1
|
Wei ZH, Koya J, Acharekar N, Trejos J, Dong XD, Schanne FA, Ashby CR, Reznik SE. N,N-dimethylacetamide targets neuroinflammation in Alzheimer's disease in in-vitro and ex-vivo models. Sci Rep 2023; 13:7077. [PMID: 37127686 PMCID: PMC10151369 DOI: 10.1038/s41598-023-34355-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/28/2023] [Indexed: 05/03/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic degenerative brain disorder with no clear pathogenesis or effective cure, accounting for 60-80% of cases of dementia. In recent years, the importance of neuroinflammation in the pathogenesis of AD and other neurodegenerative disorders has come into focus. Previously, we made the serendipitous discovery that the widely used drug excipient N,N-dimethylacetamide (DMA) attenuates endotoxin-induced inflammatory responses in vivo. In the current work, we investigate the effect of DMA on neuroinflammation and its mechanism of action in in-vitro and ex-vivo models of AD. We show that DMA significantly suppresses the production of inflammatory mediators, such as reactive oxygen species (ROS), nitric oxide (NO) and various cytokines and chemokines, as well as amyloid-β (Aβ), in cultured microglia and organotypic hippocampal slices induced by lipopolysaccharide (LPS). We also demonstrate that DMA inhibits Aβ-induced inflammation. Finally, we show that the mechanism of DMA's effect on neuroinflammation is inhibition of the nuclear factor kappa-B (NF-κB) signaling pathway and we show how DMA dismantles the positive feedback loop between NF-κB and Aβ synthesis. Taken together, our findings suggest that DMA, a generally regarded as safe compound that crosses the blood brain barrier, should be further investigated as a potential therapy for Alzheimer's disease and neuroinflammatory disorders.
Collapse
Affiliation(s)
- Zeng-Hui Wei
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Jagadish Koya
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Nikita Acharekar
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Jesus Trejos
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Xing-Duo Dong
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Francis A Schanne
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sandra E Reznik
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA.
- Departments of Pathology and Obstetrics and Gynecology and Women's Health, The University Hospital for Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, 10461, USA.
| |
Collapse
|
2
|
Saurabh K, Mbadhi MN, Prifti KK, Martin KT, Frolova AI. Sphingosine 1-Phosphate Activates S1PR3 to Induce a Proinflammatory Phenotype in Human Myometrial Cells. Endocrinology 2023; 164:bqad066. [PMID: 37120767 PMCID: PMC10201982 DOI: 10.1210/endocr/bqad066] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/01/2023]
Abstract
One of the common mechanisms responsible for obstetric complications, affecting millions of women every year, is abnormal uterine contractility. Despite the critical importance of this process for women's health, the mechanisms of uterine contraction regulation remain poorly understood. The initiation of uterine smooth muscle (myometrial) contraction is an inflammatory process, accompanied by upregulation of proinflammatory genes and cytokine release. In this study, we show that sphingolipid metabolism is activated during human labor and that sphingosine 1-phosphate (S1P), the main bioactive sphingolipid, may modify the myometrial proinflammatory phenotype. Our data in both primary and immortalized human myometrial cells show that exogenous S1P induces a proinflammatory gene signature and upregulates the expression of known inflammatory markers of parturition, such as IL8 and COX2. Using expression of IL8 as a readout for S1P activity in myometrial cells, we established that these S1P effects are mediated through the activation of S1P receptor 3 (S1PR3) and downstream activation of ERK1/2 pathways. Inhibition of S1PR3 in human myometrial cells attenuates upregulation of IL8, COX2, and JUNB both at the mRNA and protein levels. Furthermore, activation of S1PR3 with a receptor-specific agonist recapitulated the effects seen after treatment with exogenous S1P. Collectively, these results suggest a signaling pathway activated by S1P in human myometrium during parturition and propose new targets for development of novel therapeutics to alter uterine contractility during management of preterm labor or labor dystocia.
Collapse
Affiliation(s)
- Kumar Saurabh
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Magdaleena Naemi Mbadhi
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kevin K Prifti
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kaci T Martin
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Antonina I Frolova
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
3
|
Taylor J, Sharp A, Rannard SP, Arrowsmith S, McDonald TO. Nanomedicine strategies to improve therapeutic agents for the prevention and treatment of preterm birth and future directions. NANOSCALE ADVANCES 2023; 5:1870-1889. [PMID: 36998665 PMCID: PMC10044983 DOI: 10.1039/d2na00834c] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/27/2023] [Indexed: 06/19/2023]
Abstract
The World Health Organisation (WHO) estimates 15 million babies worldwide are born preterm each year, with 1 million infant mortalities and long-term morbidity in survivors. Whilst the past 40 years have provided some understanding in the causes of preterm birth, along with development of a range of therapeutic options, notably prophylactic use of progesterone or uterine contraction suppressants (tocolytics), the number of preterm births continues to rise. Existing therapeutics used to control uterine contractions are restricted in their clinical use due to pharmacological drawbacks such as poor potency, transfer of drugs to the fetus across the placenta and maternal side effects from activity in other maternal systems. This review focuses on addressing the urgent need for the development of alternative therapeutic systems with improved efficacy and safety for the treatment of preterm birth. We discuss the application of nanomedicine as a viable opportunity to engineer pre-existing tocolytic agents and progestogens into nanoformulations, to improve their efficacy and address current drawbacks to their use. We review different nanomedicines including liposomes, lipid-based carriers, polymers and nanosuspensions highlighting where possible, where these technologies have already been exploited e.g. liposomes, and their significance in improving the properties of pre-existing therapeutic agents within the field of obstetrics. We also highlight where active pharmaceutical agents (APIs) with tocolytic properties have been used for other clinical indications and how these could inform the design of future therapeutics or be repurposed to diversify their application such as for use in preterm birth. Finally we outline and discuss the future challenges.
Collapse
Affiliation(s)
- Jessica Taylor
- Department of Chemistry, University of Liverpool Crown Street Liverpool L69 7ZD UK
| | - Andrew Sharp
- Harris-Wellbeing Preterm Birth Research Centre, Department of Women's and Children's Health, Liverpool Women's Hospital, University of Liverpool Crown Street Liverpool L8 7SS UK
| | - Steve P Rannard
- Department of Chemistry, University of Liverpool Crown Street Liverpool L69 7ZD UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool Liverpool L7 3NY UK
| | - Sarah Arrowsmith
- Department of Life Sciences, Manchester Metropolitan University Chester Street Manchester M1 5GD UK
| | - Tom O McDonald
- Department of Chemistry, University of Liverpool Crown Street Liverpool L69 7ZD UK
- Department of Materials, Henry Royce Institute, The University of Manchester Manchester M13 9PL UK
| |
Collapse
|
4
|
Wei ZH, Salami OO, Koya J, Munnangi S, Pekson R, Ashby CR, Reznik SE. N,N-Dimethylformamide Delays LPS-Induced Preterm Birth in a Murine Model by Suppressing the Inflammatory Response. Reprod Sci 2022; 29:2894-2907. [PMID: 35349119 DOI: 10.1007/s43032-022-00924-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
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.
Collapse
Affiliation(s)
- Zeng-Hui Wei
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, USA
| | | | - Jagadish Koya
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, USA
| | - Swapna Munnangi
- Department of Surgery, Nassau University Medical Center, Nassau, NY, USA
| | - Ryan Pekson
- Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, USA
| | - Sandra E Reznik
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, USA.
- Departments of Pathology and Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
5
|
Vaginal Nanoformulations for the Management of Preterm Birth. Pharmaceutics 2022; 14:pharmaceutics14102019. [PMID: 36297454 PMCID: PMC9611874 DOI: 10.3390/pharmaceutics14102019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 12/01/2022] Open
Abstract
Preterm birth (PTB) is a leading cause of infant morbidity and mortality in the world. In 2020, 1 in 10 infants were born prematurely in the United States. The World Health Organization estimates that a total of 15 million infants are born prematurely every year. Current therapeutic interventions for PTB have had limited replicable success. Recent advancements in the field of nanomedicine have made it possible to utilize the vaginal administration route to effectively and locally deliver drugs to the female reproductive tract. Additionally, studies using murine models have provided important insights about the cervix as a gatekeeper for pregnancy and parturition. With these recent developments, the field of reproductive biology is on the cusp of a paradigm shift in the context of treating PTB. The present review focuses on the complexities associated with treating the condition and novel therapeutics that have produced promising results in preclinical studies.
Collapse
|
6
|
Pavlidis I, Stock SJ. Preterm Birth Therapies to Target Inflammation. J Clin Pharmacol 2022; 62 Suppl 1:S79-S93. [PMID: 36106783 PMCID: PMC9545799 DOI: 10.1002/jcph.2107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/06/2022] [Indexed: 12/04/2022]
Abstract
Preterm birth (PTB; defined as delivery before 37 weeks of pregnancy) is the leading cause of morbidity and mortality in infants and children aged <5 years, conferring potentially devastating short- and long-term complications. Despite extensive research in the field, there is currently a paucity of medications available for PTB prevention and treatment. Over the past few decades, inflammation in gestational tissues has emerged at the forefront of PTB pathophysiology. Even in the absence of infection, inflammation alone can prematurely activate the main components of parturition resulting in uterine contractions, cervical ripening and dilatation, membrane rupture, and subsequent PTB. Mechanistic studies have identified critical elements of the complex inflammatory molecular pathways involved in PTB. Here, we discuss therapeutic options that target such key mediators with an aim to prevent, postpone, or treat PTB. We provide an overview of more traditional therapies that are currently used or being tested in humans, and we highlight recent advances in preclinical studies introducing novel approaches with therapeutic potential. We conclude that urgent collaborative action is required to address the unmet need of developing effective strategies to tackle the challenge of PTB and its complications.
Collapse
Affiliation(s)
- Ioannis Pavlidis
- University of Warwick Biomedical Research Unit in Reproductive HealthCoventryUK
| | | |
Collapse
|
7
|
Reznik SE. New hope for preventing preterm birth: The promise of vaginal nanoformulations. PLACENTA AND REPRODUCTIVE MEDICINE 2022; 1:107. [PMID: 36425736 PMCID: PMC9683351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Affiliation(s)
- Sandra E. Reznik
- Department of Pharmaceutical Sciences, St. John’s University, Queens 11439, NY, USA
- Departments of Pathology and Obstetrics and Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx 10461, NY, USA
| |
Collapse
|
8
|
Chen L, Li L, Song Y, Lv T. Blocking SphK1/S1P/S1PR1 Signaling Pathway Alleviates Lung Injury Caused by Sepsis in Acute Ethanol Intoxication Mice. Inflammation 2021; 44:2170-2179. [PMID: 34109517 PMCID: PMC8189277 DOI: 10.1007/s10753-021-01490-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/12/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022]
Abstract
Acute ethanol intoxication increases the risk of sepsis and aggravates the symptoms of sepsis and lung injury. Therefore, this study aimed to explore whether sphingosine kinase 1 (SphK1)/sphingosine-1-phosphate (S1P)/S1P receptor 1 (S1PR1) signaling pathway functions in lung injury caused by acute ethanol intoxication-enhanced sepsis, as well as its underlying mechanism. The acute ethanol intoxication model was simulated by intraperitoneally administering mice with 32% ethanol solution, and cecal ligation and puncture (CLP) was used to construct the sepsis model. The lung tissue damage was observed by hematoxylin-eosin (H&E) staining, and the wet-to-dry (W/D) ratio was used to evaluate the degree of pulmonary edema. Inflammatory cell counting and protein concentration in bronchoalveolar lavage fluid (BALF) were, respectively, detected by hemocytometer and bicinchoninic acid (BCA) method. The levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1β, and IL-18 in BALF were detected by their commercial enzyme-linked immunosorbent assay (ELISA) kits. The myeloperoxidase (MPO) activity and expression of apoptosis-related proteins and SphK1/S1P/S1PR1 pathway-related proteins were, respectively, analyzed by MPO ELISA kit and Western blot analysis. The cell apoptosis in lung tissues was observed by TUNEL assay. Acute ethanol intoxication (EtOH) decreased the survival rate of mice and exacerbated the lung injury caused by sepsis through increasing pulmonary vascular permeability, neutrophil infiltration, release of inflammatory factors, and cell apoptosis. In addition, EtOH could activate the SphK1/S1P/S1PR1 pathway in CLP mice. However, PF-543, as a specific inhibitor of SphK1, could partially reverse the deleterious effects on lung injury of CLP mice. PF-543 alleviated lung injury caused by sepsis in acute ethanol intoxication rats by suppressing the SphK1/S1P/S1PR1 signaling pathway.
Collapse
Affiliation(s)
- Liang Chen
- Department of Respiratory and Critical Care Medicine, The Affiliated No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu, China
| | - Lingling Li
- Department of Respiratory and Critical Care Medicine, The Affiliated No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu, China
| | - Yong Song
- Jinling Clinical Medical College, Nanjing Medical University, 305 Zhongshan East Road, Xuanwu District, Nanjing City, 210002, Jiangsu Province, China.
| | - Tangfeng Lv
- Jinling Clinical Medical College, Nanjing Medical University, 305 Zhongshan East Road, Xuanwu District, Nanjing City, 210002, Jiangsu Province, China.
| |
Collapse
|
9
|
Next generation strategies for preventing preterm birth. Adv Drug Deliv Rev 2021; 174:190-209. [PMID: 33895215 DOI: 10.1016/j.addr.2021.04.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/22/2022]
Abstract
Preterm birth (PTB) is defined as delivery before 37 weeks of gestation. Globally, 15 million infants are born prematurely, putting these children at an increased risk of mortality and lifelong health challenges. Currently in the U.S., there is only one FDA approved therapy for the prevention of preterm birth. Makena is an intramuscular progestin injection given to women who have experienced a premature delivery in the past. Recently, however, Makena failed a confirmatory trial, resulting the Center for Drug Evaluation and Research's (CDER) recommendation for the FDA to withdrawal Makena's approval. This recommendation would leave clinicians with no therapeutic options for preventing PTB. Here, we outline recent interdisciplinary efforts involving physicians, pharmacologists, biologists, chemists, and engineers to understand risk factors associated with PTB, to define mechanisms that contribute to PTB, and to develop next generation therapies for preventing PTB. These advances have the potential to better identify women at risk for PTB, prevent the onset of premature labor, and, ultimately, save infant lives.
Collapse
|
10
|
Coler BS, Shynlova O, Boros-Rausch A, Lye S, McCartney S, Leimert KB, Xu W, Chemtob S, Olson D, Li M, Huebner E, Curtin A, Kachikis A, Savitsky L, Paul JW, Smith R, Adams Waldorf KM. Landscape of Preterm Birth Therapeutics and a Path Forward. J Clin Med 2021; 10:2912. [PMID: 34209869 PMCID: PMC8268657 DOI: 10.3390/jcm10132912] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
Preterm birth (PTB) remains the leading cause of infant morbidity and mortality. Despite 50 years of research, therapeutic options are limited and many lack clear efficacy. Tocolytic agents are drugs that briefly delay PTB, typically to allow antenatal corticosteroid administration for accelerating fetal lung maturity or to transfer patients to high-level care facilities. Globally, there is an unmet need for better tocolytic agents, particularly in low- and middle-income countries. Although most tocolytics, such as betamimetics and indomethacin, suppress downstream mediators of the parturition pathway, newer therapeutics are being designed to selectively target inflammatory checkpoints with the goal of providing broader and more effective tocolysis. However, the relatively small market for new PTB therapeutics and formidable regulatory hurdles have led to minimal pharmaceutical interest and a stagnant drug pipeline. In this review, we present the current landscape of PTB therapeutics, assessing the history of drug development, mechanisms of action, adverse effects, and the updated literature on drug efficacy. We also review the regulatory hurdles and other obstacles impairing novel tocolytic development. Ultimately, we present possible steps to expedite drug development and meet the growing need for effective preterm birth therapeutics.
Collapse
Affiliation(s)
- Brahm Seymour Coler
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| | - Oksana Shynlova
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (O.S.); (A.B.-R.); (S.L.)
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON M5G 1E2, Canada
| | - Adam Boros-Rausch
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (O.S.); (A.B.-R.); (S.L.)
| | - Stephen Lye
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (O.S.); (A.B.-R.); (S.L.)
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON M5G 1E2, Canada
| | - Stephen McCartney
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
| | - Kelycia B. Leimert
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R7, Canada; (K.B.L.); (W.X.); (D.O.)
| | - Wendy Xu
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R7, Canada; (K.B.L.); (W.X.); (D.O.)
| | - Sylvain Chemtob
- Departments of Pediatrics, Université de Montréal, Montréal, QC H3T 1J4, Canada;
| | - David Olson
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R7, Canada; (K.B.L.); (W.X.); (D.O.)
- Departments of Pediatrics and Physiology, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Miranda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
- Department of Biological Sciencies, Columbia University, New York, NY 10027, USA
| | - Emily Huebner
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
| | - Anna Curtin
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
| | - Alisa Kachikis
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
| | - Leah Savitsky
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
| | - Jonathan W. Paul
- Mothers and Babies Research Centre, School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (J.W.P.); (R.S.)
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - Roger Smith
- Mothers and Babies Research Centre, School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (J.W.P.); (R.S.)
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
- John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| | - Kristina M. Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
11
|
Fakhr Y, Brindley DN, Hemmings DG. Physiological and pathological functions of sphingolipids in pregnancy. Cell Signal 2021; 85:110041. [PMID: 33991614 DOI: 10.1016/j.cellsig.2021.110041] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 01/12/2023]
Abstract
Signaling by the bioactive sphingolipid, sphingosine 1-phosphate (S1P), and its precursors are emerging areas in pregnancy research. S1P and ceramide levels increase towards end of gestation, suggesting a physiological role in parturition. However, high levels of circulating S1P and ceramide are correlated with pregnancy disorders such as preeclampsia, gestational diabetes mellitus and intrauterine growth restriction. Expression of placental and decidual enzymes that metabolize S1P and S1P receptors are also dysregulated during pregnancy complications. In this review, we provide an in-depth examination of the signaling mechanism of S1P and ceramide in various reproductive tissues during gestation. These factors determine implantation and early pregnancy success by modulating corpus luteum function from progesterone production to luteolysis through to apoptosis. We also highlight the role of S1P through receptor signaling in inducing decidualization and angiogenesis in the decidua, as well as regulating extravillous trophoblast migration to anchor the placenta into the uterine wall. Recent advances on the role of the S1P:ceramide rheostat in controlling the fate of villous trophoblasts and the role of S1P as a negative regulator of trophoblast syncytialization to a multinucleated placental barrier are discussed. This review also explores the role of S1P in anti-inflammatory and pro-inflammatory signaling, its role as a vasoconstrictor, and the effects of S1P metabolizing enzymes and receptors in pregnancy.
Collapse
Affiliation(s)
- Yuliya Fakhr
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2S2, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - David N Brindley
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada; Signal Transduction Research Group, Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S2, Canada; Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Denise G Hemmings
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2S2, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB T6G 2S2, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2S2, Canada.
| |
Collapse
|
12
|
Zhang J, Luo X, Huang C, Pei Z, Xiao H, Luo X, Huang S, Chang Y. Erythropoietin prevents LPS-induced preterm birth and increases offspring survival. Am J Reprod Immunol 2020; 84:e13283. [PMID: 32506750 PMCID: PMC7507205 DOI: 10.1111/aji.13283] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/13/2020] [Accepted: 06/01/2020] [Indexed: 12/17/2022] Open
Abstract
PROBLEM Preterm delivery is the leading cause of neonatal mortality and contributes to delayed physical and cognitive development in children. At present, there is no efficient therapy to prevent preterm labor. A large body of evidence suggests that infections might play a significant and potentially preventable cause of premature birth. This work assessed the effects of erythropoietin (EPO) in a murine model of inflammation-associated preterm delivery, which mimics central features of preterm infections in humans. METHOD OF STUDY BALB/c mice were injected i.p. with 20 000 IU/kg EPO or normal saline twice on gestational day (GD) 15, with a 3 hours time interval between injections. An hour after the first EPO or normal saline injection, all mice received two injections of 50 μg/kg LPS, also given 3 hours apart. RESULTS EPO significantly prevented preterm labor and increased offspring survival in an LPS induced preterm delivery model. EPO prevented LPS-induced leukocyte infiltration into the placenta. Moreover, EPO inhibited the expression of pro-inflammatory cytokines, interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumour necrosis factor-α (TNF-α) in maternal serum and amniotic fluid. EPO also prevented LPS-induced increase in placental prostaglandin (PG)E2 and uterine inducible nitric oxide synthase (iNOS) production, while decreasing nuclear factor kappa-B (NF-κβ) activity in the myometrium. EPO also increased the gene expression of placental programmed cell death ligand 1 (PD-L1) in LPS-treated mice. CONCLUSIONS Our results suggest that EPO could be a potential novel therapeutic strategy to tackle infection-related preterm labor.
Collapse
Affiliation(s)
- Jie Zhang
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Xianqiong Luo
- Department of PediatricsGuangdong Women and Children HospitalGuangzhouChina
| | - Caicai Huang
- Department of ObstetricsGuangdong Women and Children HospitalGuangzhouChina
| | - Zheng Pei
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Huimei Xiao
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Xingang Luo
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Shuangmiao Huang
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Yanqun Chang
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| |
Collapse
|
13
|
Giusto K, Patki M, Koya J, Ashby CR, Munnangi S, Patel K, Reznik SE. A vaginal nanoformulation of a SphK inhibitor attenuates lipopolysaccharide-induced preterm birth in mice. Nanomedicine (Lond) 2019; 14:2835-2851. [PMID: 31793846 DOI: 10.2217/nnm-2019-0243] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/30/2019] [Indexed: 12/19/2022] Open
Abstract
Aim: Previously, we have shown that inhibition of SphK by the SphK inhibitor-II (SKI II) prevents lipopolysaccharide-induced preterm birth in mice. The aim of this study was to develop a vaginal self-nanoemulsifying drug-delivery system (SNEDDS) for SKI II. Materials & methods: A SKI II-loaded SNEDDS was characterized and tested in a murine preterm birth model. Results: The SNEDDS immediately formed a gel and then slowly emulsified to nanoglobules with over 500-fold enhancement of SKI II solubility at vaginal pH. Intravaginal administration of the SKI II SNEDDS significantly decreased lipopolysaccharide-induced preterm birth in mice. Conclusion: A vaginal nanoformulation of SKI II represents a novel, noninvasive approach to prevent preterm birth.
Collapse
Affiliation(s)
- Kiersten Giusto
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St John's University, Queens, NY 11439, USA
| | - Manali Patki
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St John's University, Queens, NY 11439, USA
| | - Jagadish Koya
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St John's University, Queens, NY 11439, USA
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St John's University, Queens, NY 11439, USA
| | - Swapna Munnangi
- Department of Surgery, Nassau University Medical Center, East Meadow, NY 11544, USA
| | - Ketan Patel
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St John's University, Queens, NY 11439, USA
| | - Sandra E Reznik
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St John's University, Queens, NY 11439, USA
- Departments of Pathology, Obstetrics, Gynecology & Women's Health, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
14
|
Yang X, Ye A, Chen L, Xia Y, Jiang W, Sun W. Involvement of calcium in 50-Hz magnetic field-induced activation of sphingosine kinase 1 signaling pathway. Bioelectromagnetics 2019; 40:180-187. [PMID: 30920672 DOI: 10.1002/bem.22181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/26/2019] [Indexed: 01/09/2023]
Abstract
Previously, we found that exposure to a 50-Hz magnetic field (MF) could induce human amniotic epithelial (FL) cell proliferation and sphingosine kinase 1 (SK1) activation, but the mechanism was not clearly understood. In the present study, the possible signaling pathways which were involved in SK1 activation induced by 50-Hz MF exposure were investigated. Results showed that MF exposure increased intracellular Ca2+ which was dependent on the L-type calcium channel, and induced Ca2+ -dependent phosphorylation of extracellular regulated protein kinase (ERK), SK1, and protein kinase C α (PKCα). Also, treatment with U0126, an inhibitor of ERK, could block MF-induced SK1 phosphorylation, but had no effect on PKCα phosphorylation. Also, the inhibitor of PKCα, Gö6976, had no effect on MF-induced SK1 activation in FL cells. In addition, the activation of ERK and PKCα could be abolished by SKI II, the inhibitor of SK1. In conclusion, the intracellular Ca2+ mediated the 50-Hz MF-induced SK1 activation which enhanced PKCα phosphorylation, and there might be a feedback mechanism between SK1 and ERK activation in responding to MF exposure in FL cells. Bioelectromagnetics. 9999:XX-XX, 2019. © 2019 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Xiaobo Yang
- Bioelectromagnetics Key Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Anfang Ye
- Department of Occupational Disease of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liangjing Chen
- Bioelectromagnetics Key Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yongpeng Xia
- Bioelectromagnetics Key Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wei Jiang
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenjun Sun
- Bioelectromagnetics Key Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Occupational Disease of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
17-α Hydroxyprogesterone Nanoemulsifying Preconcentrate-Loaded Vaginal Tablet: A Novel Non-Invasive Approach for the Prevention of Preterm Birth. Pharmaceutics 2019; 11:pharmaceutics11070335. [PMID: 31337153 PMCID: PMC6680947 DOI: 10.3390/pharmaceutics11070335] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 12/26/2022] Open
Abstract
Preterm birth (PTB) is a major cause of infant mortality in the United States and around the globe. Makena®—once-a-week intramuscular injection of 17-α Hydroxyprogesterone caproate (17P)—is the only FDA approved treatment for the prevention of PTB. Invasive delivery of 17P requires hospitalization and expert personnel for injection. Vaginal delivery of 17P would be preferable, because of high patient compliance, reduced systemic exposure, fewer side effects, and no need for hospitalization. The objective of the present study was to prepare and evaluate a self-nanoemulsifying vaginal tablet of 17P. A solid self-nanoemulsifying preconcentrate (S-SNEDDS) of 17P and dimethylacetamide (DMA) was developed using medium chain triglycerides, a non- immunogenic surfactant, and co-processed excipient (PVA-F100). The tablet prepared was characterized for emulsification time, particle size, solid state properties, and drug release. The formulation showed >50% inhibition of TNF-α release from LPS-stimulated RAW 264.7 cells. Importantly, there were significant differences in rates of PTB and average time to delivery between control and vaginal 17P-treated groups in LPS-stimulated timed pregnant E15.5 mice. Considering the lacuna of therapeutic approaches in this area, vaginal delivery of 17P for the prevention of preterm birth has significant clinical relevance.
Collapse
|
16
|
Lee DK, Min YS, Yoo SS, Shim HS, Park SY, Sohn UD. Effect of Sphingosine-1-Phosphate on Intracellular Free Ca²⁺ in Cat Esophageal Smooth Muscle Cells. Biomol Ther (Seoul) 2018; 26:546-552. [PMID: 29915165 PMCID: PMC6254643 DOI: 10.4062/biomolther.2018.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 12/31/2022] Open
Abstract
A comprehensive collection of proteins senses local changes in intracellular Ca²⁺ concentrations ([Ca²⁺]i) and transduces these signals into responses to agonists. In the present study, we examined the effect of sphingosine-1-phosphate (S1P) on modulation of intracellular Ca²⁺ concentrations in cat esophageal smooth muscle cells. To measure [Ca²⁺]i levels in cat esophageal smooth muscle cells, we used a fluorescence microscopy with the Fura-2 loading method. S1P produced a concentration-dependent increase in [Ca²⁺]i in the cells. Pretreatment with EGTA, an extracellular Ca²⁺ chelator, decreased the S1P-induced increase in [Ca²⁺]i, and an L-type Ca²⁺-channel blocker, nimodipine, decreased the effect of S1P. This indicates that Ca²⁺ influx may be required for muscle contraction by S1P. When stimulated with thapsigargin, an intracellular calcium chelator, or 2-Aminoethoxydiphenyl borate (2-APB), an InsP3 receptor blocker, the S1P-evoked increase in [Ca²⁺]i was significantly decreased. Treatment with pertussis toxin (PTX), an inhibitor of Gi-protein, suppressed the increase in [Ca²⁺]i evoked by S1P. These results suggest that the S1P-induced increase in [Ca²⁺]i in cat esophageal smooth muscle cells occurs upon the activation of phospholipase C and subsequent release of Ca²⁺ from the InsP3-sensitive Ca²⁺ pool in the sarcoplasmic reticulum. These results suggest that S1P utilized extracellular Ca²⁺ via the L type Ca²⁺ channel, which was dependent on activation of the S1P4 receptor coupled to PTX-sensitive Gi protein, via phospholipase C-mediated Ca²⁺ release from the InsP3-sensitive Ca²⁺ pool in cat esophageal smooth muscle cells.
Collapse
Affiliation(s)
- Dong Kyu Lee
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06911, Republic of Korea
| | - Young Sil Min
- Department of Pharmaceutical Engineering, College of Convergence Science and Technology, Jung Won University, Goesan 28054, Republic of Korea
| | - Seong Su Yoo
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06911, Republic of Korea
| | - Hyun Sub Shim
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06911, Republic of Korea
| | - Sun Young Park
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06911, Republic of Korea
| | - Uy Dong Sohn
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06911, Republic of Korea
| |
Collapse
|
17
|
Qiu Z, Yuan H, Li N, Yang X, Hu X, Su F, Chen B. Bidirectional effects of moxifloxacin on the pro‑inflammatory response in lipopolysaccharide‑stimulated mouse peritoneal macrophages. Mol Med Rep 2018; 18:5399-5408. [PMID: 30365072 PMCID: PMC6236266 DOI: 10.3892/mmr.2018.9569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 09/14/2018] [Indexed: 02/07/2023] Open
Abstract
Sepsis is a systemic inflammatory condition in response to life-threatening infections, and macrophages are a key source of inflammatory cytokines. Moxifloxacin (MXF) has antibacterial activity in Gram-positive and Gram-negative bacteria. The present study investigated the effects of MXF on a lipopolysaccharide (LPS)-stimulated inflammatory response and gene expression in macrophages. Peritoneal macrophages were isolated from male C57BL/6J mice and treated with LPS and/or MXF. The mRNA and protein expression of toll-like receptor 4 (TLR4), sphingosine kinase 1 (SPHK1) and nuclear factor (NF)-κB was determined by quantitative polymerase chain reaction, western blotting and immunofluorescence analysis. The expression of tumor necrosis factor (TNF)-α and interleukin (IL)-6 was determined with ELISAs. The data demonstrated that MXF dose-dependently decreased the viability of macrophages, and 8 and 16 µg/ml MXF prevented the LPS-induced increase in TLR4, SPHK1, NF-κB p65, TNF-α and IL-6 expression. The inhibition was most effective at a concentration of 16 µg/ml MXF, whereas, 64 µg/ml MXF exerted a pro-inflammatory effect. Collectively, the data demonstrated a bidirectional effect of MXF: Lower MXF concentrations (8 and 16 µg/ml) inhibited the inflammatory response; however, a higher MXF concentration (64 µg/ml) had a pro-inflammatory effect on LPS-treated mouse peritoneal macrophages. In conclusion, these results suggested the importance of MXF as an inhibitor of the inflammatory response at an optimal dose. MXF inhibition of the inflammatory response may be mediated by TLR4 signaling.
Collapse
Affiliation(s)
- Zhenyu Qiu
- Department of Infectious Diseases, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hongxia Yuan
- Department of Infectious Diseases, The First Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Na Li
- Department of Infectious Diseases, The First Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Xinjuan Yang
- Department of Infectious Diseases, The First Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Xuemei Hu
- Department of Infectious Diseases, The First Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Fengtai Su
- Department of Infectious Diseases, The First Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Baiyi Chen
- Department of Infectious Diseases, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
18
|
Taguchi A, Yamashita A, Kawana K, Nagamatsu T, Furuya H, Inoue E, Osuga Y, Fujii T. Recent Progress in Therapeutics for Inflammation-Associated Preterm Birth. Reprod Sci 2016; 24:7-18. [DOI: 10.1177/1933719115618282] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Ayumi Taguchi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- These authors contributed equally to this work
| | - Aki Yamashita
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- These authors contributed equally to this work
| | - Kei Kawana
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hitomi Furuya
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eri Inoue
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
19
|
Development of hydroxy-based sphingosine kinase inhibitors and anti-inflammation in dextran sodium sulfate induced colitis in mice. Bioorg Med Chem 2016; 24:3218-30. [DOI: 10.1016/j.bmc.2016.05.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/20/2016] [Accepted: 05/21/2016] [Indexed: 01/01/2023]
|
20
|
Migale R, Herbert BR, Lee YS, Sykes L, Waddington SN, Peebles D, Hagberg H, Johnson MR, Bennett PR, MacIntyre DA. Specific Lipopolysaccharide Serotypes Induce Differential Maternal and Neonatal Inflammatory Responses in a Murine Model of Preterm Labor. THE AMERICAN JOURNAL OF PATHOLOGY 2015. [PMID: 26212908 DOI: 10.1016/j.ajpath.2015.05.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Intrauterine inflammation is recognized as a key mediator of both normal and preterm birth but is also associated with neonatal neurological injury. Lipopolysaccharide (LPS) is often used to stimulate inflammatory pathways in animal models of infection/inflammation-induced preterm labor; however, inconsistencies in maternal and neonatal responses to LPS are frequently reported. We hypothesized that LPS serotype-specific responses may account for a portion of these inconsistencies. Four different Escherichia coli LPS serotypes (O111:B4, O55:B5, O127:B8, and O128:B12) were administered to CD1 mice via intrauterine injection at gestational day 16. Although control animals delivered at term 60 ± 15 hours postinjection (p.i.), those administered with O111:B4 delivered 7 ± 2 hours p.i., O55:B5 delivered 10 ± 3 hours p.i., O127:B8 delivered 16 ± 10 hours p.i., and O128:B12 delivered 17 ± 2 hours p.i. (means ± SD). A correlation between the onset of preterm labor and myometrial activation of the inflammatory transcription factor, activator protein 1, but not NF-κB was observed. Specific LPS serotypes induced differential activation of downstream contractile and inflammatory pathways in myometrium and neonatal pup brain. Our findings demonstrate functional disparity in inflammatory pathway activation in response to differing LPS serotypes. Selective use of LPS serotypes may represent a useful tool for targeting specific inflammatory response mechanisms in these models.
Collapse
Affiliation(s)
- Roberta Migale
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Bronwen R Herbert
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom; Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, London, United Kingdom
| | - Yun S Lee
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Lynne Sykes
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Simon N Waddington
- Gene Transfer Technology Group, Institute for Women's Health, University College London, London, United Kingdom; Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witswatersrand, Johannesburg, South Africa
| | - Donald Peebles
- UCL Centre for Perinatal Brain Protection & Repair, Institute for Women's Health, University College London, London, United Kingdom
| | - Henrik Hagberg
- Department of Clinical Sciences, Perinatal Center, University of Gothenburg, Gothenburg, Sweden; Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Mark R Johnson
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom; Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, London, United Kingdom
| | - Phillip R Bennett
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - David A MacIntyre
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom.
| |
Collapse
|