NAD+ metabolism and retinal degeneration (Review)

  • Authors:
    • Andreea Silvia Pîrvu
    • Ana Marina Andrei
    • Elena Camelia Stănciulescu
    • Ileana Monica Baniță
    • Cătălina Gabriela Pisoschi
    • Sanda Jurja
    • Radu Ciuluvica
  • View Affiliations

  • Published online on: April 23, 2021     https://doi.org/10.3892/etm.2021.10102
  • Article Number: 670
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The recent years has revealed an intense interest in the study of nicotinamide adenine dinucleotide (NAD+), particularly in regards to its intermediates, such as nicotinamide and nicotinic acid known as niacin, and also nicotinamide riboside. Besides its participation as a coenzyme in the redox transformations of nutrients during catabolism, NAD+ is also involved in DNA repair and epigenetic modification of gene expression and also plays an essential role in calcium homeostasis. Clinical and experimental data emphasize the age‑dependent decline in NAD+ levels and its relation with the onset and progression of various age‑related diseases. Maintaining optimal levels of NAD+ has aroused a therapeutic interest in such pathological conditions; NAD+ being currently regarded as an important target to extend health and lifespan. Based on a systematic exploration of the experimental data and literature surrounding the topic, this paper reviews some of the recent research studies related to the roles of the pyridine nucleotide family focusing on biosynthesis, NAD+ deficiency‑associated diseases, pathobiochemistry related to retinal degeneration and potential therapeutic effects on human vision as well.

1. Introduction

Nicotinamide adenine dinucleotide (NAD+) is considered the precursor for the pyridine nucleotide family constituted by NADH and two phosphorylated forms, nicotinamide adenine dinucleotide phosphate in its oxidized (NADP+) and reduced (NADPH) forms (1). All of these nucleotides are well-known cofactors in numerous cellular processes. NAD+ is the substrate for major redox transformations of nutrients during the catabolic phase of metabolism. Its reduced form, NADH, is the main source of electrons for mitochondrial oxidative chain and production of adenosine triphosphate through oxidative phosphorylation. NAD+ is also involved in DNA repair, as a substrate for poly(ADP-ribose) polymerases (PARP) (2), epigenetic modification of gene expression, also having an important influence on immunological function (3). NAD+ also has a critical role in calcium homeostasis, as the substrate for NAD+ glycohydrolases, enzymes related to the production of cyclic ADP-ribose, a calcium efflux effector (4).

Since the original discovery of NAD+ by Harden and Young in 1906 in cell-free yeast juices as a factor that enhanced the rate of fermentation, many studies have been developed to reveal the NAD+ biosynthetic pathway as a key player in cellular metabolism (5). NAD+ is currently regarded as an important target with which to extend lifespan and health span. Accurate evaluation of the NAD+ metabolome is of great interest due to its association with cognitive impairment, cancer, normal aging and age-related disorders (1).

Research conducted to date has elucidated the signaling pathways and cellular processes that contribute to the maintenance of pyridine nucleoside and nucleotide homeostasis; the regulation of molecular mechanisms underlying NAD+ metabolism is known but the use of its precursors in therapy are still incompletely explained.

2. NAD+ biosynthesis, metabolism and effects on the human body

NAD+ biosynthesis in mammals involves five important intermediates including tryptophan, niacin, nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN), through three biosynthetic pathways: i) Nucleotide salvage from nicotinamide (NAM), NR and NMN; ii) de novo synthesis from tryptophan and iii) Preiss-Handler pathway from nicotinic acid (NA) (6).

In mammals, the salvage pathway is the major route to generate NAD+. Conversion of NMN, one of the forms of the water-soluble vitamin B3, to NAD+ is catalyzed by nicotinamide mononucleotide adenylyltransferase (Nmnat) (Fig. 1). NMN can be synthesized by nicotinamide phosphoribosyltransferase (Nampt) from NAM. Bieganowski and Brenner revealed an alternative NAD+ salvage biosynthesis pathway, in which NR, another form of vitamin B3 that enters cells through nucleoside transporters, could be converted to NAD+ by nicotinamide riboside kinases (NMRK1 and NMRK2) and Nmnat via phosphorylation reactions (7).

In recent years, NMN and NR have been extensively investigated in various experiments on rodents and humans and evidence suggests that with age NAD+ levels decline at a systemic level (8,9), causing profound metabolic changes (10). These precursors of NAD+ have an essential influence on the elevation of NAD+ concentration in a variety of tissues, automatically suggesting a beneficial therapeutic effect (8,11,12).

The metabolic balance of NAD+ in the cellular environment can be accomplished in several ways and involves multiple metabolites in an oxidized and reduced form such as NADP+, NADH, NADPH, and nicotinic acid adenine dinucleotide phosphate (NAADP), all with an essential role in energy production and cellular metabolism, acting as one proton-accepting or donating coenzymes. NAD+ derivatives are important for regulating cellular redox status, intracellular Ca2+ pools, DNA damage and repair, cell cycle timing and lipid and energy metabolism (13). NAD+ is a major cofactor for mitochondrial ATP production and for NAD-dependent enzymes including sirtuins and poly-ADP-ribosylpolymerases, essential players in fundamental processes such as cell division and proliferation, apoptosis, aging, senescence and stress resistance (14). Lin and Guarente argued that sirtuins can sense the NADH/NAD+ ratio in cells by the recognition of the oxidized dinucleotides, but it seems that other cellular factors also respond to the alterations within NADH levels (15).

For many years it was believed that NAD+ breakdown is a nonspecific process but in the last decade evidence has shown that NAD+ consumption is linked with the signaling reactions inside and outside the cells (16). For example, the oxidation of glucose and fatty acids lead to the reduction of oxidized NAD+ to NADH and alterations in its concentration were found to be involved in multiple pathogenic signaling pathways, including heart failure (17). The last two decades generated multiple discoveries regarding NAD+ and its precursors, identifying the important role in DNA repair, immune activation and epigenetic control through protein deacetylation (3). Even if the metabolism of NAD+ has been the subject of numerous research studies in various cells, tissues, and organs, those regarding pyridine nucleotide metabolism in eye structures and its involvement in visual dysfunctions are scarce.

3. Diet and NAD+ level

Dietary supplementation is the only way to increase body NAD+ levels. Since NAD+ administration is not efficient enough to increase NAD+ levels, its precursors, such as NAM and NA known as niacin, NMN and NR could be useful to increase NAD+ levels in animal models and humans (18).

Most raw foods provide these substances which can regulate cellular activities and the timing of changes that lead to aging phenotypes (13). Another important precursor of NAD+ is the amino acid tryptophan which is synthetized via the kynurenine pathway. While NR is the form of vitamin B3 found in humans and cow milk and other foods, NA is produced by plants and algae, and tryptophan is the most abundant amino acid found in animal and plant proteins (19).

Many of the biochemical studies performed recently have shown that a poor diet lacking niacin and tryptophan intake or a chronic immune activation can lead to inefficient production of NAD+, where catabolism exceeds anabolism producing a consequent cellular dysfunction (20). By contrast, a reduced energy load due to activities such exercise, calorie restriction, fasting and glucose deprivation can increase the concentration of NAD+ (8).

Although studies on rodents have shown that both NMN and NR enhance NAD+ biosynthesis and have beneficial effects in multiple disorders (21-23), it still remains unclear what mechanisms mediate their beneficial effect. Presently, the pharmacokinetics and metabolic fates of NAD+ precursors are still under investigation, multiple human clinical trials being conducted to study and understand the safety of NAD+ precursor supplementation, mainly NR and NMN (24-26). The concentrations of NAD+ and NAM in blood are in the micromolar range and can be boosted by oral administration of their precursors, NR and NMN. In their study, Trammell et al showed that a daily dose of 1000 mg NR leads to a 2.7-fold rise in blood NAD+ after one dose of NR (24). Another clinical study demonstrated that daily doses of NR up to 1000 mg are well tolerated and efficient to increase NAD+ levels by almost 60% in peripheral blood mononuclear cells (25). Irie et al conducted a human study regarding the safety of a single oral NMN dose (100, 250 or 500 mg) in healthy Japanese men by investigating the pharmacokinetics of NMN metabolites for 5 h after each intervention and found that NMN supplementation is safe without causing any significant deleterious effects (27).

4. NAD+ and diseases

Multiple studies strongly confirm that deficiencies in NAD+ levels can lead to degenerative diseases and multiple pathologies such as metabolic disorders, heart and renal diseases, cognitive impairment and even cancer (28,29).

In the past few years, research on the biology of NAD+ has provided many critical insights into the pathogenesis of age-associated functional decline (30).

Studies on rodents have demonstrated that under normal and pathophysiological conditions, systemic NMN administration enhanced NAD+ biosynthesis in various peripheral tissues such as the pancreas, liver and white adipose tissue of diabetic mice (21), heart (31,32), skeletal muscle (33), kidney (34,35), eyes (36) and blood vessels (37,38). Recent studies on mouse and rat models suggest that NMN improves the neuronal function with visible benefits on cognition and memory (39-41).

NR is also a direct factor involved in NAD+ metabolism with a recognized efficacy in metabolic and skeleton-muscular disorders. Similar to NMN, NR also exhibits protective effects against aging and age-related diseases, improves liver health and protects against diabetic and chemotherapy induced neuropathy (42).

In the past few years, metabolic syndrome has become a global health concern due to its critical risk in regards to the development of various life-threatening conditions such as cardiovascular disease, stroke and cancer (43). Obesity, diabetes, dyslipidemia, fatty liver and hypertension are common diseases linked with the metabolic syndrome and are closely associated with impaired nutrient status and lifestyle (44). A number of studies have demonstrated that NAD+ levels decline with aberrant nutritional status and its prevention is regarded as a promising strategy with which to combat metabolic disorders (18). Administration of NMN and NR can ameliorate diet-associated weight gain, dietary intervention, their use being a promising treatment against obesity (45). In addition, the administration of NR in mice was found to increase fatty acid oxidation and energy expenditure and also to improve insulin sensitivity (45). On other hand, studies on various diabetic rodent models, have revealed that NAM administration ameliorates hyperglycemia by increasing β-cell proliferation (46).

Recent studies indicate that NAD+ is involved in tumor cell progression being considered a promising therapeutic target for cancer (47-49). NAD+-metabolizing enzymes play an important role in conditioning several aspects of cancer and immune cell fate and functions (50). Cancer cells display a unique energy metabolism and the continuous replenishment of NAD+ promotes the proliferation and survival of fast-dividing cancer cells as elevated NAD+ levels enhance anaerobic glycolysis (51). Increased intracellular NAD+ levels accompanied by overexpression of NAD+ salvage biosynthesis pathway enzymes sustain tumor cell proliferation and promote survival against anticancer cell agents (52).

Overexpression of Nampt is frequently observed in several types of malignant tumors, for example, breast, colorectal, gastric, ovarian, prostate cancers, and malignant lymphomas (48,52). A recent study showed that high expression of Nampt and Naprt predicts a poor prognosis in colorectal cancer and was associated with vascular invasion. This finding suggests that these two enzymes of the NAD+ salvage synthesis pathway may be novel markers for the diagnosis and treatment of colorectal cancer (53).

5. NAD+ in normal and abnormal function of the retina

The retina is a highly organized eye structure, light-sensitive, intensively innervated and vascularized. It consists of numerous cell types that function in a coordinated manner to give rise to a signal transmitted to the brain. To create an image, light photons are sensed by the retinal receptors and the signal from the photoreceptors is then transmitted to secondary neurons before being transmitted to the retinal ganglion cells. Retinal photoreceptor death is a common cause of blindness in retinal degenerative diseases (54-56).

Retinal degenerative diseases are a major cause of morbidity because visual impairment significantly decreases the quality of life of patients. These have different etiologies, being either acquired, clinical manifestation of a systemic problem or inherited (54).

Despite their diversity, these diseases focus on the death of photoreceptors. The huge metabolic and energetic requirements of the retinal photoreceptors and their progressive impairment in various retinal degenerative diseases have been for a long time the subject of research.

Given the key metabolic role of NAD+ and the high metabolism in photoreceptor cells, there is a growing interest in debating the role of the ‘NADome’ in controlling retinal metabolism and mediating the pathogenesis of retinal degeneration.

The involvement of NAD+ biosynthesis pathways in retinal photoreceptor dysfunction has been previously investigated. The mutation of Nmnat1, one of the key enzymes involved in NAD+ biosynthesis, was identified as a cause of a childhood blinding disease (57). Rodent studies that replicated this mutation showed the same alterations (for example, rapidly photoreceptor degeneration, retinal pigment epithelial cell loss, retinal vasculature attenuation) and confirm its importance for retinal survival (58).

Data reported by Kaja and colleagues showed that altered circulating levels of Nampt were also significantly associated with risk for retinal vein occlusion, ischemia and metabolic alteration (59). Other studies confirmed the relevance of Nampt for retinal function proving its importance for maintenance of functional retinal pigment epithelial and endothelial cells. For example, inhibition of Nampt with pharmacological inhibitors or deficits in NAD+ bioavailability promoted an early senescent phenotype in these cells (60,61).

NAD+ metabolism is also important for cell longevity. Age-dependent decline in NAD+ levels reported for multiple human organs has been demonstrated to occur also in the retina, especially in photoreceptor, ganglion, endothelial, and retinal pigment epithelial cells (62). That is why extensive research has focused on the role of NAD+ as a modulator of sirtuin activity, known that sirtuins play important roles in the retina, confer protection against oxidative stress and retinal degeneration being able to increase lifespan (63). All seven mammal sirtuins (SRT1-SRT7) are highly expressed in the mouse retina (64) and could enhance retinal metabolism, reduce photoreceptor death, and improve vision.

Given the roles of sirtuins in retinal function, pharmacological modulation of sirtuin activation may be a therapeutic strategy for preventing retinal degeneration. The supply with NAD+ precursors, such as NMN and NR, represent an interesting approach to this condition. In a mouse model of light-induced acute retinal degeneration, Zhang and colleagues found that NR treatment prevented NAD+ diminution following toxic light exposure (65). Maintenance of NAD+ levels allowed continued activity of enzymes for which NAD+ is a substrate, for example mitochondrial sirtuins, on which optimal activity relies NAD+-dependent retinal homeostasis (66). Supplementation with these precursors was found to lead to increased intracellular NAD+ bioavailability and enhanced sirtuin function, especially in contexts of cellular stress when NAD+ requirements may be increased and have been shown to be critical to retinal health. Further studies are important to ensure the development of new safe and efficient therapeutic strategies.

6. Conclusions

While the first century of NAD+ research has revealed multiple discoveries which prove that NAD+ plays a key role in the regulation of cell metabolism, stress and immune responses to physiological or pathological signals, it is widely recognized that additional research is required to fully reveal the complex metabolic fate of the ‘NADome’ in order to sustain the paramount importance of the supplementation of NAD+ precursors to promote health and improve therapeutic safety in various diseases, including retinal disturbances.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

All information discussed in this review is documented by relevant references.

Authors' contributions

ASP, AMA, ECS, IMB, CGP, SJ and RC made equal contributions to the conception and editing of this manuscript. ASP, AMA, ECS, CGP, SJ and RC contributed to the acquisition of the data. ASP, AMA, ECS, CGP, IMB, SJ and RC selected, processed the data acquired and wrote appropriate parts of this manuscript. SJ, RC, CGP and IMB revised its contents. All authors read and approved the final manuscript to be published.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Clement J, Wong M, Poljak A, Sachdev P and Braidy N: The plasma NAD+ metabolome is dysregulated in ‘normal’ aging. Rejuvenation Res. 22:121–130. 2019.PubMed/NCBI View Article : Google Scholar

2 

Surjana D, Halliday GM and Damian DL: Role of nicotinamide in DNA damage, mutagenesis, and DNA repair. J Nucleic Acids. 2010(157591)2010.PubMed/NCBI View Article : Google Scholar

3 

Braidy N, Beg J, Clement J, Khorshidi F, Poljak A, Jayasena T, Grant R and Sachdev P: Role of nicotinamide adenine dinucleotide and related precursors as therapeutic targets for age-related degenerative diseases: Rationale, biochemistry, pharmacokinetics, and outcomes. Antioxid Redox Signal. 30:251–294. 2019.PubMed/NCBI View Article : Google Scholar

4 

Horenstein AL, Sizzano F, Lusso R, Besso FG, Ferrero E, Deaglio S, Corno F and Malavasi F: CD38 and CD157 ectoenzymes mark cell subsets in the human corneal limbus. Mol Med. 15:76–84. 2009.PubMed/NCBI View Article : Google Scholar

5 

Croft T, Venkatakrishnan P and Lin SJ: NAD+ metabolism and regulation: Lessons from yeast. Biomolecules. 10(330)2020.PubMed/NCBI View Article : Google Scholar

6 

Wherry EJ and Kurachi M: Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 15:486–499. 2015.PubMed/NCBI View Article : Google Scholar

7 

Bieganowski P and Brenner C: Discoveries of nicotinamide riboside as a nutrient and conserved NRK genes establish a Preiss-Handler independent route to NAD+ in fungi and humans. Cell. 117:495–502. 2004.PubMed/NCBI View Article : Google Scholar

8 

Canto C, Menzies KJ and Auwerx J: NAD+ metabolism and the control of energy homeostasis: A balancing act between mitochondria and the nucleus. Cell Metab. 22:31–53. 2015.PubMed/NCBI View Article : Google Scholar

9 

Verdin E: NAD+ in aging, metabolism, and neurodegeneration. Science. 350:1208–1213. 2015.PubMed/NCBI View Article : Google Scholar

10 

Finley LW and Haigis MC: The coordination of nuclear and mitochondrial communication during aging and calorie restriction. Age Res Rev. 8:173–188. 2009.PubMed/NCBI View Article : Google Scholar

11 

Chini CC, Tarrago MG and Chini EN: NAD and the aging process: Role in life, death and everything in between. Mol Cell Endocrinol. 455:62–74. 2017.PubMed/NCBI View Article : Google Scholar

12 

Fang EF, Lautrup S, Hou Y, Demarest TG, Croteau DL, Mattson MP and Bohr VA: NAD+ in aging: Molecular mechanisms and translational implications. Trends Mol Med. 23:899–916. 2017.PubMed/NCBI View Article : Google Scholar

13 

Xu P and Sauve A: Vitamin B3, the nicotinamide adenine dinucleotides and aging. Mech Ageing Dev. 131:287–298. 2010.PubMed/NCBI View Article : Google Scholar

14 

van de Ven RA, Santos D and Haigis MC: Mitochondrial sirtuins and molecular mechanisms of aging. Trends Mol Med. 23:320–331. 2017.PubMed/NCBI View Article : Google Scholar

15 

Lin SJ and Guarente L: Nicotinamide adenine dinucleotide, a metabolic regulator of transcription, longevity and disease. Curr Opin Cell Biol. 15:241–246. 2003.PubMed/NCBI View Article : Google Scholar

16 

Belenky P, Bogan KL and Brenner C: NAD+ metabolism in health and disease. Trends Biochem Sci. 32:12–19. 2007.PubMed/NCBI View Article : Google Scholar

17 

Mericskay M: Nicotinamide adenine dinucleotide homeostasis and signalling in heart disease: Pathophysiological implications and therapeutic potential. Arch Cardiovasc Dis. 109:207–215. 2016.PubMed/NCBI View Article : Google Scholar

18 

Okabe K, Yaki K, Tobe K and Nakagawa T: Implications of altered NAD metabolism in metabolic disorders. J Biomed Sci. 26(34)2019.PubMed/NCBI View Article : Google Scholar

19 

Trammell SA, Yu L, Redpath P, Migaud ME and Brenner C: Nicotinamide riboside is a major NAD+ precursor vitamin in cow milk. J Nutr. 146:957–963. 2016.PubMed/NCBI View Article : Google Scholar

20 

Tang K, Sham H, Hui E and Kirkland JB: Niacin deficiency causes oxidative stress in rat bone marrow cells but not through decreased NADPH or glutathione status. J Nutr Biochem. 19:746–753. 2008.PubMed/NCBI View Article : Google Scholar

21 

Yoshino J, Mills KF, Yoon MJ and Imai SI: Nicotinamide mononucleotide, a key NAD(+) intermediate, treats the pathophysiology of diet- and age-induced diabetes in mice. Cell Metab. 14:528–536. 2011.PubMed/NCBI View Article : Google Scholar

22 

Zhang H, Ryu D, Wu Y, Gariani K, Wang X, Luan P, D'Amico D, Ropelle ER, Lutolf MP, Aebersold R, et al: NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science. 352:1436–1443. 2016.PubMed/NCBI View Article : Google Scholar

23 

Mills KF, Yoshida S, Stein LR, Grozio A, Kubota S, Sasaki Y, Redpath P, Migaud ME, Apte RS, Uchida K, et al: Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cell Metab. 24:795–806. 2016.PubMed/NCBI View Article : Google Scholar

24 

Trammell SAJ, Schmidt MS, Weidemann BJ, Redpath P, Jaksch F, Dellinger RW, Li Z, Dale Abel E, Migaud ME and Brenner C: Nicotinamide riboside is uniquely and orally bioavailable in mice and humans. Nat Commun. 7(12948)2016.PubMed/NCBI View Article : Google Scholar

25 

Martens CR, Denman BA, Mazzo MR, Armstrong ML, Reisdorph N, McQueen MB, Chonchol M and Seals DR: Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults. Nat Commun. 9(1286)2018.PubMed/NCBI View Article : Google Scholar

26 

Elhassan YS, Kluckova K, Fletcher RS, Schmidt MS, Garten A, Doig CL, Cartwright DM, Oakey L, Burley CV, Jenkinson N, et al: Nicotinamide riboside augments the aged human skeletal muscle NAD+ metabolome and induces transcriptomic and anti-inflammatory signatures. Cell Rep. 28:1717–1728.e6. 2019.PubMed/NCBI View Article : Google Scholar

27 

Irie J, Inagaki E, Fujita M, Nakaya H, Mitsuishi M, Yamaguchi S, Yamashita K, Shigaki S, Ono T, Yukioka H, et al: Effect of oral administration of nicotinamide mononucleotide on clinical parameters and nicotinamide metabolite levels in healthy Japanese men. Endocr J. 67:153–160. 2020.PubMed/NCBI View Article : Google Scholar

28 

Ralto KM, Rhee EP and Parikh SM: NAD+ homeostasis in renal health and disease. Nat Rev Nephrol. 16:99–111. 2020.PubMed/NCBI View Article : Google Scholar

29 

Katsyuba E, Romani M, Hofer D and Auwerx J: NAD+ homeostasis in health and disease. Nat Metab. 2:9–31. 2020.PubMed/NCBI View Article : Google Scholar

30 

Johnson S and Imai S: NAD+ biosynthesis, aging, and disease. F1000Res. 7(132)2018.PubMed/NCBI View Article : Google Scholar

31 

Yamamoto T, Byun J, Zhai P, Ikeda Y, Oka S and Sadoshima J: Nicotinamide mononucleotide, an intermediate of NAD+ synthesis, protects the heart from ischemia and reperfusion. PLoS One. 9(e98972)2014.PubMed/NCBI View Article : Google Scholar

32 

Martin AS, Abraham DM, Hershberger KA, Bhatt DP, Mao L, Cui H, Liu J, Liu X, Muehlbauer MJ, Grimsrud PA, et al: Nicotinamide mononucleotide requires SIRT3 to improve cardiac function and bioenergetics in a Friedreich's ataxia cardiomyopathy model. JCI Insight. 2(e93885)2017.PubMed/NCBI View Article : Google Scholar

33 

Ryu D, Zhang H, Ropelle ER, Sorrentino V, Mázala DAG, Mouchiroud L, Marshall PL, Campbell MD, Ali AS, Knowels GM, et al: NAD+ repletion improves muscle function in muscular dystrophy and counters global PARylation. Sci Transl Med. 8(361ra139)2016.PubMed/NCBI View Article : Google Scholar

34 

Guan Y, Wang SR, Huang XZ, Xie QH, Xu YY, Shang D and Hao CM: Nicotinamide mononucleotide, an NAD+ precursor, rescues age-associated susceptibility to AKI in a sirtuin 1-dependent manner. J Am Soc Nephrol. 28:2337–2352. 2017.PubMed/NCBI View Article : Google Scholar

35 

Yoshino J, Baur JA and Imai SI: NAD+ intermediates: The biology and therapeutic potential of NMN and NR. Cell Metab. 27:513–528. 2018.PubMed/NCBI View Article : Google Scholar

36 

Lin JB, Kubota S, Ban N, Yoshida M, Santeford A, Sene A, Nakamura R, Zapata N, Kubota M, Tsubota K, et al: NAMPT-mediated NAD(+) biosynthesis is essential for vision in mice. Cell Rep. 17:69–85. 2016.PubMed/NCBI View Article : Google Scholar

37 

Zhao C, Li W, Duan H, Li Z, Jia Y, Zhang S, Wang X, Zhou Q and Shi W: NAD+ precursors protect corneal endothelial cells from UVB-induced apoptosis. Cell Physiol. 318:C796–C805. 2020.PubMed/NCBI View Article : Google Scholar

38 

de Picciotto NE, Gano LB, Johnson LC, Martens CR, Sindler AL, Mills KF, Imai S and Seals DR: Nicotinamide mononucleotide supplementation reverses vascular dysfunction and oxidative stress with aging in mice. Aging Cell. 15:522–530. 2016.PubMed/NCBI View Article : Google Scholar

39 

Wang X, Hu X, Yang Y, Takata T and Sakurai T: Nicotinamide mononucleotide protects against β-amyloid oligomer-induced cognitive impairment and neuronal death. Brain Res. 1643:1–9. 2016.PubMed/NCBI View Article : Google Scholar

40 

Tarantini S, Valcarcel-Ares MA, Toth P, Yabluchanskiy A, Tucsek Z, Kiss T, Hertelendy P, Kinter M, Ballabh P, Süle Z, et al: Nicotinamide mononucleotide (NMN) supplementation rescues cerebromicrovascular endothelial function and neurovascular coupling responses and improves cognitive function in aged mice. Redox Biol. 24(101192)2019.PubMed/NCBI View Article : Google Scholar

41 

Wang X, Hu X, Zhang L, Xu X and Sakurai T: Nicotinamide mononucleotide administration after sever hypoglycemia improves neuronal survival and cognitive function in rats. Brain Res Bull. 160:98–106. 2020.PubMed/NCBI View Article : Google Scholar

42 

Aman Y, Qiu Y, Tao J and Fang EF: Therapeutic potential of boosting NAD+ in aging and age-related diseases. Transl Med Aging. 2018:30–37. 2018.PubMed/NCBI View Article : Google Scholar

43 

Saklayen MG: The global epidemic of the metabolic syndrome. Curr Hypertens Rep. 20(12)2018.PubMed/NCBI View Article : Google Scholar

44 

O'Neill S and O'Driscoll L: Metabolic syndrome: A closer look at the growing epidemic and its associated pathologies. Obes Rev. 16:1–12. 2015.PubMed/NCBI View Article : Google Scholar

45 

Canto C, Houtkooper RH, Pirinen E, Youn DY, Oosterveer MH, Cen Y, Fernandez-Marcos PJ, Yamamoto H, Andreux PA, Cettour-Rose P, et al: The NAD(+) precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab. 15:838–847. 2012.PubMed/NCBI View Article : Google Scholar

46 

Shima K, Zhu M and Kuwajima M: A role of nicotinamide-induced increase in pancreatic beta-cell mass on blood glucose control after discontinuation of the treatment in partially pancreatectomized OLETF rats. Diabetes Res Clin Pract. 41:1–8. 1998.PubMed/NCBI View Article : Google Scholar

47 

Park SY, Lee KB, Lee MJ, Bae SC and Jang JJ: Nicotinamide inhibits the early stage of carcinogen-induced hepatocarcinogenesis in mice and suppresses human hepatocellular carcinoma cell growth. J Cell Physiol. 227:899–908. 2012.PubMed/NCBI View Article : Google Scholar

48 

Hong SM, Hwang SW, Wang T, Park CW, Ryu YM, Jung JH, Shin JH, Kim SY, Lee JL, Kim CW, et al: Increased nicotinamide adenine dinucleotide pool promotes colon cancer progression by suppressing reactive oxygen species level. Cancer Sci. 110:629–638. 2019.PubMed/NCBI View Article : Google Scholar

49 

Galbraith AR, Seabloom DE, Wuertz BR, Antonides JD, Steele VE, Wattenberg LW and Ondrey FG: Chemoprevention of lung carcinogenesis by dietary nicotinamide and inhaled budesonide. Cancer Prev Res. 12:69–78. 2019.PubMed/NCBI View Article : Google Scholar

50 

Audrito V, Managò A, Gaudino F, Sorci L, Messana VG, Raffaelli N and Deaglio S: NAD-biosynthetic and consuming enzymes as central players of metabolic regulation of innate and adaptive immune responses in cancer. Front Immunol. 10(1720)2019.PubMed/NCBI View Article : Google Scholar

51 

Yamamoto M, Inohara H and Nakagawa T: Targeting metabolic pathways for head and neck cancers therapeutics. Cancer Metastasis Rev. 36:503–514. 2017.PubMed/NCBI View Article : Google Scholar

52 

Yaku K, Okabe K, Hikosaka K and Nakagawa T: NAD metabolism in cancer therapeutics. Front Oncol. 8(622)2018.PubMed/NCBI View Article : Google Scholar

53 

Li X, Lei J, Mao L, Wang Q, Xu F, Ran T, Zhou Z and He S: NAMPT and NAPRT, key enzymes in NAD salvage synthesis pathway, are of negative prognostic value in colorectal cancer. Front Oncol. 9(736)2019.PubMed/NCBI View Article : Google Scholar

54 

Lin JB and Apte RS: NAD+ and sirtuins in retinal degenerative diseases: A look at future therapies. Prog Retin Eye Res. 67:118–129. 2018.PubMed/NCBI View Article : Google Scholar

55 

Jurja S, Hincu M, Dobrescu MA, Golu AE, Balasoiu AT and Coman M: Ocular cells and light: Harmony or conflict? Rom J Morphol Embryol. 55:257–261. 2014.PubMed/NCBI

56 

Jurja S, Coman M and Hincu M: The ultraviolet influence upon soft eye tissues. Rom J Morphol Embryol. 58:45–52. 2017.PubMed/NCBI

57 

Koenekoop RK, Wang H, Majewski J, Wang X, Lopez I, Ren H, Chen Y, Li Y, Fishman GA, Genead M, et al: Mutations in NMNAT1 cause Leber congenital amaurosis and identify a new disease pathway for retinal degeneration. Nat Genet. 44:1035–1039. 2012.PubMed/NCBI View Article : Google Scholar

58 

Greenwald SH, Charette JR, Staniszewska M, Shi LY, Brown SD, Stone L, Liu Q, Hicks WL, Collin GB, Bowl MR, et al: Mouse models of NMNAT1-Leber congenital Amaurosis (LCA9) recapitulate key features of the human disease. Am J Pathol. 186:1925–1938. 2016.PubMed/NCBI View Article : Google Scholar

59 

Kaja S, Shah A, Haji S, Patel KB, Naumchuk Y, Zabaneh A, Gerdes BC, Kunjukunju N, Sabates NR, Cassell MA, et al: Nampt/PBEF/visfatin serum levels: A new biomarker for retinal blood vessel occlusions. Clin Ophthalmol. 9:611–618. 2015.PubMed/NCBI View Article : Google Scholar

60 

Zabka TS, Singh J, Dhawan P, Liederer BM, Oeh J, Kauss MA, Xiao Y, Zak M, Lin T, McCray B, et al: Retinal toxicity, in vivo and in vitro, associated with inhibition of nicotinamide phosphoribosyltransferase. Toxicol Sci. 144:163–172. 2015.PubMed/NCBI View Article : Google Scholar

61 

Jadeja RN, Powell FL and Jones MA: Loss of NAMPT in aging retinal pigment epithelium reduces NAD+ availability and promotes cellular senescence. Aging. 10:1306–1323. 2018.PubMed/NCBI View Article : Google Scholar

62 

Jadeja RN, Thounaojam MC, Bartoli M and Martin PM: Implications of NAD+ metabolism in the aging retina and retinal degeneration. Oxid Med Cell Longev. 2020(2692794)2020.PubMed/NCBI View Article : Google Scholar

63 

Balaiya S, Abu-Amero KK, Kondkar AA and Chalam KV: Sirtuins expression and their role in retinal diseases. Oxid Med Cell Longev. 2017(3187594)2017.PubMed/NCBI View Article : Google Scholar

64 

Ban N, Ozawa Y, Inaba T, Miyake S, Watanabe M, Shinmura K and Tsubota K: Light-dark condition regulates sirtuin mRNA levels in the retina. Experimental Gerontol. 48:1212–1217. 2013.PubMed/NCBI View Article : Google Scholar

65 

Zhang X, Henneman NF, Girardot PE, Sellers JT, Chrenek MA, Li Y, Wang J, Brenner C, Nickerson JM and Boatright JH: Systemic treatment with nicotinamide riboside is protective in a mouse model of light-induced retinal degeneration. Invest Ophthalmol Vis Sci. 61(47)2020.PubMed/NCBI View Article : Google Scholar

66 

Lin JB, Kubota S, Mostoslavsky R and Apte RS: Role of sirtuins in retinal function under basal conditions. Adv Exp Med Biol. 1074:561–567. 2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

July-2021
Volume 22 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pîrvu AS, Andrei AM, Stănciulescu EC, Baniță IM, Pisoschi CG, Jurja S and Ciuluvica R: NAD<sup>+</sup> metabolism and retinal degeneration (Review). Exp Ther Med 22: 670, 2021
APA
Pîrvu, A.S., Andrei, A.M., Stănciulescu, E.C., Baniță, I.M., Pisoschi, C.G., Jurja, S., & Ciuluvica, R. (2021). NAD<sup>+</sup> metabolism and retinal degeneration (Review). Experimental and Therapeutic Medicine, 22, 670. https://doi.org/10.3892/etm.2021.10102
MLA
Pîrvu, A. S., Andrei, A. M., Stănciulescu, E. C., Baniță, I. M., Pisoschi, C. G., Jurja, S., Ciuluvica, R."NAD<sup>+</sup> metabolism and retinal degeneration (Review)". Experimental and Therapeutic Medicine 22.1 (2021): 670.
Chicago
Pîrvu, A. S., Andrei, A. M., Stănciulescu, E. C., Baniță, I. M., Pisoschi, C. G., Jurja, S., Ciuluvica, R."NAD<sup>+</sup> metabolism and retinal degeneration (Review)". Experimental and Therapeutic Medicine 22, no. 1 (2021): 670. https://doi.org/10.3892/etm.2021.10102