity, and cellular excitation and recovery. Triggering events during or following recovery from action potential excitation could thereby lead to sustained arrhythmia. These surface membrane processes were modified by alterations in cellular Ca2+ homeostasis and energetics, as well as cellular and tissue structural change. Study of murine systems thus offers major insights into both our understanding of normal cardiac activity and its propagation, and their relationship to mechanisms generating clinical arrhythmias.
Collapse
Affiliation(s)
- Christopher L-H Huang
- Physiological Laboratory and the Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
18
|
Salmina AB, Komleva YK, Lopatina OL, Kuvacheva NV, Gorina YV, Panina YA, Uspenskaya YA, Petrova MM, Demko IV, Zamay AS, Malinovskaya NA. Astroglial control of neuroinflammation: TLR3-mediated dsRNA-sensing pathways are in the focus. Rev Neurosci 2016; 26:143-59. [PMID: 25528762 DOI: 10.1515/revneuro-2014-0052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 10/16/2014] [Indexed: 01/06/2023]
Abstract
Neuroinflammation is as an important component of pathogenesis in many types of brain pathology. Immune mechanisms regulate neuroplasticity, memory formation, neurogenesis, behavior, brain development, cognitive functions, and brain metabolism. It is generally believed that essential homeostatic functions of astrocytes - astroglia-neuron metabolic coupling, gliovascular control, regulation of proliferation, and migration of cells in the neurogenic niches - are compromised in neuroinflammation resulting in excitotoxicity, neuronal and glial cell death, and alterations of intercellular communication. Viral neuroinfection, release of non-coding RNAs from the cells at the sites of brain injury or degeneration, and application of siRNA or RNA aptamers as therapeutic agents would require dsRNA-sensing pathways in the cells of neuronal and non-neuronal origin. In this review, we analyze the data regarding the role of astrocytes in dsRNA-initiated innate immune response in neuroinflammation and their contribution to progression of neurodegenerative and neurodevelopmental pathology.
Collapse
|
19
|
Camacho-Pereira J, Tarragó MG, Chini CCS, Nin V, Escande C, Warner GM, Puranik AS, Schoon RA, Reid JM, Galina A, Chini EN. CD38 Dictates Age-Related NAD Decline and Mitochondrial Dysfunction through an SIRT3-Dependent Mechanism. Cell Metab 2016; 23:1127-1139. [PMID: 27304511 PMCID: PMC4911708 DOI: 10.1016/j.cmet.2016.05.006] [Citation(s) in RCA: 602] [Impact Index Per Article: 66.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 02/08/2016] [Accepted: 05/22/2016] [Indexed: 11/17/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD) levels decrease during aging and are involved in age-related metabolic decline. To date, the mechanism responsible for the age-related reduction in NAD has not been elucidated. Here we demonstrate that expression and activity of the NADase CD38 increase with aging and that CD38 is required for the age-related NAD decline and mitochondrial dysfunction via a pathway mediated at least in part by regulation of SIRT3 activity. We also identified CD38 as the main enzyme involved in the degradation of the NAD precursor nicotinamide mononucleotide (NMN) in vivo, indicating that CD38 has a key role in the modulation of NAD-replacement therapy for aging and metabolic diseases.
Collapse
Affiliation(s)
- Juliana Camacho-Pereira
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Instituto de Bioquímica Médica, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ 21941-590, Brazil
| | - Mariana G Tarragó
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Claudia C S Chini
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Veronica Nin
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Carlos Escande
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Gina M Warner
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Amrutesh S Puranik
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Renee A Schoon
- Oncology Research, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Joel M Reid
- Oncology Research, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Antonio Galina
- Instituto de Bioquímica Médica, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ 21941-590, Brazil
| | - Eduardo N Chini
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| |
Collapse
|
20
|
Graeff RM, Lee HC. Determination of ADP-ribosyl cyclase activity, cyclic ADP-ribose, and nicotinic acid adenine dinucleotide phosphate in tissue extracts. Methods Mol Biol 2013; 1016:39-56. [PMID: 23681571 DOI: 10.1007/978-1-62703-441-8_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Cyclic ADP-ribose (cADPR) is a novel second messenger that releases calcium from intracellular stores. Although first shown to release calcium in the sea urchin egg, cADPR has been shown since to be active in a variety of cells and tissues, from plant to human. cADPR stimulates calcium release via ryanodine receptors although the mechanism is still not completely understood. cADPR is produced enzymatically from NAD by ADP-ribosyl cyclases; several of these proteins have been identified including one isolated from Aplysia californica, two types found in mammals (CD38 and CD157), and three forms in sea urchin. A cyclase activity has been measured in extracts from Arabidopsis thaliana although the protein is still unidentified. Nicotinic acid adenine dinucleotide phosphate (NAADP) is another novel messenger that releases calcium from internal stores and is produced by these same enzymes by an exchange reaction. NAADP targets lysosomal stores whereas cADPR releases calcium from the endoplasmic reticulum. Due to their importance in cell signaling, cADPR and NAADP have been the focus of numerous investigations over the last 25 years. This chapter describes several assay methods for the measurements of cADPR and NAADP concentration and cyclase activity in extracts from cells.
Collapse
Affiliation(s)
- Richard M Graeff
- Department of Physiology, The University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
21
|
Abstract
Cell-surface expression of CD38 in CLL has been recognised recently as a marker of progressive disease and poor outcome. In contrast to traditional staging systems, CD38 is able to identify progressive cases at an early stage. Measurement of CD38, in conjunction with other novel prognostic factors such as p53 and ZAP-70 helps to identify patients who might benefit from early and more intensive therapy. In addition, CD38 positivity can predict unmutated IgVH gene mutation status in most cases. These features, together with its easy applicability, render CD38 a valuable tool in the routine diagnostics of CLL. Questions remaining to be clarified about CD38 include the incidence and significance of its variations during the course of the disease, the optimal method to define CD38 positivity and the impact of different methodologies on results. Only after these issues are resolved can the definitive place of CD38 be defined in the diagnostics of CLL.
Collapse
Affiliation(s)
- Zoltan Matrai
- Department of Clinical Haematology, national Medical Center, Budapest, Hungary.
| |
Collapse
|
22
|
Abstract
CD38, adenosine-5'-diphosphate-ribosyl cyclase 1, is a multifunctional enzyme, expressed on a wide variety of cell types. CD38 has been assigned diverse functions, including generation of calcium-mobilizing metabolites, cell activation, and chemotaxis. Using a murine Listeria monocytogenes infection model, we found that CD38 knockout (KO) mice were highly susceptible to infection. Enhanced susceptibility was already evident within 3 days of infection, suggesting a function of CD38 in the innate immune response. CD38 was expressed on neutrophils and inflammatory monocytes, and especially inflammatory monocytes further upregulated CD38 during infection. Absence of CD38 caused alterations of the migration pattern of both cell types to sites of infection. We observed impaired accumulation of cells in the spleen but surprisingly similar or even higher accumulation of cells in the liver. CD38 KO and wild-type mice showed similar changes in the composition of neutrophils and inflammatory monocytes in blood and bone marrow, indicating that mobilization of these cells from the bone marrow was CD38 independent. In vitro, macrophages of CD38 KO mice were less efficient in uptake of listeria but still able to kill the bacteria. Dendritic cells also displayed enhanced CD38 expression following infection. However, absence of CD38 did not impair the capacity of mice to prime CD8(+) T cells against L. monocytogenes, and CD38 KO mice could efficiently control secondary listeria infection. In conclusion, our results demonstrate an essential role for CD38 in the innate immune response against L. monocytogenes.
Collapse
|
23
|
Dölle C, Rack JGM, Ziegler M. NAD and ADP-ribose metabolism in mitochondria. FEBS J 2013; 280:3530-41. [PMID: 23617329 DOI: 10.1111/febs.12304] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 04/18/2013] [Accepted: 04/23/2013] [Indexed: 12/29/2022]
Abstract
Mitochondrial metabolism is intimately connected to the universal coenzyme NAD. In addition to its role in redox reactions of energy transduction, NAD serves as substrate in regulatory reactions that lead to its degradation. Importantly, all types of the known NAD-consuming signalling reactions have been reported to take place in mitochondria. These reactions include the generation of second messengers, as well as post-translational protein modifications such as ADP-ribosylation and protein deacetylation. Therefore, the availability and redox state of NAD emerged as important factors in the regulation of mitochondrial metabolism. Molecular mechanisms and targets of mitochondrial NAD-dependent protein deacetylation and mono-ADP-ribosylation have been established, whereas poly-ADP-ribosylation and NAD-derived messenger generation in the organelles await in-depth characterization. In this review, we highlight the major NAD-dependent reactions occurring within mitochondria and describe their metabolic and regulatory functions. We also discuss the metabolic fates of the NAD-degradation products, nicotinamide and ADP-ribose, and how the mitochondrial NAD pool is restored.
Collapse
Affiliation(s)
- Christian Dölle
- Department of Molecular Biology, University of Bergen, Bergen, Norway
| | | | | |
Collapse
|
24
|
O-Uchi J, Pan S, Sheu SS. Perspectives on: SGP symposium on mitochondrial physiology and medicine: molecular identities of mitochondrial Ca2+ influx mechanism: updated passwords for accessing mitochondrial Ca2+-linked health and disease. ACTA ACUST UNITED AC 2013; 139:435-43. [PMID: 22641638 PMCID: PMC3362516 DOI: 10.1085/jgp.201210795] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Jin O-Uchi
- Department of Medicine, Center for Translational Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | |
Collapse
|
25
|
Masuda W, Jimi E. CD38/ADP-ribosyl cyclase in the rat sublingual gland: Subcellular localization under resting and saliva-secreting conditions. Arch Biochem Biophys 2011; 513:131-9. [DOI: 10.1016/j.abb.2011.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 07/06/2011] [Accepted: 07/07/2011] [Indexed: 12/01/2022]
|
26
|
Blair HC, Robinson LJ, Huang CLH, Sun L, Friedman PA, Schlesinger PH, Zaidi M. Calcium and bone disease. Biofactors 2011; 37:159-67. [PMID: 21674636 PMCID: PMC3608212 DOI: 10.1002/biof.143] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 12/18/2010] [Indexed: 11/12/2022]
Abstract
Calcium transport and calcium signaling are of basic importance in bone cells. Bone is the major store of calcium and a key regulatory organ for calcium homeostasis. Bone, in major part, responds to calcium-dependent signals from the parathyroids and via vitamin D metabolites, although bone retains direct response to extracellular calcium if parathyroid regulation is lost. Improved understanding of calcium transporters and calcium-regulated cellular processes has resulted from analysis of genetic defects, including several defects with low or high bone mass. Osteoblasts deposit calcium by mechanisms including phosphate and calcium transport with alkalinization to absorb acid created by mineral deposition; cartilage calcium mineralization occurs by passive diffusion and phosphate production. Calcium mobilization by osteoclasts is mediated by acid secretion. Both bone forming and bone resorbing cells use calcium signals as regulators of differentiation and activity. This has been studied in more detail in osteoclasts, where both osteoclast differentiation and motility are regulated by calcium.
Collapse
Affiliation(s)
- Harry C Blair
- Department of Pathology, University of Pittsburgh, Veterans Affairs Health System, PA, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Ryu SY, Beutner G, Kinnally KW, Dirksen RT, Sheu SS. Single channel characterization of the mitochondrial ryanodine receptor in heart mitoplasts. J Biol Chem 2011; 286:21324-9. [PMID: 21524998 DOI: 10.1074/jbc.c111.245597] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Heart mitochondria utilize multiple Ca(2+) transport mechanisms. Among them, the mitochondrial ryanodine receptor provides a fast Ca(2+) uptake pathway across the inner membrane to control "excitation and metabolism coupling." In the present study, we identified a novel ryanodine-sensitive channel in the native inner membrane of heart mitochondria and characterized its pharmacological and biophysical properties by directly patch clamping mitoplasts. Four distinct channel conductances of ∼100, ∼225, ∼700, and ∼1,000 picosiemens (pS) in symmetrical 150 mm CsCl were observed. The 225 pS cation-selective channel exhibited multiple subconductance states and was blocked by high concentrations of ryanodine and ruthenium red, known inhibitors of ryanodine receptors. Ryanodine exhibited a concentration-dependent modulation of this channel, with low concentrations stabilizing a subconductance state and high concentrations abolishing activity. The 100, 700, and 1,000 pS conductances exhibited different channel characteristics and were not inhibited by ryanodine. Taken together, these findings identified a novel 225 pS channel as the native mitochondrial ryanodine receptor channel activity in heart mitoplasts with biophysical and pharmacological properties that distinguish it from previously identified mitochondrial ion channels.
Collapse
Affiliation(s)
- Shin-Young Ryu
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | |
Collapse
|
28
|
Ge Y, Jiang W, Gan L, Wang L, Sun C, Ni P, Liu Y, Wu S, Gu L, Zheng W, Lund FE, Xin HB. Mouse embryonic fibroblasts from CD38 knockout mice are resistant to oxidative stresses through inhibition of reactive oxygen species production and Ca(2+) overload. Biochem Biophys Res Commun 2010; 399:167-72. [PMID: 20638362 DOI: 10.1016/j.bbrc.2010.07.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 07/13/2010] [Indexed: 02/05/2023]
Abstract
CD38 is a multifunctional enzyme that has both ADP-ribosyl cyclase and cADPR hydrolase activities, being capable of cleaving NAD(+) to cyclic ADP ribose (cADPR) and hydrolyzing cADPR to ADPR. It has been reported that there is markedly a reduction of cADPR and elevation of NAD in many tissues from CD38 knockout (CD38(-/-)) mice. Cyclic ADPR is a potent second messenger for intracellular Ca(2+) mobilization, and NAD is a key cellular metabolite for cellular energetic and a crucial regulator for multiple signaling pathways in cells. We hypothesize that CD38 knockout may have a protective effect in oxidative stresses through elevating NAD and decreasing cADPR. In the present study, we observed that the mouse embryonic fibroblasts (MEFs) from CD38(-/-) mice were significantly resistant to oxidative stress such as H(2)O(2) injury and hypoxia/reoxygenation compared with wild type MEFs (WT MEFs). We further found that production of reactive oxygen species (ROS) and concentrations of intracellular Ca(2+) ([Ca(2+)](i)) in CD38(-/-) MEFs were markedly reduced compared with WT MEFs during hypoxia/reoxygenation. Coincidence with these results, a remarkably lower mRNA level of Nox1, one of the enzymes responsible for ROS generation, was observed in CD38(-/-) MEFs. Furthermore, we found that transcription of Nox1 mRNA in WT MEFs could be elevated by calcium ionophore ionomycin in a dose-dependent manner, indicating that the expression of Nox1 mRNA can be regulated by elevation of intracellular [Ca(2+)]. Therefore we concluded that CD38(-/-) MEFs are resistant to oxidative stresses through inhibiting intracellular Ca(2+) overload and ROS production which may be regulated by Ca(2+)-mediated inhibition of Nox1 expression. Our data should provide an insight for elucidating the roles of CD38 in oxidative stresses and a novel perspective of dealing with the ischemia/reperfusion-related diseases.
Collapse
Affiliation(s)
- Yan Ge
- Laboratory of Cardiovascular Diseases, West China Hospital, The State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
The role of dietary niacin intake and the adenosine-5'-diphosphate-ribosyl cyclase enzyme CD38 in spatial learning ability: is cyclic adenosine diphosphate ribose the link between diet and behaviour? Nutr Res Rev 2009; 21:42-55. [PMID: 19079853 DOI: 10.1017/s0954422408945182] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The pyridine nucleotide NAD+ is derived from dietary niacin and serves as the substrate for the synthesis of cyclic ADP-ribose (cADPR), an intracellular Ca signalling molecule that plays an important role in synaptic plasticity in the hippocampus, a region of the brain involved in spatial learning. cADPR is formed in part via the activity of the ADP-ribosyl cyclase enzyme CD38, which is widespread throughout the brain. In the present review, current evidence of the relationship between dietary niacin and behaviour is presented following investigations of the effect of niacin deficiency, pharmacological nicotinamide supplementation and CD38 gene deletion on brain nucleotides and spatial learning ability in mice and rats. In young male rats, both niacin deficiency and nicotinamide supplementation significantly altered brain NAD+ and cADPR, both of which were inversely correlated with spatial learning ability. These results were consistent across three different models of niacin deficiency (pair feeding, partially restricted feeding and niacin recovery). Similar changes in spatial learning ability were observed in Cd38- / - mice, which also showed decreases in brain cADPR. These findings suggest an inverse relationship between spatial learning ability, dietary niacin intake and cADPR, although a direct link between cADPR and spatial learning ability is still missing. Dietary niacin may therefore play a role in the molecular events regulating learning performance, and further investigations of niacin intake, CD38 and cADPR may help identify potential molecular targets for clinical intervention to enhance learning and prevent or reverse cognitive decline.
Collapse
|
30
|
Salmina AB, Okuneva OS, Malinovskaya NA, Taranushenko TE, Morgun AV, Mantorova NS, Mikhutkina SV. NAD+-dependent mechanisms of disturbances of viability of brain cells during the acute period of hypoxic-ischemic perinatal injury. NEUROCHEM J+ 2008. [DOI: 10.1134/s1819712408030136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Blair HC, Zaidi M, Huang CLH, Sun L. The developmental basis of skeletal cell differentiation and the molecular basis of major skeletal defects. Biol Rev Camb Philos Soc 2008; 83:401-15. [PMID: 18710437 DOI: 10.1111/j.1469-185x.2008.00048.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Vertebrate skeletal differentiation retains elements from simpler phyla, and reflects the differentiation of supporting tissues programmed by primary embryonic development. This developmental scheme is driven by homeotic genes expressed in sequence, with subdivision of skeletal primordia driven by a combination of seven transmembrane-pass receptors responding to Wnt-family signals, and by bone morphogenetic family signals that define borders of individual bones. In sea-dwelling vertebrates, an essentially complete form of the skeleton adapted by the land-living vertebrates develops in cartilage, based on type II collagen and hydrophilic proteoglycans. In bony fishes, this skeleton is mineralized to form a solid bony skeleton. In the land-living vertebrates, most of the skeleton is replaced by an advanced vascular mineralized skeleton based on type I collagen, which reduces skeletal mass while facilitating use of skeletal mineral for metabolic homeostasis. Regulation of the mammalian skeleton, in this context, reflects practical adaptations to the needs for life on land that are related to ancestral developmental signals. This regulation includes central nervous system regulation that integrates bone turnover with overall metabolism. Recent work on skeletal development, in addition, demonstrates molecular mechanisms that cause developmental bone diseases.
Collapse
Affiliation(s)
- Harry C Blair
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | | | | | |
Collapse
|
32
|
Gambara G, Billington RA, Debidda M, D'Alessio A, Palombi F, Ziparo E, Genazzani AA, Filippini A. NAADP-induced Ca(2+ signaling in response to endothelin is via the receptor subtype B and requires the integrity of lipid rafts/caveolae. J Cell Physiol 2008; 216:396-404. [PMID: 18288637 DOI: 10.1002/jcp.21407] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We have investigated the role of NAADP-mediated Ca(2+) mobilization in endothelin (ET) signaling via endothelin receptor subtype A (ETA) and endothelin receptor subtype B (ETB) in rat peritubular smooth muscle cells. Microinjection and extracellular application of NAADP were both able to elicit Ca(2+) release which was blocked by inhibitory concentrations of NAADP, by impairing Ca(2+) uptake in acidic stores with bafilomycin, and by thapsigargin. Ca(2+) release in response to selective ETB stimulation was abolished by inhibition of NAADP signaling through the same strategies, while these treatments only partially impaired ETA-dependent Ca(2+) signaling, showing that transduction of the ETB signal is dependent on NAADP. In addition, we show that lipid rafts/caveolae contain ETA, ETB, and NAADP/cADPR generating enzyme CD38 and that stimulation of ETB receptors results in increased CD38 activity; interestingly, ETB- (but not ETA-) mediated Ca(2+) responses were antagonized by disruption of lipid rafts/caveolae with methyl-beta-cyclodextrin. These data demonstrate a primary role of NAADP in ETB-mediated Ca(2+) signaling and strongly suggest a novel role of lipid rafts/caveolae in triggering ET-induced NAADP signaling.
Collapse
Affiliation(s)
- Guido Gambara
- Istituto Pasteur-Fondazione Cenci Bolognetti, Department of Histology and Medical Embryology, University of Rome La Sapienza, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Moreschi I, Bruzzone S, Bodrato N, Usai C, Guida L, Nicholas RA, Kassack MU, Zocchi E, De Flora A. NAADP+ is an agonist of the human P2Y11 purinergic receptor. Cell Calcium 2007; 43:344-55. [PMID: 17707504 DOI: 10.1016/j.ceca.2007.06.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2006] [Revised: 05/08/2007] [Accepted: 06/28/2007] [Indexed: 11/30/2022]
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP+) is an intracellular second messenger releasing Ca2+ from intracellular stores in different cell types. In addition, it is also active in triggering [Ca2+](i) increase when applied extracellularly and various underlying mechanisms have been proposed. Here, we used hP2Y(11)-transfected 1321N1 astrocytoma cells to unequivocally establish whether extracellular NAADP+ is an agonist of the P2Y(11) receptor, as previously reported for beta-NAD+ [I. Moreschi, S. Bruzzone, R.A. Nicholas, et al., Extracellular NAD+ is an agonist of the human P2Y11 purinergic receptor in human granulocytes, J. Biol. Chem. 281 (2006) 31419-31429]. Extracellular NAADP+ triggered a concentration-dependent two-step elevation of [Ca2+](i) in 1321N1-hP2Y(11) cells, but not in wild-type 1321N1 cells, secondary to the intracellular production of IP(3), cAMP and cyclic ADP-ribose (cADPR). Specifically, the transient [Ca2+](i) rise proved to be related to IP(3) overproduction and to consequent Ca2+ mobilization, while the sustained [Ca2+](i) elevation was caused by the cAMP/ADP-ribosyl cyclase (ADPRC)/cADPR signalling cascade and by influx of extracellular Ca2+. In human granulocytes, endogenous P2Y(11) proved to be responsible for the NAADP+-induced cell activation (as demonstrated by the use of NF157, a selective and potent inhibitor of P2Y(11)), unveiling a role of NAADP+ as a pro-inflammatory cytokine. In conclusion, we provide unequivocal evidence for the activation of a member of the P2Y receptor subfamily by NAADP+.
Collapse
Affiliation(s)
- Iliana Moreschi
- Department of Experimental Medicine, Section of Biochemistry, and Center of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV/1, Genoa, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
The use of genetically manipulated mouse models, gene and protein discovery and the cataloguing of genetic mutations have each allowed us to obtain new insights into skeletal morphogenesis and remodeling. These techniques have made it possible to identify molecules that are obligatory for specific cellular functions, and to exploit these molecules for therapeutic purposes. New insights into the pathophysiology of diseases have also enabled us to understand molecular defects in a way that was not possible a decade ago. This review summarizes our current understanding of the carefully orchestrated cross-talk between cells of the bone marrow and between bone cells and the brain through which bone is constantly remodeled during adult life. It also highlights molecular aberrations that cause bone cells to become dysfunctional, as well as therapeutic options and opportunities to counteract skeletal loss.
Collapse
Affiliation(s)
- Mone Zaidi
- The Mount Sinai Bone Program, Department of Medicine, Box 1055, Mount Sinai School of Medicine, New York, New York 10029, USA.
| |
Collapse
|
35
|
Higashida H, Salmina AB, Olovyannikova RY, Hashii M, Yokoyama S, Koizumi K, Jin D, Liu HX, Lopatina O, Amina S, Islam MS, Huang JJ, Noda M. Cyclic ADP-ribose as a universal calcium signal molecule in the nervous system. Neurochem Int 2007; 51:192-9. [PMID: 17664018 DOI: 10.1016/j.neuint.2007.06.023] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Revised: 05/30/2007] [Accepted: 06/01/2007] [Indexed: 01/09/2023]
Abstract
beta-NAD(+) is as abundant as ATP in neuronal cells. beta-NAD(+) functions not only as a coenzyme but also as a substrate. beta-NAD(+)-utilizing enzymes are involved in signal transduction. We focus on ADP-ribosyl cyclase/CD38 which synthesizes cyclic ADP-ribose (cADPR), a universal Ca(2+) mobilizer from intracellular stores, from beta-NAD(+). cADPR acts through activation/modulation of ryanodine receptor Ca(2+) releasing Ca(2+) channels. cADPR synthesis in neuronal cells is stimulated or modulated via different pathways and various factors. Subtype-specific coupling of various neurotransmitter receptors with ADP-ribosyl cyclase confirms the involvement of the enzyme in signal transduction in neurons and glial cells. Moreover, cADPR/CD38 is critical in oxytocin release from the hypothalamic cell dendrites and nerve terminals in the posterior pituitary. Therefore, it is possible that pharmacological manipulation of intracellular cADPR levels through ADP-ribosyl cyclase activity or synthetic cADPR analogues may provide new therapeutic opportunities for treatment of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Haruhiro Higashida
- Department of Biophysical Genetics, Kanazawa University Graduate School of Medicine, Kanazawa 920-8640, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
CD38 is a novel multifunctional protein that serves not only as an antigen but also as an enzyme. It catalyzes the metabolism of cyclic ADP-ribose and nicotinic acid adenine dinucleotide phosphate, two structurally and functionally distinct Ca(2+) messengers targeting, respectively, the endoplasmic reticulum and lysosomal Ca(2+) stores. The protein has recently been crystallized and its three-dimensional structure solved to a resolution of 1.9 A. The crystal structure of a binary complex reveals critical interactions between residues at the active site and a bound substrate, providing mechanistic insights to its novel multi-functional catalysis. This article reviews the current advances in the understanding of the structural determinants that control the multiple enzymatic reactions catalyzed by CD38.
Collapse
Affiliation(s)
- Hon Cheung Lee
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
37
|
Lee HC. Structure and enzymatic functions of human CD38. MOLECULAR MEDICINE (CAMBRIDGE, MASS.) 2007; 12:317-23. [PMID: 17380198 PMCID: PMC1829193 DOI: 10.2119/2006–00086.lee] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/08/2006] [Accepted: 12/07/2006] [Indexed: 12/12/2022]
Abstract
CD38 is a novel multifunctional protein that serves not only as an antigen but also as an enzyme. It catalyzes the metabolism of cyclic ADP-ribose and nicotinic acid adenine dinucleotide phosphate, two structurally and functionally distinct Ca(2+) messengers targeting, respectively, the endoplasmic reticulum and lysosomal Ca(2+) stores. The protein has recently been crystallized and its three-dimensional structure solved to a resolution of 1.9 A. The crystal structure of a binary complex reveals critical interactions between residues at the active site and a bound substrate, providing mechanistic insights to its novel multi-functional catalysis. This article reviews the current advances in the understanding of the structural determinants that control the multiple enzymatic reactions catalyzed by CD38.
Collapse
Affiliation(s)
- Hon Cheung Lee
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
38
|
Abstract
Calcium transport and calcium signalling mechanisms in bone cells have, in many cases, been discovered by study of diseases with disordered bone metabolism. Calcium matrix deposition is driven primarily by phosphate production, and disorders in bone deposition include abnormalities in membrane phosphate transport such as in chondrocalcinosis, and defects in phosphate-producing enzymes such as in hypophosphatasia. Matrix removal is driven by acidification, which dissolves the mineral. Disorders in calcium removal from bone matrix by osteoclasts cause osteopetrosis. On the other hand, although bone is central to management of extracellular calcium, bone is not a major calcium sensing organ, although calcium sensing proteins are expressed in both osteoblasts and osteoclasts. Intracellular calcium signals are involved in secondary control including cellular motility and survival, but the relationship of these findings to specific diseases is not clear. Intracellular calcium signals may regulate the balance of cell survival versus proliferation or anabolic functional response as part of signalling cascades that integrate the response to primary signals via cell stretch, estrogen, tyrosine kinase, and tumor necrosis factor receptors.
Collapse
Affiliation(s)
- H C Blair
- Department of Pathology, University of Pittsburgh, PA 15261, USA
| | | | | | | |
Collapse
|
39
|
Sun L, Peng Y, Sharrow AC, Iqbal J, Zhang Z, Papachristou DJ, Zaidi S, Zhu LL, Yaroslavskiy BB, Zhou H, Zallone A, Sairam MR, Kumar TR, Bo W, Braun J, Cardoso-Landa L, Schaffler MB, Moonga BS, Blair HC, Zaidi M. FSH Directly Regulates Bone Mass. Cell 2006; 125:247-60. [PMID: 16630814 DOI: 10.1016/j.cell.2006.01.051] [Citation(s) in RCA: 515] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2005] [Revised: 09/02/2005] [Accepted: 01/23/2006] [Indexed: 11/29/2022]
Abstract
Postmenopausal osteoporosis, a global public health problem, has for decades been attributed solely to declining estrogen levels. Although FSH levels rise sharply in parallel, a direct effect of FSH on the skeleton has never been explored. We show that FSH is required for hypogonadal bone loss. Neither FSHbeta nor FSH receptor (FSHR) null mice have bone loss despite severe hypogonadism. Bone mass is increased and osteoclastic resorption is decreased in haploinsufficient FSHbeta+/- mice with normal ovarian function, suggesting that the skeletal action of FSH is estrogen independent. Osteoclasts and their precursors possess G(i2alpha)-coupled FSHRs that activate MEK/Erk, NF-kappaB, and Akt to result in enhanced osteoclast formation and function. We suggest that high circulating FSH causes hypogonadal bone loss.
Collapse
Affiliation(s)
- Li Sun
- Mount Sinai Bone Program, Department of Medicine and Department of Orthopedics, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sun L, Iqbal J, Zaidi S, Zhu LL, Zhang X, Peng Y, Moonga BS, Zaidi M. Structure and functional regulation of the CD38 promoter. Biochem Biophys Res Commun 2006; 341:804-9. [PMID: 16442077 DOI: 10.1016/j.bbrc.2006.01.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Accepted: 01/11/2006] [Indexed: 11/17/2022]
Abstract
CD38 has multiple roles in biology, including T lymphocyte signaling, neutrophil migration, neurotransmission, cell proliferation, apoptosis, and bone remodeling. To study the transcriptional control of the CD38 gene, we cloned a putative 1.8 kb promoter fragment from a rabbit genomic DNA library. Primer extension analysis indicated two transcription start sites consistent with the absence of a TATA box. Sequence analysis revealed several AP-1, AP-4, myo-D, GATA, and SP-1 sequences. MC3T3.E1 (osteoblast) or RAW-C3 (osteoclast precursor macrophage) cells were then transfected with the CD38 promoter or its deletion fragments ligated to the luciferase reporter gene. Except for the shortest 41 bp fragment, all fragments showed significant luciferase activity. There was a marked stimulation of basal activity in the 93 bp fragment that contained a GC box and SP-1 site. Furthermore, there were significant differences in the activity of the fragments in MC3T3.E1 and RAW-C3 cells. Intracellular Ca(2+) elevations by ionomycin (10muM) in MC3T3.E1 cells inhibited promoter activity, except in the short 41 bp. In contrast, cAMP elevation by exposure to forskolin (100 microM) inhibited activation of all fragments, except the 0.6 and 1.2kb fragments. Finally, TNF-alpha stimulated promoter activity in RAW-C3 cells transfected with the 93 bp and 1.0 kb fragments, consistent with the stimulation of CD38 mRNA by TNF-alpha. Physiologically, therefore, modulation of the expression of the NAD(+)-sensing enzyme, CD38, by Ca(2+), cAMP, and cytokines, such as TNF-alpha may contribute to coupling the intense metabolic activity of osteoclasts and osteoblasts to their respective bone-resorbing and bone-forming functions.
Collapse
Affiliation(s)
- Li Sun
- Mount Sinai Bone Program and Department of Medicine, Mount Sinai School of Medicine, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Drummond FJ, Mackrill JJ, O'sullivan K, Daly M, Shanahan F, Molloy MG. CD38 is associated with premenopausal and postmenopausal bone mineral density and postmenopausal bone loss. J Bone Miner Metab 2006; 24:28-35. [PMID: 16369895 DOI: 10.1007/s00774-005-0642-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2004] [Accepted: 08/15/2005] [Indexed: 11/25/2022]
Abstract
One goal of osteoporosis research is to identify the genes and environmental factors that contribute to low bone mineral density (BMD) and fracture. Linkage analyses have identified quantitative trait loci (QTLs), however, the genes contributing to low BMD are largely unknown. We examined the potential association of an intronic polymorphism in CD38 with BMD and postmenopausal bone loss. CD38 resides in 4p15, where a QTL for BMD has been described. CD38-/- mice display an osteoporotic phenotype at 3 months, with normalization of BMD by 5 months. The CD38 polymorphism was identified by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis in 457 postmenopausal and 173 premenopausal Caucasian women whose spine and hip BMD was measured by dual energy X-ray absorptiometry (DXA). Influence of the CD38 polymorphism on bone loss was analyzed in 273 postmenopausal women over a follow-up of 2.94 +/- 1.50 years. The CD38-PvuII polymorphism was significantly associated with premenopausal and postmenopausal (P = 0.001) lumbar spine BMD. Women homozygous for the G allele had >14% lower spinal BMD than women with GC/CC genotypes. An allele dose effect was observed at the spine in premenopausal (P = 0.002) and postmenopausal (P < 0.001) cohorts. The CD38-PvuII polymorphism was significantly associated with femoral neck BMD in pre- and postmenopausal women (P = 0.002 and P = 0.011, respectively). However, significance was lost following adjustment of hip BMD for covariates in the postmenopausal cohort (P = 0.081). The CD38-PvuII polymorphism was weakly associated with bone loss at the spine (P = 0.024), in postmenopausal women not taking hormone replacement therapy. We suggest that the CD38-PvuII polymorphism may influence the attainment and maintenance of peak BMD and postmenopausal bone loss.
Collapse
Affiliation(s)
- Frances J Drummond
- Department of Rheumatology and Medicine, Clinical Sciences Building, Cork University Hospital, National University of Ireland, Cork, Ireland.
| | | | | | | | | | | |
Collapse
|
42
|
Bréchard S, Brunello A, Bueb JL, Tschirhart EJ. Modulation by cADPr of Ca2+ mobilization and oxidative response in dimethylsulfoxide- or retinoic acid-differentiated HL-60 cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1763:129-36. [PMID: 16413069 DOI: 10.1016/j.bbamcr.2005.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Revised: 11/17/2005] [Accepted: 12/05/2005] [Indexed: 11/18/2022]
Abstract
In human phagocytic cells, reactive oxygen species (ROS) generation in response to N-formyl-L-Methionyl-L-Leucyl-L-Phenylalanine (fMLF) is largely dependent on cytosolic free calcium concentration ([Ca2+]i). Cyclic ADP-ribose (cADPr) is able to regulate Ca2+ release from intracellular stores through the ryanodine receptor but its potential role in biological responses has so far not been determined. In this study, we examined whether extracellular and intracellular cADPr is required in fMLF-induced [Ca2+]i rise and consequently in the oxidative response in human neutrophil-like HL-60 cells differentiated with dimethylsulfoxide or all-trans-retinoic acid (ATRA). We establish that extracellular cADPr cannot elicit [Ca2+]i elevation. Furthermore, we demonstrate that 8-Br-cADPr, a functional antagonist of cADPr, inhibits Ca2+ entry into HL-60 cells differentiated with ATRA and stimulated with fMLF (95+/-4 and 148+/-5 nM respectively, n=3). Finally, we show that this partial inhibition of Ca2+ mobilization is unrelated to ROS production (10.0+/-0.3 vs. 9.6+/-0.5 A.U., n=3). In conclusion, we showed that cADPr can control fMLF-induced Ca2+ influx but is unable to regulate a Ca2+-dependent biological response, i.e. H2O2 production.
Collapse
Affiliation(s)
- S Bréchard
- Laboratoire de Biologie et Physiologie Intégrée. Faculté des Sciences, de la Technologie et de la Communication. Université du Luxembourg, Luxembourg
| | | | | | | |
Collapse
|
43
|
Evans AM, Wyatt CN, Kinnear NP, Clark JH, Blanco EA. Pyridine nucleotides and calcium signalling in arterial smooth muscle: from cell physiology to pharmacology. Pharmacol Ther 2005; 107:286-313. [PMID: 16005073 DOI: 10.1016/j.pharmthera.2005.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2005] [Indexed: 10/25/2022]
Abstract
It is generally accepted that the mobilisation of intracellular Ca2+ stores plays a pivotal role in the regulation of arterial smooth muscle function, paradoxically during both contraction and relaxation. However, the spatiotemporal pattern of different Ca2+ signals that elicit such responses may also contribute to the regulation of, for example, differential gene expression. These findings, among others, demonstrate the importance of discrete spatiotemporal Ca2+ signalling patterns and the mechanisms that underpin them. Of fundamental importance in this respect is the realisation that different Ca2+ storing organelles may be selected by the discrete or coordinated actions of multiple Ca2+ mobilising messengers. When considering such messengers, it is generally accepted that sarcoplasmic reticulum (SR) stores may be mobilised by the ubiquitous messenger inositol 1,4,5 trisphosphate. However, relatively little attention has been paid to the role of Ca2+ mobilising pyridine nucleotides in arterial smooth muscle, namely, cyclic adenosine diphosphate-ribose (cADPR) and nicotinic acid adenine dinucleotide phosphate (NAADP). This review will therefore focus on these novel mechanisms of calcium signalling and their likely therapeutic potential.
Collapse
Affiliation(s)
- A Mark Evans
- Division of Biomedical Sciences, School of Biology, Bute Building, University of St. Andrews, St. Andrews, Fife KY16 9TS, UK.
| | | | | | | | | |
Collapse
|
44
|
Gerth A, Nieber K, Oppenheimer N, Hauschildt S. Extracellular NAD+ regulates intracellular free calcium concentration in human monocytes. Biochem J 2005; 382:849-56. [PMID: 15233622 PMCID: PMC1133960 DOI: 10.1042/bj20040979] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2004] [Revised: 06/30/2004] [Accepted: 07/02/2004] [Indexed: 01/07/2023]
Abstract
Ca(2+) ions play a critical role in the biochemical cascade of signal transduction pathways, leading to the activation of immune cells. In the present study, we show that the exposure of freshly isolated human monocytes to NAD(+) results in a rapid concentration-dependent elevation of [Ca(2+)](i) (intracellular free Ca(2+) concentration) caused by the influx of extracellular Ca(2+). NAD(+) derivatives containing a modified adenine or nicotinamide ring failed to trigger a Ca(2+) increase. Treating monocytes with ADPR (ADP-ribose), a major degradation product of NAD(+), also resulted in a rise in [Ca(2+)](i). Selective inhibition of CD38, an NAD-glycohydrolase that generates free ADPR from NAD(+), does not abolish the effect of NAD(+), excluding the possibility that NAD(+) might act via ADPR. The NAD(+)-induced Ca(2+) response was prevented by the prior addition of ADPR and vice versa, indicating that both compounds share some mechanisms mediating the rise in [Ca(2+)](i). NAD(+), as well as ADPR, were ineffective when applied following ATP, suggesting that ATP controls events that intersect with NAD(+) and ADPR signalling.
Collapse
Affiliation(s)
- Anja Gerth
- *Department of Immunobiology, Institute of Zoology, University of Leipzig, Liebigstrasse 18, D-04103 Leipzig, Germany
| | - Karen Nieber
- †Department of Pharmacology for Natural Sciences, Institute of Pharmacy, University of Leipzig, Liebigstrasse 18, D-04103 Leipzig, Germany
| | - Norman J. Oppenheimer
- ‡Department of Pharmaceutical Chemistry S-926, University of California, San Francisco, CA 94143-0446, U.S.A
| | - Sunna Hauschildt
- *Department of Immunobiology, Institute of Zoology, University of Leipzig, Liebigstrasse 18, D-04103 Leipzig, Germany
- To whom correspondence should be addressed (email )
| |
Collapse
|
45
|
Bai N, Lee HC, Laher I. Emerging role of cyclic ADP-ribose (cADPR) in smooth muscle. Pharmacol Ther 2004; 105:189-207. [PMID: 15670626 DOI: 10.1016/j.pharmthera.2004.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2004] [Accepted: 10/14/2004] [Indexed: 10/26/2022]
Abstract
Cyclic adenosine diphosphate ribose (cADPR) is a naturally occurring cyclic nucleotide and represents a novel class of endogenous Ca(2+) messengers implicated in the regulation of the gating properties of ryanodine receptors (RyRs). This action of cADPR occurs independently from the inositol-1,4,5-trisphosphate (IP(3)) receptor. The regulation of intracellular Ca(2+) release is a fundamental element of cellular Ca(2+) homeostasis since a number of smooth muscle functions (tone, proliferation, apoptosis, and gene expression) are modulated by intracellular Ca(2+) concentration ([Ca(2+)](i)). There has been a surge in the efforts aimed at understanding the mechanisms of cADPR-mediated Ca(2+) mobilization and its impact on smooth muscle function. This review summarizes the proposed roles of cADPR in the regulation of smooth muscle tone.
Collapse
Affiliation(s)
- Ni Bai
- Department of Pharmacology and Therapeutics, University of British Columbia Vancouver, BC, Canada V6T 1Z3
| | | | | |
Collapse
|
46
|
Zhang AY, Yi F, Teggatz EG, Zou AP, Li PL. Enhanced production and action of cyclic ADP-ribose during oxidative stress in small bovine coronary arterial smooth muscle. Microvasc Res 2004; 67:159-67. [PMID: 15020207 DOI: 10.1016/j.mvr.2003.11.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2003] [Indexed: 11/28/2022]
Abstract
Recent studies in our lab and by others have indicated that cyclic ADP-ribose (cADPR) as a novel second messenger is importantly involved in vasomotor response in various vascular beds. However, the mechanism regulating cADPR production and actions remains poorly understood. The present study determined whether changes in redox status influence the production and action of cADPR in coronary arterial smooth muscle cells (CASMCs) and thereby alters vascular tone in these arteries. HPLC analyses demonstrated that xanthine (X, 40 microM)/xanthine oxidase (XO, 0.1 U/ml), a superoxide-generating system, increased the ADP-ribosyl cyclase activity by 59% in freshly isolated bovine CASMCs. However, hydrogen peroxide (H2O2, 1-100 microM) had no significant effect on ADP-ribosyl cyclase activity. In these CASMCs, X/XO produced a rapid increase in [Ca2+]i (Delta[Ca2+]i=201 nM), which was significantly attenuated by a cADPR antagonist, 8-Br-cADPR. Both inhibition of cADPR production by nicotinamide (Nicot) and blockade of Ca2+-induced Ca2+ release (CICR) by tetracaine (TC) and ryanodine (Rya) significantly reduced X/XO-induced rapid Ca2+ responses. In isolated, perfused, and pressurized small bovine coronary arteries, X at 2.5-80 microM with a fixed XO level produced a concentration-dependent vasoconstriction with a maximal decrease in arterial diameter of 45%. This X/XO-induced vasoconstriction was significantly attenuated by 8-Br-cADPR, Nicot, TC, or Rya. We conclude that superoxide activates cADPR production, and thereby mobilizes intracellular Ca2+ from the SR and produces vasoconstriction in coronary arteries.
Collapse
Affiliation(s)
- Andrew Y Zhang
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | |
Collapse
|
47
|
Lacapère JJ, Boulla G, Lund FE, Primack J, Oppenheimer N, Schuber F, Deterre P. Fluorometric studies of ligand-induced conformational changes of CD38. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2003; 1652:17-26. [PMID: 14580993 DOI: 10.1016/j.bbapap.2003.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The lymphoid surface antigen CD38 is a NAD(+)-glycohydrolase that also catalyzes the transformation of NAD(+) into cyclic ADP-ribose, a calcium mobilizing second messenger. In addition, ligation of CD38 by antibodies triggers signaling in lymphoid cells. Since the cytoplasmic tail of CD38 is dispensable for this latter property, we have previously proposed that CD38-mediated receptor signal transduction might be regulated by its conformational state. We have now examined the molecular changes of this protein during its interaction with NAD(+) by measuring the intrinsic fluorescence of CD38. We have shown that addition of the substrate produced a dramatic decrease in the fluorescence of the catalytically active recombinant soluble ectodomain of murine CD38. Analysis of this event revealed that the catalytic cycle involves a state of the enzyme that is characterized by a low fluorescence which, upon substrate turnover, reverts to the initial high intrinsic fluorescence level. In contrast, non-hydrolyzable substrates trap CD38 in its altered low fluorescence state. Studies with the hydrophilic quencher potassium iodide revealed that the tryptophan residues that are mainly involved in the observed changes in fluorescence, are remote from the active site. Similar data were also obtained with human CD38, indicating that studies of intrinsic fluorescence will be useful in monitoring the transconformation of CD38 from different species. Together, these data demonstrate that CD38 undergoes a reversible conformational change after substrate binding, and suggest a mechanism by which this change could alter interactions with different cell-surface partners.
Collapse
Affiliation(s)
- Jean-Jacques Lacapère
- INSERM U410, Faculté de Médecine Xavier Bichat, 16 rue Henri Huchard, 75018 Paris, France
| | | | | | | | | | | | | |
Collapse
|
48
|
Muñoz P, Navarro MDC, Pavón EJ, Salmerón J, Malavasi F, Sancho J, Zubiaur M. CD38 Signaling in T Cells Is Initiated within a Subset of Membrane Rafts Containing Lck and the CD3-ζ Subunit of the T Cell Antigen Receptor. J Biol Chem 2003; 278:50791-802. [PMID: 14523017 DOI: 10.1074/jbc.m308034200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In this study we present data supporting that most CD38 is pre-assembled in a subset of Brij 98-resistant raft vesicles, which were stable at 37 degrees C, and have relatively high levels of Lck and the CD3-zeta subunit of T cell antigen receptor-CD3 complex in contrast with a Brij 98-soluble pool, where CD38 is associated with CD3-zeta, and Lck is not detected. Our data further indicate that following CD38 engagement, LAT and Lck are tyrosine phosphorylated exclusively in Brij 98-resistant rafts, and some key signaling components translocate into rafts (i.e. Sos and p85-phosphatidylinositol 3-kinase). Moreover, N-Ras results activated within rafts immediately upon CD38 ligation, whereas activated Erk was mainly found in soluble fractions with delayed kinetics respective to Ras activation. Furthermore, full phosphorylation of CD3-zeta and CD3-epsilon only occurs in rafts, whereas partial CD3-zeta tyrosine phosphorylation occurs exclusively in the soluble pool, which correlated with increased levels of c-Cbl tyrosine phosphorylation in the non-raft fractions. Taken together, these results suggest that, unlike the non-raft pool, CD38 in rafts is able to initiate and propagate several activating signaling pathways, possibly by facilitating critical associations within other raft subsets, for example, LAT rafts via its capacity to interact with Lck and CD3-zeta. Overall, these findings provide the first evidence that CD38 operates in two functionally distinct microdomains of the plasma membrane.
Collapse
Affiliation(s)
- Pilar Muñoz
- Instituto de Parasitología y Biomedicina, Consejo Superior de Investigaciones Científicas, 18001 Granada, Spain
| | | | | | | | | | | | | |
Collapse
|
49
|
Lu YC, Kuo SY, Jiann BP, Chang HT, Chen WC, Huang JK, Jan CR. Triethyltin increases cytosolic Ca(2+) levels in human osteoblasts. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2003; 14:1-7. [PMID: 21782656 DOI: 10.1016/s1382-6689(03)00004-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2002] [Accepted: 12/18/2002] [Indexed: 05/31/2023]
Abstract
In human osteosarcoma MG63 cells, effect of triethyltin, an environmental toxicant, on intracellular Ca(2+) concentration ([Ca(2+)](i)) was measured by using fura-2. Triethyltin (1-50 μM) caused a rapid and sustained plateau rise of [Ca(2+)](i) in a concentration-dependent manner (EC(50)=10 μM). Triethyltin-induced [Ca(2+)](i) rise was prevented by 50% by removal of extracellular Ca(2+) but was not altered by voltage-gated Ca(2+) channel blockers. In Ca(2+)-free medium, thapsigargin, an inhibitor of the endoplasmic reticulum (ER) Ca(2+)-ATPase, caused a monophasic [Ca(2+)](i) rise, after which the increasing effect of triethyltin on [Ca(2+)](i) was attenuated by 60%; also, pretreatment with triethyltin abolished thapsigargin-induced [Ca(2+)](i) increase. Depletion of mitochondrial Ca(2+) with carbonylcyanide m-chlorophenylhydrazone (CCCP; 2 μM) did not affect triethyltin-induced Ca(2+) release. U73122, an inhibitor of phoispholipase C, abolished ATP (but not triethyltin)-induced [Ca(2+)](i) rise. A low concentration (1 μM) of triethyltin failed to alter ATP and bradykinin-induced [Ca(2+)](i) rises. These findings suggest that triethyltin rapidly increases [Ca(2+)](i) in osteoblasts by stimulating both extracellular Ca(2+) influx and intracellular Ca(2+) release via as yet unidentified mechanism(s).
Collapse
Affiliation(s)
- Yih-Chau Lu
- Department of Orthopaedic Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Members of the transient receptor potential (TRP) family for which mRNA can be demonstrated in neutrophil granulocytes with RT-PCR include TRPC6 (as only "short" TRP), TRPM2, TRPV1, TRPV2, TRPV5 and TRPV6. When these are analyzed in heterologous overexpression experiments, TRPM2 is the only cation channel with characteristic properties that can be used as fingerprint to provide functional evidence for its expression in neutrophil granulocytes. As cells transfected with TRPM2, neutrophil granulocytes display non-selective cation currents and typical channel activity evoked by intracellular ADP-ribose and NAD. Thus, stimulation of TRPM2 is likely to occur after activation of CD38 (producing ADP-ribose) and during the oxidative burst (enhancing the NAD concentration). This novel mode of cation entry regulation may be of particular importance for the response of granulocytes to chemoattractants. TRPV6 is a likely but not exclusive candidate as subunit of the channels mediating store-operated Ca2+ entry (SOCE). Evidence for SOCE in granulocytes has been presented with the fura-2 technique but not with electrophysiological methods although Ca2+-selective store-operated currents can be demonstrated in HL-60 cells, a cell culture model of neutrophil granulocytes.
Collapse
Affiliation(s)
- Inka Heiner
- Institut für Physiologie, Universitätsklinikum der RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | | | | |
Collapse
|