Nicotinamide mononucleotide improves spermatogenic function in streptozotocin-induced diabetic mice via modulating the glycolysis pathway

Spermatogenic dysfunction is one of the major secondary complications of diabetes; however, the underlying mechanisms remain ill-defined, and there is no available drug or strategy for the radical treatment of diabetic spermatogenic dysfunction. Therefore, the objective of this study is to investigate the protective effects of nicotinamide mononucleotide (NMN) on testicular spermatogenic function in streptozotocin (STZ)-induced diabetic mice. The results show that oral administration of NMN significantly increases the body and testis weight and the number of sperms. Moreover, the abnormal sperm count and the rate of sperm malformation are significantly decreased compared with the saline-treated diabetic mice. Histological analysis reveals that NMN treatment significantly increases the area and diameter of seminiferous tubules, accompanied by an increased number of spermatogenic cells and sperms. Immunohistochemistry and qRT-PCR results show that NMN increases Bcl-2 expression and decreases Bax expression in the testis. NMN also increases the protein expression of Vimentin and the mRNA expressions of WT1 and GATA4. In addition, qRT-PCR, western blot analysis and immunohistochemistry results also show that NMN increases the expressions of glycolysis-related rate-limiting enzymes including HK2, PKM2, and LDHA. In summary, this study demonstrates the protective effects of NMN on the testis in an STZ-induced diabetic mice model. NMN exerts its protective effects via reducing spermatogenic cell apoptosis by regulating glycolysis of Sertoli cells in diabetic mice. This study provides an experimental basis for the future clinical application of NMN in diabetes-induced spermatogenic dysfunction.

[1]  Suyun Li,et al.  Icariin improves testicular dysfunction via enhancing proliferation and inhibiting mitochondria-dependent apoptosis pathway in high-fat diet and streptozotocin-induced diabetic rats , 2021, Reproductive Biology and Endocrinology.

[2]  H. Tae,et al.  Ethanol Extract of Maclura tricuspidata Fruit Protects SH-SY5Y Neuroblastoma Cells against H2O2-Induced Oxidative Damage via Inhibiting MAPK and NF-κB Signaling , 2021, International journal of molecular sciences.

[3]  Yang Liu,et al.  KISS1/KISS1R mediates Sertoli cell apoptosis via the PI3K/AKT signalling pathway in a high‑glucose environment. , 2021, Molecular medicine reports.

[4]  Chang-cheng Zhang,et al.  Common markers of testicular Sertoli cells , 2021, Expert review of molecular diagnostics.

[5]  K. Cho,et al.  Nicotinamide Supplementation Improves Oocyte Quality and Offspring Development by Modulating Mitochondrial Function in an Aged Caenorhabditis elegans Model , 2021, Antioxidants.

[6]  R. Agarwal,et al.  Glutathione (GSH) improves sperm quality and testicular morphology in streptozotocin-induced diabetic mice , 2021, Asian journal of andrology.

[7]  F. Lu,et al.  Protective Effects of Hu-Lu-Ba-Wan (葫芦巴丸) against Oxidative Stress in Testis of Diabetic Rats through PKCα/NAPDH Oxidase Signaling Pathway , 2021, Chinese Journal of Integrative Medicine.

[8]  T. Kuloğlu,et al.  Expression of asprosin in rat hepatic, renal, heart, gastric, testicular and brain tissues and its changes in a streptozotocin-induced diabetes mellitus model. , 2020, Tissue & cell.

[9]  A. Mäkitie,et al.  Expression and Role of E-Cadherin, β-Catenin, and Vimentin in Human Papillomavirus–Positive and Human Papillomavirus–Negative Oropharyngeal Squamous Cell Carcinoma , 2020, The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society.

[10]  R. Rui,et al.  Nicotinamide Mononucleotide Supplementation Reverses the Declining Quality of Maternally Aged Oocytes. , 2020, Cell reports.

[11]  J. Rabinowitz,et al.  Lactate: the ugly duckling of energy metabolism , 2020, Nature Metabolism.

[12]  L. Hua,et al.  Long Non-Coding RNA DUXAP8 Facilitates Cell Viability, Migration, and Glycolysis in Non-Small-Cell Lung Cancer via Regulating HK2 and LDHA by Inhibition of miR-409-3p , 2020, OncoTargets and therapy.

[13]  M. Saghafi-Asl,et al.  The potential therapeutic effects of Lactobacillus plantarum and inulin on serum and testicular reproductive markers in diabetic male rats , 2020, Diabetology & Metabolic Syndrome.

[14]  Xiawei Wei,et al.  Nicotinamide Mononucleotide: A Promising Molecule for Therapy of Diverse Diseases by Targeting NAD+ Metabolism , 2020, Frontiers in Cell and Developmental Biology.

[15]  Zhongcheng Mo,et al.  Lycium barbarum Polysaccharides Improve Testicular Spermatogenic Function in Streptozotocin-Induced Diabetic Rats , 2020, Frontiers in Endocrinology.

[16]  A. Khaki,et al.  Evaluation of carvacrol on pituitary and sexual hormones and their receptors in the testicle of male diabetic rats , 2020, Human & experimental toxicology.

[17]  A. Ahmadi,et al.  Protective effects of Equisetum arvense methanolic extract on sperm characteristics and in vitro fertilization potential in experimental diabetic mice: An experimental study , 2020, International journal of reproductive biomedicine.

[18]  T. B. Mendes,et al.  Sertoli Cell Alterations in Peripubertal Varicocelized Rats: Evidence of Primary Damage on Spermatogenesis , 2020, The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society.

[19]  B. Ma,et al.  Protective effect of Dioscorea zingiberensis ethanol extract on the disruption of blood–testes barrier in high‐fat diet/streptozotocin‐induced diabetic mice by upregulating ZO‐1 and Nrf2 , 2020, Andrologia.

[20]  P. Zheng,et al.  Protective effects of Salidroside on spermatogenesis in streptozotocin induced type-1 diabetic male mice by inhibiting oxidative stress mediated blood-testis barrier damage. , 2020, Chemico-biological interactions.

[21]  Wei Yu,et al.  Metformin Ameliorates Testicular Damage in Male Mice with Streptozotocin-Induced Type 1 Diabetes through the PK2/PKR Pathway , 2019, Oxidative medicine and cellular longevity.

[22]  Shuai Xiao,et al.  [Role and regulatory mechanism of glycometabolism of Sertoli cells in spermatogenesis]. , 2019 .

[23]  Eman Said,et al.  A comparative study of the role of crocin and sitagliptin in attenuation of STZ-induced diabetes mellitus and the associated inflammatory and apoptotic changes in pancreatic β-islets. , 2019, Environmental toxicology and pharmacology.

[24]  Jia Wang,et al.  High temperature suppressed SSC self-renewal through S phase cell cycle arrest but not apoptosis , 2019, Stem Cell Research & Therapy.

[25]  M. Salahshoor,et al.  Effects of Crocin on Sperm Parameters and Seminiferous Tubules in Diabetic Rats , 2019, Advanced biomedical research.

[26]  A. Letai,et al.  Regulation of apoptosis in health and disease: the balancing act of BCL-2 family proteins , 2019, Nature Reviews Molecular Cell Biology.

[27]  Sanjana Haque,et al.  Nicotinamide Mononucleotide: Exploration of Diverse Therapeutic Applications of a Potential Molecule , 2019, Biomolecules.

[28]  S. Salimi,et al.  Genetic and epigenetic analysis of the BAX and BCL2 in the placenta of pregnant women complicated by preeclampsia , 2019, Apoptosis.

[29]  H. Lane,et al.  Dual Inhibition of the Lactate Transporters MCT1 and MCT4 Is Synthetic Lethal with Metformin due to NAD+ Depletion in Cancer Cells , 2018, Cell reports.

[30]  Z. Kong,et al.  Modulation of Diabetes Mellitus-Induced Male Rat Reproductive Dysfunction with Micro-Nanoencapsulated Echinacea purpurea Ethanol Extract , 2018, BioMed research international.

[31]  Xiaofang Lu,et al.  Hyperglycemia induced testicular damage in type 2 diabetes mellitus rats exhibiting microcirculation impairments associated with vascular endothelial growth factor decreased via PI3K/Akt pathway , 2018, Oncotarget.

[32]  J. Baur,et al.  NAD+ Intermediates: The Biology and Therapeutic Potential of NMN and NR. , 2017, Cell metabolism.

[33]  Yang Du,et al.  Effect of varicocelectomy treatment on spermatogenesis and apoptosis via the induction of heat shock protein 70 in varicocele-induced rats , 2017, Molecular medicine reports.

[34]  C. Hao,et al.  Nicotinamide Mononucleotide, an NAD+ Precursor, Rescues Age-Associated Susceptibility to AKI in a Sirtuin 1-Dependent Manner. , 2017, Journal of the American Society of Nephrology : JASN.

[35]  M. I. Sousa,et al.  High glucose levels affect spermatogenesis: an in vitro approach. , 2017, Reproduction, fertility, and development.

[36]  S. Imai,et al.  Long-Term Administration of Nicotinamide Mononucleotide Mitigates Age-Associated Physiological Decline in Mice. , 2016, Cell metabolism.

[37]  Luís Rato,et al.  White tea intake prevents prediabetes-induced metabolic dysfunctions in testis and epididymis preserving sperm quality. , 2016, The Journal of nutritional biochemistry.

[38]  M. Alves,et al.  Pioglitazone increases the glycolytic efficiency of human Sertoli cells with possible implications for spermatogenesis. , 2016, The international journal of biochemistry & cell biology.

[39]  T. Sakurai,et al.  Nicotinamide mononucleotide protects against β-amyloid oligomer-induced cognitive impairment and neuronal death , 2016, Brain Research.

[40]  Dany Va,et al.  Diabetes Mellitus, Testicular Damages and Seafood: A Curious Relationship , 2016 .

[41]  J. Toppari,et al.  GATA4 Regulates Blood-Testis Barrier Function and Lactate Metabolism in Mouse Sertoli Cells. , 2016, Endocrinology.

[42]  B. Furman,et al.  Streptozotocin‐Induced Diabetic Models in Mice and Rats , 2015, Current protocols in pharmacology.

[43]  M. Alves,et al.  Metabolic fingerprints in testicular biopsies from type 1 diabetic patients , 2015, Cell and Tissue Research.

[44]  Luís Rato,et al.  Testicular Metabolic Reprogramming in Neonatal Streptozotocin-Induced Type 2 Diabetic Rats Impairs Glycolytic Flux and Promotes Glycogen Synthesis , 2015, Journal of diabetes research.

[45]  G. Ding,et al.  The effects of diabetes on male fertility and epigenetic regulation during spermatogenesis , 2015, Asian journal of andrology.

[46]  T. Kadowaki,et al.  l-cysteine reversibly inhibits glucose-induced biphasic insulin secretion and ATP production by inactivating PKM2 , 2015, Proceedings of the National Academy of Sciences.

[47]  L. Cai,et al.  Protection by sulforaphane from type 1 diabetes-induced testicular apoptosis is associated with the up-regulation of Nrf2 expression and function. , 2014, Toxicology and applied pharmacology.

[48]  Z. Xin,et al.  Prophylactic Protective Effects and Its Potential Mechanisms of IcarisideII on Streptozotocin Induced Spermatogenic Dysfunction , 2014, International journal of molecular sciences.

[49]  D. Sinclair,et al.  SIRT2 induces the checkpoint kinase BubR1 to increase lifespan , 2014, The EMBO journal.

[50]  M. Maiorino,et al.  Diabetes and sexual dysfunction: current perspectives , 2014, Diabetes, metabolic syndrome and obesity : targets and therapy.

[51]  P. Moreira,et al.  Metformin and male reproduction: effects on Sertoli cell metabolism , 2014, British journal of pharmacology.

[52]  P. Kempler,et al.  Sexual dysfunction in diabetes. , 2014, Handbook of clinical neurology.

[53]  T. Jiang,et al.  The Wilms Tumor Gene, Wt1, Is Critical for Mouse Spermatogenesis via Regulation of Sertoli Cell Polarity and Is Associated with Non-Obstructive Azoospermia in Humans , 2013, PLoS genetics.

[54]  Luís Rato,et al.  Molecular mechanisms beyond glucose transport in diabetes-related male infertility. , 2013, Biochimica et biophysica acta.

[55]  M. Alves,et al.  Diabetes, insulin-mediated glucose metabolism and Sertoli/blood-testis barrier function , 2013, Tissue barriers.

[56]  Erwin Goldberg,et al.  Lactate Dehydrogenase C and Energy Metabolism in Mouse Sperm1 , 2011, Biology of reproduction.

[57]  N. Mellen,et al.  Exacerbation of diabetes-induced testicular apoptosis by zinc deficiency is most likely associated with oxidative stress, p38 MAPK activation, and p53 activation in mice. , 2011, Toxicology letters.

[58]  J. Loscalzo,et al.  High glucose inhibits glucose‐6‐phosphate dehydrogenase, leading to increased oxidative stress and β‐cell apoptosis , 2010, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[59]  S. Imai,et al.  Age‐associated loss of Sirt1‐mediated enhancement of glucose‐stimulated insulin secretion in beta cell‐specific Sirt1‐overexpressing (BESTO) mice , 2008, Aging cell.

[60]  J. Milbrandt,et al.  Nampt/PBEF/Visfatin regulates insulin secretion in beta cells as a systemic NAD biosynthetic enzyme. , 2007, Cell metabolism.

[61]  T. Itano,et al.  Altered distribution of Sertoli cell vimentin and increased apoptosis in cryptorchid rats. , 2002, Journal of pediatric surgery.