Skip to main content

Advertisement

Log in

N,N-Dimethylformamide Delays LPS-Induced Preterm Birth in a Murine Model by Suppressing the Inflammatory Response

  • Pregnancy: Original Article
  • Published:
Reproductive Sciences Aims and scope Submit manuscript

Abstract

Preterm birth accounts for the majority of perinatal mortality worldwide, and there remains no FDA-approved drug to prevent it. Recently, we discovered that the common drug excipient, N,N-dimethylacetamide (DMA), delays inflammation-induced preterm birth in mice by inhibiting NF-κB. Since we reported this finding, it has come to light that a group of widely used, structurally related aprotic solvents, including DMA, N-methyl-2-pyrrolidone (NMP) and dimethylformamide (DMF), have anti-inflammatory efficacy. We show here that DMF suppresses LPS-induced TNFα secretion from RAW 264.7 cells and IL-6 and IL-8 secretion from HTR-8 cells at concentrations that do not significantly affect cell viability. Like DMA, DMF protects IκBα from degradation and prevents the p65 subunit of NF-κB from translocating to the nucleus. In vivo, DMF decreases LPS-induced inflammatory cell infiltration and expression of TNFα and IL-6 in the placental labyrinth, all to near baseline levels. Finally, DMF decreases the rate of preterm birth in LPS-induced pregnant mice (P<.0001) and the rate at which pups are spontaneously aborted (P<.0001). In summary, DMF, a widely used solvent structurally related to DMA and NMP, delays LPS-induced preterm birth in a murine model without overt toxic effects. Re-purposing the DMA/DMF/NMP family of small molecules as anti-inflammatory drugs is a promising new approach to delaying or reducing the incidence of inflammation-induced preterm birth and potentially attenuating other inflammatory disorders as well.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Data Availability

The data that support the findings in this study are available from the corresponding author upon reasonable request.

Code Availability

N/A.

References

  1. Liu L, Oza S, Hogan D, et al. Global, regional, and national causes of under-5 mortality in 2000-15: An updated systematic analysis with implications for the Sustainable Development Goals. Lancet. 2016;388:3027–35.

    PubMed  PubMed Central  Google Scholar 

  2. Blencowe H, Cousens S, Oestergaard M, Chou D, Moller AB, Narwal R, Adler A, Garcia CV, Rohde S, Say L, Lawn JE. National, regional and worldwide estimates of preterm birth. Lancet. 2012;379:2162–72.

    PubMed  Google Scholar 

  3. Martin JA, Osterman MJK. Describing the increase in preterm births in the United States, 2014–2016. National Center for Health Statistics 2018;Data Brief No. 312.

  4. Kerkhof G, Breukhoven PE, Leunissen RW, Willemsen RH, Hokken-Koelega AC. Does preterm birth influence cardiovascular risk in early adulthood? J Pediatr. 2012;161:390–6.

    PubMed  Google Scholar 

  5. Nuyt A, Lavoie JC, Mohamed I, Paquette K, Luu TM. Adult consequences of extremely preterm birth: cardiovascular and metabolic diseases risk factors, mechanisms, and prevention avenues. Clin Perinatol. 2017;44:315–32.

    PubMed  Google Scholar 

  6. Peelen MJ, Luef BM, Lamont RF, et al. The influence of the vaginal microbiota on preterm birth: A systematic review and recommendations for a minimum dataset for future research. Placenta. 2019;79:30–9.

    PubMed  Google Scholar 

  7. Manuel CR, Latuga MS, Ashby CR, Reznik SE. Immune tolerance attenuates gut dysbiosis, dysregulated uterine gene expression and high-fat diet potentiated preterm birth in mice. Am J Obstet Gynecol. 2019;220(596):e1–28.

    Google Scholar 

  8. Salomon C, Nuzhat Z, Dixon CL, Menon R. Placental exosomes during gestation: liquid biopsies carrying signals for the regulation of human parturition. Curr Pharm Des. 2018;24:974–82.

    CAS  PubMed  Google Scholar 

  9. Menon R. Initiation of human parturition: signaling from senescent fetal tissues via extracellular vesicle mediated paracrine mechanism. Obstet Gynecol Sci. 2019;62:199–211.

    PubMed  PubMed Central  Google Scholar 

  10. Keelan J. Intrauterine inflammatory activation, functional progesterone withdrawal, and the timing of term and preterm birth. J Reprod Immunol. 2017;125:89–99.

    PubMed  Google Scholar 

  11. Manuck TA. 17-alpha hydroxyprogesterone caproate for preterm birth prevention: Where have be been, how did we get here, and where are we going? Semin Perinatol. 2017;41:461–7.

    PubMed  Google Scholar 

  12. Caritis SN, Hauspurg A, Venkataramanan R, Lemon L. Defining the clinical response to 17-alpha-hydroxyprogesterone caproate. Am J Obstet Gynecol. 2018;219:623–5.

    PubMed  Google Scholar 

  13. O’Brien JM, Lewis DF. Prevention of preterm birth with vaginal progesterone or 17-alpha-hydroxyprogesterone caproate: A critical examination of safety and efficacy. Am J Obstet Gynecol. 2016;214:45–56.

    PubMed  Google Scholar 

  14. Heyborne KD, Allshouse AA, Carey JC. Does 17-alpha hydroxyprogesterone caproate prevent preterm birth in obese women? Am J Obstet Gynecol. 2015;213(844):e1–6.

    Google Scholar 

  15. Co AL, Walker HC, Hade EM, Iams JD. Relation of body mass index to frequency of recurrent preterm birth in women treated with 17-alpha hydroxyprogesterone caproate. Am J Obstet Gynecol. 2015;213(233):e1–5.

    Google Scholar 

  16. Lee LM, Liu LY, Sakowicz A, Bolden JR, Miller ES. Racial and ethnic disparities in use of 17-alpha hydroxyprogesterone caproate for prevention of preterm birth. Am J Obstet Gynecol. 2016;214(374):e1–5.

    Google Scholar 

  17. Timofeev J, Singh J, Istwan N, Rhea D, Driggers RW. Spontaneous preterm birth in African American and Caucasian women receiving 17-alpha hydroxyprogesterone caproate. J Matern Fetal Neonatal Med. 2013;26:881–4.

    CAS  PubMed  Google Scholar 

  18. NCD Risk Factor Collaboration. Trends in adult body-mass index in 200 countries from 1975-2014: A pooled analysis of 1698 population-based measurement studies with 19.2 million participants. Lancet. 2016;387:1377–96.

    Google Scholar 

  19. Mohamed SA, Thota C, Browne PC, Diamond MP, Al-Hendy A. Why is preterm birth stubbornly higher in African-Americans? Obstet Gynecol Int J. 2015;1:00019.

    Google Scholar 

  20. Blackwell SC, Gyamfi-Bannerman C, Biggio JR, et al. 17-OHPC to prevent recurrent preterm birth in singleton gestations (PROLONG study): A multicenter, international, randomized double-blind trial. Am J Perinatol. 2019;37:127–36.

    PubMed  Google Scholar 

  21. Sundaram S, Ashby CR, Pekson R, et al. N, N-Dimethylacetamide regulates the pro-inflammatory response associated with endotoxin and prevents preterm birth. Am J Pathol. 2013;183:422–30.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Pekson R, Poltoratsky V, Gorasiya S, Sundaram S, Ashby CR, Vancurova I, Reznik SE. N,N-Dimethylacetamide significantly attenuates LPS- and TNFα-induced proinflammatory responses via inhibition of the nuclear factor kappa B pathway. Mol Med. 2016;22:747–58.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Ghayor C, Gjoksi B, Dong J, Siegenthaler B, Caflisch A, Weber FE. N,N-Dimethylacetamide a drug excipient that acts as bromodomain ligand for osteoporosis treatment. Sci Rep. 2017;7:42108.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Chen TH, Weber FE, Malina-Altzinger J, Ghayor C. Epigenetic drugs as new therapy for tumor necrosis factor-α-compromised bone healing. Bone. 2019;127:49–58.

    CAS  PubMed  Google Scholar 

  25. Vyas V, Ashby CR, Olgun NS, et al. Inhibition of sphingosine kinase prevents lipopolysacccharide induced preterm birth and suppresses pro-inflammatory responses in a murine model. Am J Pathol. 2015;185:862–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Williams L, Burgos E, Vuguin P, et al. N-Acetylcysteine resolves placental inflammatory-vasculopathic changes in mice consuming a high fat diet. Am J Path. 2019;189:2246–57.

  27. Munnangi S, Gross SJ, Madankumar R, Reznik SE. Pregnancy associated plasma protein-A2: A novel biomarker for Down syndrome. Placenta. 2014;35:900–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Novembri R, De Clemente C, Funghi L, Torricelli M, Voltolini C, Challis JR, Petraglia F. Corticotropin releasing hormone and Urocortin 2 activate inflammatory pathways in cultured trophoblast cell line. Eur J Obstet Gynecol Reprod Biol. 2019;195:200–5.

    Google Scholar 

  29. Gomez R, Romero R, Ghezzi F, Yoon BH, Mazor M, Berry SM. The fetal inflammatory response syndrome. Am J Obstet Gynecol. 1998;179:194–202.

    CAS  PubMed  Google Scholar 

  30. Kadhim H, Tabarki B, Verellen G, De Prez C, Rona AM, Sébire G. Inflammatory cytokines in the pathogenesis of periventricular leukomalacia. Neurology. 2001;56:1278–84.

    CAS  PubMed  Google Scholar 

  31. Gisslen T, Singh G, Georgieff MK. Fetal inflammation is associated with persistent systemic and hippocampal inflammation and dysregulation of hippocampal glutamatergic homeostasis. Pediatr Res. 2019;85:703–10.

    PubMed  PubMed Central  Google Scholar 

  32. Muñoz-Pérez VM, Ortiz MI, Cariño-Cortés R, Fernández-Martínez E, Rocha-Zavaleta L, Bautista-Ávila M. Preterm birth, inflammation and infection: New alternative strategies for their prevention. Curr Pharm Biotechnol. 2019;20:354–65.

    PubMed  Google Scholar 

  33. Reznik SE. Editorial: Novel pharmacotherapeutic targets and emerging approaches to preterm birth, Part 1. Curr Pharm Des. 2017;23:6097.

    CAS  PubMed  Google Scholar 

  34. Reznik SE. Editorial: Novel pharmacotherapeutic targets and emerging approaches to preterm birth, Part 2. Curr Pharm Des. 2018;24:959.

    CAS  PubMed  Google Scholar 

  35. Manuel CR, Ashby CR, Reznik SE. Discrepancies in animal models of preterm birth. Curr Pharm Des. 2017;23:6142–8.

    CAS  PubMed  Google Scholar 

  36. Zierden HC, Ortiz JI, DeLong K, Yu J, et al. Enhanced drug delivery to the reproductive tract using nanomedicine reveals therapeutic options for prevention of preterm birth. Sci Transl Med. 2021;13:eabc6245.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Patki M, Giusto K, Gorasiya S, Patel K, Reznik SE. 17-α-Hydroxyprogesterone nanoemulsifying preconcentrate loaded vaginal tablet: A novel non-invasive approach for the prevention of preterm birth. Pharmaceutics. 2019;11:335. https://doi.org/10.3390/pharmaceutics11070335.

    Article  CAS  PubMed Central  Google Scholar 

  38. Giusto K, Patki M, Koya J, Ashby CR, Munnangi S, Patel K, Reznik SE. A vaginal nanoformulation of a sphingosine kinase inhibitor attenuates LPS-induced preterm birth in mice. Nanomedicine (London). 2019;14:2835–51.

    CAS  Google Scholar 

  39. Golia E, Limongelli G, Natale F, et al. Inflammation and cardiovascular disease: from pathogenesis to therapeutic target. Curr Atheroscler Rep. 2014;16:435.

    PubMed  Google Scholar 

  40. Dai L, Golembiewska E, Lindholm B, Stenvinkel P. Contrib Nephrol. 2017;191:32–43.

    CAS  PubMed  Google Scholar 

  41. Tuomisto AE, Mäkinen MJ, Väyrynen JP. Systemic inflammation in colorectal cancer: Underlying factors, effects and prognostic significance. World J Gastroenterol. 2019;25:4383–404.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Diakos CI, Charles KA, McMillan DC, Clarke SJ. Cancer-related inflammation and treatment effectiveness. Lancet Oncol. 2014;15:e493–503.

    PubMed  Google Scholar 

  43. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357:539–45.

    CAS  PubMed  Google Scholar 

  45. Adams B, Nunes JM, Page MJ, et al. Parkinson’s disease: A systemic inflammatory disease accompanied by bacterial inflammagens. Front Aging Neurosci. 2019;11:210.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Xu L, He D, Bai Y. Microglia-mediated inflammation and neurodegenerative disease. Mol Neurobiol. 2016;53:6709–15.

    CAS  PubMed  Google Scholar 

  47. Fischer R, Maier O. Interrelation of oxidative stress and inflammation in neurodegenerative disease: role of TNF. Oxidative Med Cell Longev. 2015;2015:610813.

    Google Scholar 

Download references

Acknowledgments

This work was funded by a grant from the National Institutes of Health to SER (1R01NS069577). We are grateful to Helen Scaramell and the entire staff of the St. John’s University Animal Care Center for maintenance of our mouse colony and assistance with in vivo protocols. We also thank Ms. Ernestine Middleton of Montefiore Medical Center, Bronx, NY, for her expert technical support in preparing the histology slides.

Author information

Authors and Affiliations

Authors

Contributions

Z.-H. W. and O.O.S. performed all of the experiments and contributed equally to this manuscript. J.K. assisted in the performance of the in vitro experiments. S.M. assisted in the performance of the in vivo experiments. R.P. assisted with the statistical analyses. C.R.A. contributed to the original idea of the project and assisted with the writing of the manuscript. S.E.R. contributed to the original idea of the project, wrote the manuscript and provided funding.

Corresponding author

Correspondence to Sandra E. Reznik.

Ethics declarations

Ethics Approval

Approval was obtained from the St. John’s University IACUC, assurance number D16-00346 (A3574-01).

Consent to Participate

N/A.

Consent for Publication

N/A.

Conflicts of Interest/Competing Interests

None of the authors has any conflicts to disclose.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplemental Figure 1.

DMF’s effect on cell viability. A, RAW 264.7 cells and B, HTR-8 cells were incubated with or without concentrations of DMF ranging from 0.1 to 40 mM. Cell viability was assessed using the MTT assay. The double asterisks indicate P<0.01 and the quadruple asterisks indicate P<.0001, as compared to the control group. HTR-8, HTR-8/SVNeo; DMF, N,N-dimethylformamide; LPS, lipopolysaccharide; MTT, 3-(4,5-dimethyl thiazolyl-2)-2,5-diphenyl tetrazolium bromide. (DOCX 3647 kb)

Supplementary file1 (DOCX 43 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wei, ZH., Salami, O.O., Koya, J. et al. N,N-Dimethylformamide Delays LPS-Induced Preterm Birth in a Murine Model by Suppressing the Inflammatory Response. Reprod. Sci. 29, 2894–2907 (2022). https://doi.org/10.1007/s43032-022-00924-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s43032-022-00924-z

Keywords

Navigation