1
|
Mir A, Acosta T, Concheiro-Guisan M, Yellon SM, Patel K, Reznik SE. Improving the safety of N,N-dimethylacetamide (DMA) as a potential treatment for preterm birth in a pregnant mouse model using a vaginal nanoformulation. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167822. [PMID: 40174791 PMCID: PMC11994577 DOI: 10.1016/j.bbadis.2025.167822] [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: 11/20/2024] [Revised: 03/27/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Vaginal administration and the uterine first pass effect allow for preferential delivery of drugs to the reproductive tract. Dimethylacetamide has previously been shown to delay preterm birth in a pregnant mouse model when given intraperitoneally but the effectiveness of a vaginal nanoformulation of dimethylacetamide has yet to be tested. The purpose of this study was to compare the two formulations of dimethylacetamide for efficacy in rescuing pups from preterm birth in an inflammation-induced mouse model, effects on the maternal fetal interface, and pharmacokinetic profiles in maternal plasma. Timed pregnant CD1 mice were given a 1.56 mg/kg intraperitoneal dose of lipopolysaccharide followed by 3 doses of either vaginal dimethylacetamide or intraperitoneal dimethylacetamide. Mice were monitored for 48 h and times of deliveries were recorded. Additionally, CD1 mice in late gestation were given a single dose of either vaginal or intraperitoneal dimethylacetamide and blood was drawn at 3 different time points following administration. Vaginal administration of dimethylacetamide had similar efficacy in delaying inflammation induced preterm birth as intraperitoneal administration but resulted in lower concentrations in the systemic circulation and decreased effects on the maternal fetal interface. Vaginal nanoformulations should be explored for their potential therapeutic value for the delay of preterm birth.
Collapse
Affiliation(s)
- Asad Mir
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY 11439, USA
| | - Teeshavi Acosta
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY 11439, USA; Department of Sciences, John Jay College of Criminal Justice, City University of New York, 524 W 59th St, New York, NY 10019, USA
| | - Marta Concheiro-Guisan
- Department of Sciences, John Jay College of Criminal Justice, City University of New York, 524 W 59th St, New York, NY 10019, USA
| | - Steven M Yellon
- Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Ketan Patel
- 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; Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
2
|
Colon-Caraballo M, Russell SR, Myers KM, Mahendroo M. Collagen turnover during cervical remodeling involves both intracellular and extracellular collagen degradation pathways†. Biol Reprod 2025; 112:709-727. [PMID: 39823285 PMCID: PMC11996760 DOI: 10.1093/biolre/ioaf012] [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: 09/19/2024] [Revised: 12/20/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025] Open
Abstract
Reproductive success requires accurately timed remodeling of the cervix to orchestrate the maintenance of pregnancy, the process of labor, and birth. Prior work in mice established that a combination of continuous turnover of fibrillar collagen and reduced formation of collagen cross-links allows for the gradual increase in tissue compliance and delivery of the fetus during labor. However, the mechanism for continuous collagen degradation to ensure turnover during cervical remodeling is still unknown. This study demonstrates the functional role of extracellular and intracellular collagen degradative pathways in two different settings of cervical remodeling: physiological term remodeling and inflammation-mediated premature remodeling. Extracellular collagen degradation is achieved by the activity of fibroblast-derived matrix metalloproteases MMP14, MMP2, and fibroblast activation protein (FAP). In parallel, we demonstrate the function of an intracellular collagen degradative pathway in fibroblast cells mediated by the collagen endocytic mannose receptor type-2 (MRC2). These pathways appear to be functionally redundant as loss of MRC2 does not obstruct collagen turnover or cervical function in pregnancy. While both extracellular and intracellular pathways are also utilized in inflammation-mediated premature cervical remodeling, the extracellular collagen degradation pathway uniquely employs fibroblast and immune-cell-derived proteases. In sum, these findings identify the dual utilization of two distinct degradative pathways as a failsafe mechanism to achieve continuous collagen turnover in the cervix, thereby allowing dynamic shifts in cervical tissue mechanics and function.
Collapse
Affiliation(s)
- Mariano Colon-Caraballo
- Department of Ob/Gyn and Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Serena R Russell
- Department of Mechanical Engineering, Columbia University, New York, New York City, United States of America
| | - Kristin M Myers
- Department of Mechanical Engineering, Columbia University, New York, New York City, United States of America
| | - Mala Mahendroo
- Department of Ob/Gyn and Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
3
|
Madhukaran S, Fomina YY, Mahendroo M. Cervical function in pregnancy and disease: new insights from single-cell analysis. Am J Obstet Gynecol 2025; 232:S81-S94. [PMID: 40253084 DOI: 10.1016/j.ajog.2024.07.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 04/21/2025]
Abstract
The uterine cervix plays an essential role in regulating fertility, maintaining pregnancy, remodeling in preparation for parturition, and protecting the reproductive tract from infection. A compromise in cervical function contributes to adverse clinical outcomes. Understanding molecular events that drive the multifunctional and temporally defined roles of the cervix is necessary to effectively treat infertility, reproductive tract infections, preterm birth, labor dystocia, and cervical cancer. The application of single-cell technologies to study cervical pathophysiology, while in its infancy, underscores the potential of these approaches in developing clinically relevant biomarkers of disease and preventative therapies. This review focuses on insights gained from single-cell transcriptomic studies in human and mouse cervical tissue and highlights outstanding questions in the field. One collective advance from single-cell analysis is the dynamic plasticity of cervical epithelial cells during the reproductive cycle in health and disease. Single-cell comparisons between upper and lower regions of the reproductive tract also highlight the distinct and divergent immunological responses elicited in the cervix during the reproductive lifespan. These findings may reconcile prior controversies in the role of proinflammatory mediators during parturition. In addition to providing obstetric insights, single-cell technologies elucidate the molecular pathways that drive cervical cancer progression. Thus far, these technologies have uncovered cellular heterogeneity in the tumor microenvironment and have identified potential cancer stem cells. While single-cell technology alone will not uncover all the molecular underpinnings contributing to preterm birth or cervical cancer, the insights derived from this valuable technology will accelerate our understanding of cervical biology in health and disease, which ultimately will help develop biomarkers for disease prediction and prevention therapies.
Collapse
Affiliation(s)
- ShanmugaPriyaa Madhukaran
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yevgenia Y Fomina
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Mala Mahendroo
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
4
|
Jia S, Zhao F. Decoding Müllerian Duct Epithelial Regionalization. Mol Reprod Dev 2025; 92:e70018. [PMID: 39994938 PMCID: PMC11850963 DOI: 10.1002/mrd.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025]
Abstract
Müllerian ducts (MD), also known as paramesonephric ducts, are the primordial anlage of the female reproductive tract organs including the oviduct, uterus, cervix and upper vagina along the craniocaudal axis. Although the general architecture of MD-derived organs is conserved, each organ possesses their unique epithelial structures and cell types to confer their region-specific functions, which collectively coordinate successful fertilization and pregnancy. MD epithelial fate decisions and differentiation along the craniocaudal axis is dependent on spatiotemporal regulation of intrinsic transcription factors and extrinsic signals derived from the mesenchyme. Findings from genetic mouse models, single-cell sequencing studies, and organoid cultures have significantly advanced our understanding of the cellular and molecular mechanisms of MD regionalization. In this review, we first discuss the diversity of epithelial morphologies and cell types in the female reproductive tract organs. Then, we discuss the roles of key transcription factors (Hox, transcriptional cascade driving multiciliogenesis, Foxa2, and P63), signaling pathways (estrogen/ESR1, Wnt/β-catenin, hedgehog, and retinoic acid), and epigenetic factors (microRNAs, chromatin remodeling factors, and histone modification enzymes) in region-specific MD differentiation. Further deciphering molecular mechanisms of MD craniocaudal patterning will open new avenues to improve our strategies for prevention, diagnosis, and treatment of Müllerian anomalies and female reproductive tract disorders.
Collapse
Affiliation(s)
- Shuai Jia
- Department of Comparative Biosciences, School of Veterinary MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Fei Zhao
- Department of Comparative Biosciences, School of Veterinary MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| |
Collapse
|
5
|
Mir A, Acosta T, Concheiro-Guisan M, Yellon SM, Patel K, Reznik SE. Improving the Safety of N,N-Dimethylacetamide (DMA) as a Potential Treatment for Preterm Birth in a Pregnant Mouse Model Using a Vaginal Nanoformulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633348. [PMID: 39896642 PMCID: PMC11785104 DOI: 10.1101/2025.01.16.633348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Vaginal administration and the uterine first pass effect allow for preferential delivery of drugs to the reproductive tract. Dimethylacetamide has previously been shown to delay preterm birth in a pregnant mouse model when given intraperitoneally but the effectiveness of a vaginal nanoformulation of dimethylacetamide has yet to be tested. The purpose of this study was to compare the two formulations of dimethylacetamide for efficacy in rescuing pups from preterm birth in an inflammation-induced mouse model, effects on the maternal fetal interface, and pharmacokinetic profiles in maternal plasma. Timed pregnant CD1 mice were given a 1.56 mg/kg intraperitoneal dose of lipopolysaccharide followed by 3 doses of either vaginal dimethylacetamide or intraperitoneal dimethylacetamide. Mice were monitored for 48 hours and times of deliveries were recorded. Additionally, CD1 mice in late gestation were given a single dose of either vaginal or intraperitoneal dimethylacetamide and blood was drawn at 3 different time points following administration. Vaginal administration of dimethylacetamide had similar efficacy in delaying inflammation induced preterm birth as intraperitoneal administration but resulted in lower concentrations in the systemic circulation and decreased effects on the maternal fetal interface. Vaginal nanoformulations should be explored for their potential therapeutic value for the delay of preterm birth.
Collapse
Affiliation(s)
- Asad Mir
- Department of Pharmaceutical Sciences, St. John’s University, Queens, NY, 11439 USA
| | - Teeshavi Acosta
- Department of Pharmaceutical Sciences, St. John’s University, Queens, NY, 11439 USA
- Department of Sciences, John Jay College of Criminal Justice, City University of New York, 524 W 59 St, New York, NY, 10019 USA
| | - Marta Concheiro-Guisan
- Department of Sciences, John Jay College of Criminal Justice, City University of New York, 524 W 59 St, New York, NY, 10019 USA
| | - Steven M. Yellon
- Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92350
| | - Ketan Patel
- 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
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Obstetrics and Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx, NY, 10461 USA
| |
Collapse
|
6
|
Hansen CJ, Rogers JH, Brown AJ, Boatwright N, Siricilla S, O’Brien CM, Panja S, Nichols CM, Devanathan K, Hardy BM, Does MD, Anderson AW, Paria BC, Mahadevan-Jansen A, Reese J, Herington JL. Regional differences in three-dimensional fiber organization, smooth muscle cell phenotype, and contractility in the pregnant mouse cervix. SCIENCE ADVANCES 2024; 10:eadr3530. [PMID: 39693423 PMCID: PMC11654679 DOI: 10.1126/sciadv.adr3530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024]
Abstract
The orientation and function of smooth muscle in the cervix may contribute to the important biomechanical properties that change during pregnancy. Thus, this study examined the three-dimensional structure, smooth muscle phenotype, and mechanical and contractile functions of the upper and lower cervix of nongravid (not pregnant) and gravid (pregnant) mice. In gravid cervix, we uncovered region-specific changes in the structure and organization of fiber tracts. We also detected a greater proportion of contractile smooth muscle cells (SMCs), but an equal proportion of synthetic SMCs, in the upper versus lower cervix. Furthermore, we revealed that the lower cervix had infrequent spontaneous contractions, distension had a minimal effect on contractility, and the upper cervix had forceful contractions in response to labor-inducing agents (oxytocin and prostaglandin E2). These findings identify regional differences in cervix contractility related to contractile SMC content and fiber organization, which could be targeted with diagnostic technologies and for therapeutic intervention.
Collapse
Affiliation(s)
- Christopher J. Hansen
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Jackson H. Rogers
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexus J. Brown
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Naoko Boatwright
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shajila Siricilla
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christine M. O’Brien
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Sourav Panja
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cameron M. Nichols
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kanchana Devanathan
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Biophotonics Center, Vanderbilt University, Nashville TN USA
| | - Benjamin M. Hardy
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mark D. Does
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adam W. Anderson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bibhash C. Paria
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anita Mahadevan-Jansen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Biophotonics Center, Vanderbilt University, Nashville TN USA
- Department of Surgery, Neurological Surgery and Otolaryngology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Jeff Reese
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Jennifer L. Herington
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
7
|
Balan TA, Balan RA, Socolov D, Gheorghiță VR, Buțureanu TA, Păvăleanu I, Coșovanu ET, Căruntu ID. Pregnancy-Related Precancerous Cervical Lesions: Pathogenesis, Diagnosis, Evolution, and Impact upon Gestation and Fertility. J Clin Med 2024; 13:6718. [PMID: 39597862 PMCID: PMC11595265 DOI: 10.3390/jcm13226718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
More common than cervical cancer, cervical intraepithelial neoplasia (CIN) represents a precursor lesion of cervical carcinoma, being associated with HPV infection. Due to the bidirectional relationship between HPV and estrogen and progesterone in pregnancy, most of the published data claim that precancerous lesions remain stable or even regress during pregnancy, although several studies have indicated the tendency of HSILs to persist. It is considered that pregnancy-related cervical precancerous lesions undergo a postpartum regression, due to stimulatory effects of the immune microenvironment. Due to the rarity of publications on this subject, we aimed to offer a concise overview of and new insights into the current knowledge regarding the pathogenesis, diagnosis, and evolution of pregnancy-associated precancerous lesions, as well as their impact upon gestation and fertility.
Collapse
Affiliation(s)
- Teodora Ana Balan
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.A.B.); (D.S.); (T.A.B.); (I.P.); (E.T.C.); (I.-D.C.)
| | - Raluca Anca Balan
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.A.B.); (D.S.); (T.A.B.); (I.P.); (E.T.C.); (I.-D.C.)
- “Elena Doamna” Clinical Hospital of Obstetrics and Gynecology, 700398 Iasi, Romania
| | - Demetra Socolov
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.A.B.); (D.S.); (T.A.B.); (I.P.); (E.T.C.); (I.-D.C.)
- “Cuza Voda” Clinical Hospital of Obstetrics and Gynecology, 700038 Iasi, Romania
| | | | - Tudor Andrei Buțureanu
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.A.B.); (D.S.); (T.A.B.); (I.P.); (E.T.C.); (I.-D.C.)
- “Elena Doamna” Clinical Hospital of Obstetrics and Gynecology, 700398 Iasi, Romania
| | - Ioana Păvăleanu
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.A.B.); (D.S.); (T.A.B.); (I.P.); (E.T.C.); (I.-D.C.)
- “Elena Doamna” Clinical Hospital of Obstetrics and Gynecology, 700398 Iasi, Romania
| | - Elena Teona Coșovanu
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.A.B.); (D.S.); (T.A.B.); (I.P.); (E.T.C.); (I.-D.C.)
| | - Irina-Draga Căruntu
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.A.B.); (D.S.); (T.A.B.); (I.P.); (E.T.C.); (I.-D.C.)
- Department of Pathology, “Dr. C. I. Parhon” Clinical Hospital, 700503 Iasi, Romania
| |
Collapse
|
8
|
Ulibarri MR, Lin Y, Ramprashad JC, Han G, Hasan MH, Mithila FJ, Ma C, Gopinath S, Zhang N, Milner JJ, Beura LK. Epithelial organoid supports resident memory CD8 T cell differentiation. Cell Rep 2024; 43:114621. [PMID: 39153200 PMCID: PMC11401477 DOI: 10.1016/j.celrep.2024.114621] [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: 12/15/2023] [Revised: 07/01/2024] [Accepted: 07/26/2024] [Indexed: 08/19/2024] Open
Abstract
Resident memory T cells (TRMs) play a vital role in regional immune defense. Although laboratory rodents have been extensively used to study fundamental TRM biology, poor isolation efficiency and low cell survival rates have limited the implementation of TRM-focused high-throughput assays. Here, we engineer a murine vaginal epithelial organoid (VEO)-CD8 T cell co-culture system that supports CD8 TRM differentiation. These in-vitro-generated TRMs are phenotypically and transcriptionally similar to in vivo TRMs. Pharmacological and genetic approaches showed that transforming growth factor β (TGF-β) signaling plays a crucial role in their differentiation. The VEOs in our model are susceptible to viral infections and the CD8 T cells are amenable to genetic manipulation, both of which will allow a detailed interrogation of antiviral CD8 T cell biology. Altogether we have established a robust in vitro TRM differentiation system that is scalable and can be subjected to high-throughput assays that will rapidly add to our understanding of TRMs.
Collapse
Affiliation(s)
- Max R Ulibarri
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Ying Lin
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA; Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
| | - Julian C Ramprashad
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Geongoo Han
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Mohammad H Hasan
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Farha J Mithila
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA; Molecular Biology, Cell Biology and Biochemistry Graduate Program, Brown University, Providence, RI 02912, USA
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Smita Gopinath
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Cambridge, MA 02115, USA
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, TX 78229, USA; South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - J Justin Milner
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Lalit K Beura
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
9
|
Izadifar Z, Cotton J, Chen S, Horvath V, Stejskalova A, Gulati A, LoGrande NT, Budnik B, Shahriar S, Doherty ER, Xie Y, To T, Gilpin SE, Sesay AM, Goyal G, Lebrilla CB, Ingber DE. Mucus production, host-microbiome interactions, hormone sensitivity, and innate immune responses modeled in human cervix chips. Nat Commun 2024; 15:4578. [PMID: 38811586 PMCID: PMC11137093 DOI: 10.1038/s41467-024-48910-0] [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: 04/22/2023] [Accepted: 03/22/2024] [Indexed: 05/31/2024] Open
Abstract
Modulation of the cervix by steroid hormones and commensal microbiome play a central role in the health of the female reproductive tract. Here we describe organ-on-a-chip (Organ Chip) models that recreate the human cervical epithelial-stromal interface with a functional epithelial barrier and production of mucus with biochemical and hormone-responsive properties similar to living cervix. When Cervix Chips are populated with optimal healthy versus dysbiotic microbial communities (dominated by Lactobacillus crispatus and Gardnerella vaginalis, respectively), significant differences in tissue innate immune responses, barrier function, cell viability, proteome, and mucus composition are observed that are similar to those seen in vivo. Thus, human Cervix Organ Chips represent physiologically relevant in vitro models to study cervix physiology and host-microbiome interactions, and hence may be used as a preclinical testbed for development of therapeutic interventions to enhance women's health.
Collapse
Affiliation(s)
- Zohreh Izadifar
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
- Urology Department, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Justin Cotton
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Siyu Chen
- Department of Chemistry, University of California Davis, Davis, California, Davis, CA, 95616, USA
| | - Viktor Horvath
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Anna Stejskalova
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Aakanksha Gulati
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Nina T LoGrande
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Bogdan Budnik
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Sanjid Shahriar
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Erin R Doherty
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Yixuan Xie
- Department of Chemistry, University of California Davis, Davis, California, Davis, CA, 95616, USA
| | - Tania To
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Sarah E Gilpin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Adama M Sesay
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Carlito B Lebrilla
- Department of Chemistry, University of California Davis, Davis, California, Davis, CA, 95616, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA.
- Vascular Biology Program, Boston Children's Hospital and Department of Pathology, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, 02134, USA.
| |
Collapse
|
10
|
Kowsar R, Sadeghi K, Hashemzadeh F, Miyamoto A. Ovarian sex steroid and epithelial control of immune responses in the uterus and oviduct: human and animal models†. Biol Reprod 2024; 110:230-245. [PMID: 38038990 PMCID: PMC10873282 DOI: 10.1093/biolre/ioad166] [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: 09/29/2023] [Revised: 11/08/2023] [Accepted: 11/30/2023] [Indexed: 12/02/2023] Open
Abstract
The female reproductive tract (FRT), including the uterus and oviduct (Fallopian tube), is responsible for maintaining an optimal microenvironment for reproductive processes, such as gamete activation and transportation, sperm capacitation, fertilization, and early embryonic and fetal development. The mucosal surface of the FRT may be exposed to pathogens and sexually transmitted microorganisms due to the opening of the cervix during mating. Pathogens and endotoxins may also reach the oviduct through the peritoneal fluid. To maintain an optimum reproductive environment while recognizing and killing pathogenic bacterial and viral agents, the oviduct and uterus should be equipped with an efficient and rigorously controlled immune system. Ovarian sex steroids can affect epithelial cells and underlying stromal cells, which have been shown to mediate innate and adaptive immune responses. This, in turn, protects against potential infections while maintaining an optimal milieu for reproductive events, highlighting the homeostatic involvement of ovarian sex steroids and reproductive epithelial cells. This article will discuss how ovarian sex steroids affect the immune reactions elicited by the epithelial cells of the non-pregnant uterus and oviduct in the bovine, murine, and human species. Finally, we propose that there are regional and species-specific differences in the immune responses in FRT.
Collapse
Affiliation(s)
- Rasoul Kowsar
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | | | - Farzad Hashemzadeh
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Akio Miyamoto
- Global Agromedicine Research Center, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| |
Collapse
|
11
|
Kircher BK, Stanley EL, Behringer RR. Anatomy of the female reproductive tract organs of the brown anole (Anolis sagrei). Anat Rec (Hoboken) 2024; 307:395-413. [PMID: 37506227 PMCID: PMC11683880 DOI: 10.1002/ar.25293] [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] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/13/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023]
Abstract
Female reproduction in squamate reptiles (lizards and snakes) is highly diverse and mode of reproduction, clutch size, and reproductive tract morphology all vary widely across this group of ~11,000 species. Recently, CRISPR genome editing techniques that require manipulation of the female reproductive anatomy have been developed in this group, making a more complete understanding of this anatomy essential. We describe the adult female reproductive anatomy of the model reptile the brown anole (Anolis sagrei). We show that the brown anole female reproductive tract has three distinct anterior-to-posterior regions, the infundibulum, the glandular uterus, and the nonglandular uterus. The infundibulum has a highly ciliated epithelial lip, a region where the epithelium is inverted so that cilia are present on the inside and outside of the tube. The glandular uterus has epithelial ducts that are patent with a lumen as well as acinar structures with a lumen. The nonglandular uterus has a heterogeneous morphology from anterior to posterior, with a highly folded, ciliated epithelium transitioning to a stratified squamous epithelium. This transition is accompanied by a loss of keratin-8 expression and together, these changes are similar to the morphological and gene expression changes that occur in the mammalian cervix. We recommend that description of the nonglandular uterus include the regional sub-specification of a "cervix" and "vagina" as this terminology change more accurately describes the morphology. Our data extend histological studies of reproductive organ morphology in reptiles and expand our understanding of the variation in reproductive system anatomy across squamates and vertebrates.
Collapse
Affiliation(s)
- Bonnie K. Kircher
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Edward L. Stanley
- Florida Museum of Natural History, University of Florida, Gainesville, Florida 32601
| | - Richard R. Behringer
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
12
|
Madhukaran S, Hon GC, Mahendroo M. Protocol to dissociate epithelia from non-pregnant and pregnant mouse cervical tissue for single-cell RNA-sequencing. STAR Protoc 2023; 4:102631. [PMID: 37897730 PMCID: PMC10751548 DOI: 10.1016/j.xpro.2023.102631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/24/2023] [Accepted: 09/20/2023] [Indexed: 10/30/2023] Open
Abstract
A challenge in studying cervical epithelial cell biology at the single-cell level is that differentiated subtypes, in particular mucus-secreting goblet cells, are sensitive to disassociating enzymes making isolation of all epithelial subpopulations difficult. Here we present a protocol to dissociate epithelia from non-pregnant and pregnant mouse cervical tissue for single-cell RNA-sequencing. We describe steps for harvesting cervices, preparing cervical tissue, dissociation of cervical cells, and viability checks. We then detail library preparation, sequencing, and procedure for data analysis. For complete details on the use and execution of this protocol, please refer to Cooley et al. (2023).1.
Collapse
Affiliation(s)
- ShanmugaPriyaa Madhukaran
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gary C Hon
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mala Mahendroo
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
13
|
Ulibarri MR, Lin Y, Ramprashad JR, Han G, Hasan MH, Mithila FJ, Ma C, Gopinath S, Zhang N, Milner JJ, Beura LK. Epithelial organoid supports resident memory CD8 T cell differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569395. [PMID: 38076957 PMCID: PMC10705482 DOI: 10.1101/2023.12.01.569395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Resident Memory T cells (TRM) play a vital role in regional immune defense in barrier organs. Although laboratory rodents have been extensively used to study fundamental TRM biology, poor isolation efficiency, sampling bias and low cell survival rates have limited our ability to conduct TRM-focused high-throughput assays. Here, we engineered a murine vaginal epithelial organoid (VEO)-CD8 T cell co-culture system that supports CD8 TRM differentiation in vitro. The three-dimensional VEOs established from murine adult stem cells resembled stratified squamous vaginal epithelium and induced gradual differentiation of activated CD8 T cells into epithelial TRM. These in vitro generated TRM were phenotypically and transcriptionally similar to in vivo TRM, and key tissue residency features were reinforced with a second cognate-antigen exposure during co-culture. TRM differentiation was not affected even when VEOs and CD8 T cells were separated by a semipermeable barrier, indicating soluble factors' involvement. Pharmacological and genetic approaches showed that TGF-β signaling played a crucial role in their differentiation. We found that the VEOs in our model remained susceptible to viral infections and the CD8 T cells were amenable to genetic manipulation; both of which will allow detailed interrogation of antiviral CD8 T cell biology in a reductionist setting. In summary, we established a robust model which captures bonafide TRM differentiation that is scalable, open to iterative sampling, and can be subjected to high throughput assays that will rapidly add to our understanding of TRM.
Collapse
Affiliation(s)
- Max R. Ulibarri
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
| | - Ying Lin
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
- Pathobiology Graduate Program, Brown University, Providence, RI, 02912
| | - Julian R. Ramprashad
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
| | - Geongoo Han
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
| | - Mohammad H. Hasan
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
| | - Farha J. Mithila
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
- Molecular Biology, Cell Biology and Biochemistry Graduate Program, Brown University, Providence, RI, 02912
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, TX, 78229
| | - Smita Gopinath
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Cambridge, MA, 02115
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, TX, 78229
- South Texas Veterans Health Care System, San Antonio, TX, 78229
| | - J. Justin Milner
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, 27599
| | - Lalit K. Beura
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
| |
Collapse
|
14
|
Zhao H, Wang Y, Xu H, Liu M, Xu X, Zhu S, Liu Z, Cai H, Wang Y, Lu J, Yang X, Kong S, Bao H, Wang H, Deng W. Stromal cells-specific retinoic acid determines parturition timing at single-cell and spatial-temporal resolution. iScience 2023; 26:107796. [PMID: 37720083 PMCID: PMC10502414 DOI: 10.1016/j.isci.2023.107796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/23/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023] Open
Abstract
The underlying mechanisms governing parturition remain largely elusive due to limited knowledge of parturition preparation and initiation. Accumulated evidences indicate that maternal decidua plays a critical role in parturition initiation. To comprehensively decrypt the cell heterogeneity in decidua approaching parturition, we investigate the roles of various cell types in mouse decidua process and reveal previously unappreciated insights in parturition initiation utilizing single-cell RNA sequencing (scRNA-seq). We enumerate the cell types in decidua and identity five different stromal cells populations and one decidualized stromal cells. Furthermore, our study unravels that stromal cells prepare for parturition by regulating local retinol acid (RA) synthesis. RA supplement decreases expression of extracellular matrix-related genes in vitro and accelerates the timing of parturition in vivo. Collectively, the discovery of contribution of stromal cells in parturition expands current knowledge about parturition and opens up avenues for the intervention of preterm birth (PTB).
Collapse
Affiliation(s)
- Hui Zhao
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yang Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Hui Xu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Meng Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xinmei Xu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Sijing Zhu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhao Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Han Cai
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yinan Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiaoqing Yang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Nantong University, Xisi Road, Nantong, Jiangsu, China
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Haili Bao
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wenbo Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|