1
|
Sipes J, Rayamajhi S, Bantis LE, Madan R, Mitra A, Puri RV, Rahman MM, Ahmmed F, Pathak HB, Godwin AK. Spatial transcriptomic profiling of the human fallopian tube epithelium reveals region-specific gene expression patterns. Commun Biol 2025; 8:520. [PMID: 40158048 PMCID: PMC11954873 DOI: 10.1038/s42003-025-07871-w] [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: 02/22/2024] [Accepted: 03/03/2025] [Indexed: 04/01/2025] Open
Abstract
The fallopian tube (FT) plays a crucial role in fertility, gynecological health, and high-grade serous ovarian cancer (HGSOC) development. Despite its importance, the spatial transcriptome of the FT's distinct anatomical regions (fimbria, infundibulum, ampulla, and isthmus) remains underexplored. Using the GeoMx Digital Spatial Profiler (DSP) and a targeted ~1800 gene panel, we analyze premenopausal FT epithelium, identifying region-specific gene expression patterns. Our analysis reveals upregulation of mature ciliated cell markers (FOXJ1, MLF1, SPA17, and CTSS) approaching the fimbria, elevated ROS and apoptosis-related transcripts (TXNIP, PRDX5, BAD, GAS1) in the distal FT, and a switch in cell-cell adhesion transcripts (CDH1, CDH3) along the distal-to-proximal axis. We also provide evidence that MHC-II transcripts in the FT are differentially regulated throughout the menstrual cycle, with lower expression in follicular phase. These results suggest spatially regulated expression of FT transcripts with implications for fertilization and early neoplastic changes contributing to HGSOC.
Collapse
Affiliation(s)
- Jared Sipes
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Bioengineering Program, The University of Kansas, Lawrence, KS, 64111, USA
| | - Sagar Rayamajhi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Leonidas E Bantis
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Rashna Madan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Amrita Mitra
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Rajni V Puri
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Mohammod Mahmudur Rahman
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Foyez Ahmmed
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Department of Statistics, Comilla University, Cumilla, 3506, Bangladesh
| | - Harsh B Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Bioengineering Program, The University of Kansas, Lawrence, KS, 64111, USA.
- The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
2
|
Gatimel N, Perez G, Bruno E, Sagnat D, Rolland C, Tanguy-Le-Gac Y, Di Donato E, Racaud C, Léandri R, Bettiol C, Deraison C, Motta JP, Huyghe E, Vergnolle N. Human fallopian tube organoids provide a favourable environment for sperm motility. Hum Reprod 2025; 40:503-517. [PMID: 39792911 DOI: 10.1093/humrep/deae258] [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: 04/29/2024] [Revised: 10/01/2024] [Indexed: 01/12/2025] Open
Abstract
STUDY QUESTION Does a human fallopian tube (HFT) organoid model offer a favourable apical environment for human sperm survival and motility? SUMMARY ANSWER After differentiation, the apical compartment of a new HFT organoid model provides a favourable environment for sperm motility, which is better than commercial media. WHAT IS KNOWN ALREADY HFTs are the site of major events that are crucial for achieving an ongoing pregnancy, such as gamete survival and competence, fertilization steps, and preimplantation embryo development. In order to better understand the tubal physiology and tubal factors involved in these reproductive functions, and to improve still suboptimal in vitro conditions for gamete preparation and embryo culture during IVF, we sought to develop an HFT organoid model from isolated adult stem cells to allow spermatozoa co-culture in the apical compartment. STUDY DESIGN, SIZE, DURATION Over a 2-year period, fallopian tube tissues were collected for organoid culture purposes from 10 'donor' patients undergoing bilateral salpingectomy by laparoscopy for definitive sterilization. After tissue digestion, isolated cells from the isthmus and ampulla regions were separately seeded in 3D Matrigel and cultured with conventional growth factors for organoid culture and specific factors for differentiation of the female genital tract. PARTICIPANTS/MATERIALS, SETTING, METHODS HFT organoids were characterized by light microscopy, scanning and transmission electron microscopy, immunofluorescence, and transcriptome analysis. Following simultaneous organoid culture on specific inserts, spermatozoa from five donors were placed either in control media or in the apical compartment of colon or HFT organoids (isthmus and ampulla separately) for 96 h. Vitality and motility and kinematic parameters were assessed at 0, 48, and 96 h on 200 spermatozoa in each condition and in duplicate and compared using the Wilcoxon test. MAIN RESULTS AND THE ROLE OF CHANCE Specific fallopian tube differentiation of our model was confirmed by immunofluorescence, transcriptome analysis, and electron microscopy observations that exhibited ciliated and secretory cells. We succeeded in releasing spermatozoa in the apical compartment of HFT organoids and in recovering them for sperm analysis. Sperm vitality values were similar in HFT organoids and in commercial sperm media. We demonstrated a superiority of the HFT organoid apical compartment for sperm motility compared with other controls (colon organoids, organoid culture media, and conventional commercial sperm fertilization media). At 48 h of incubation, progressive sperm motility was higher in the apical compartment of HFT organoids (ampulla 31% ± 17, isthmus 29% ± 15) than in commercial fertilization media (15% ± 15) (P < 0.05) and compared with all other conditions. At 96 h, progressive sperm motility was almost nil (<1%) in all conditions except for spermatozoa in HFT organoids (P < 0.05): 12% ± 15 and 13% ± 17 in ampulla and isthmus organoids, respectively. Computer-assisted sperm analysis (CASA) analysis also showed that the organoids were able to maintain significantly higher levels of kinematic parameters (curvilinear velocity, average path velocity, straight linear velocity, and amplitude of lateral movement of the head) and therefore more efficient mobility compared with commercial IVF media. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION This was an in vitro study in which conditions of organoid culture could not exactly mimic the in vivo environment of the extracellular matrix and vascularization of fallopian tubes. WIDER IMPLICATIONS OF THE FINDINGS This work opens up perspectives for better understanding of HFT physiology. For the first time, it highlights the possibility of developing HFT organoids for reproductive purposes. In the future, it could help us to improve gamete fertilizing abilities and embryo culture conditions during human ARTs. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by a grant from the Occitanie region, and by financial allocations from the DEFE and IRSD research teams. The authors have no conflicts of interest to report.
Collapse
Affiliation(s)
- Nicolas Gatimel
- Department of Reproductive Medicine, Hôpital Paule de Viguier, CHU Toulouse, Toulouse, France
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III-Paul Sabatier (UPS), Toulouse, France
- DEFE (Développement Embryonnaire, Fertilité, Environnement-Embryonic Development, Fertility, Environment), UMR1203, INSERM-Universities of Toulouse and Montpellier, Paule de Viguier Hospital, Toulouse, France
- Toulouse Organoid Platform of IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Guillaume Perez
- DEFE (Développement Embryonnaire, Fertilité, Environnement-Embryonic Development, Fertility, Environment), UMR1203, INSERM-Universities of Toulouse and Montpellier, Paule de Viguier Hospital, Toulouse, France
| | - Eloïse Bruno
- DEFE (Développement Embryonnaire, Fertilité, Environnement-Embryonic Development, Fertility, Environment), UMR1203, INSERM-Universities of Toulouse and Montpellier, Paule de Viguier Hospital, Toulouse, France
- Toulouse Organoid Platform of IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - David Sagnat
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III-Paul Sabatier (UPS), Toulouse, France
- Toulouse Organoid Platform of IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Corinne Rolland
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Yan Tanguy-Le-Gac
- Department of Reproductive Medicine, Hôpital Paule de Viguier, CHU Toulouse, Toulouse, France
| | - Emeline Di Donato
- Department of Reproductive Medicine, Hôpital Paule de Viguier, CHU Toulouse, Toulouse, France
| | - Claire Racaud
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Roger Léandri
- Department of Reproductive Medicine, Hôpital Paule de Viguier, CHU Toulouse, Toulouse, France
| | - Célia Bettiol
- Department of Reproductive Medicine, Hôpital Paule de Viguier, CHU Toulouse, Toulouse, France
- DEFE (Développement Embryonnaire, Fertilité, Environnement-Embryonic Development, Fertility, Environment), UMR1203, INSERM-Universities of Toulouse and Montpellier, Paule de Viguier Hospital, Toulouse, France
| | - Céline Deraison
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Jean-Paul Motta
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Eric Huyghe
- Department of Reproductive Medicine, Hôpital Paule de Viguier, CHU Toulouse, Toulouse, France
- DEFE (Développement Embryonnaire, Fertilité, Environnement-Embryonic Development, Fertility, Environment), UMR1203, INSERM-Universities of Toulouse and Montpellier, Paule de Viguier Hospital, Toulouse, France
| | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III-Paul Sabatier (UPS), Toulouse, France
- Toulouse Organoid Platform of IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III-Paul Sabatier (UPS), Toulouse, France
| |
Collapse
|
3
|
Rizo JA, Ahmad V, Pru JM, Winuthayanon S, Challa S, Kim TH, Jeong JW, Spencer TE, Kelleher AM. Uterine organoids reveal insights into epithelial specification and plasticity in development and disease. Proc Natl Acad Sci U S A 2025; 122:e2422694122. [PMID: 39883834 PMCID: PMC11804710 DOI: 10.1073/pnas.2422694122] [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: 11/11/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025] Open
Abstract
Understanding how epithelial cells in the female reproductive tract (FRT) differentiate is crucial for reproductive health, yet the underlying mechanisms remain poorly defined. At birth, FRT epithelium is highly malleable, allowing differentiation into various epithelial types, but the regulatory pathways guiding these early cell fate decisions are unclear. Here, we use neonatal mouse endometrial organoids and assembloid coculture models to investigate how innate cellular plasticity and external mesenchymal signals influence epithelial differentiation. Our findings demonstrate that uterine epithelium undergoes marked age-dependent changes, transitioning from a highly plastic state capable of forming both monolayered and multilayered structures to a more restricted fate as development progresses. Interestingly, parallels emerge between the developmental plasticity of neonatal uterine epithelium and pathological conditions such as endometrial cancer, where similar regulatory mechanisms may reactivate, driving abnormal epithelial differentiation and tumorigenesis. These results not only deepen our understanding of early uterine development but also offer a valuable model for studying the progression of reproductive diseases and cancers.
Collapse
Affiliation(s)
- Jason A. Rizo
- Division of Animal Sciences, University of Missouri, Columbia, MO65211
| | - Vakil Ahmad
- Division of Animal Sciences, University of Missouri, Columbia, MO65211
| | - Jacob M. Pru
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO65211
| | - Sarayut Winuthayanon
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO65211
| | - Sridevi Challa
- The University of Chicago Comprehensive Cancer Center, The University of Chicago, Chicago, IL60637
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL60637
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO65211
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO65211
| | - Thomas E. Spencer
- Division of Animal Sciences, University of Missouri, Columbia, MO65211
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO65211
| | - Andrew M. Kelleher
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO65211
| |
Collapse
|
4
|
Santativongchai P, Klaeui CC, Kosonsiriluk S, Saqui-Salces M, Reed KM, Wileman BW, Studniski MM, Boukherroub KS. Protocol to establish turkey oviductal organoids as an in vitro model. STAR Protoc 2024; 5:103384. [PMID: 39388356 PMCID: PMC11735998 DOI: 10.1016/j.xpro.2024.103384] [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: 07/03/2024] [Revised: 09/03/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
The study of reproductive function in turkey hens has been difficult due to the lack of a reliable, representative in vitro model for investigating profound physiological aspects. This article presents a protocol to establish turkey oviductal organoids, including steps for isolating turkey oviduct epithelial cells followed by seeding and maintaining 3D organoid cultures. We also detail procedures for organoid fixation for histological analysis. This organoid model could serve as a valuable in vitro tool for understanding the intricacies of turkey reproductive physiology.
Collapse
Affiliation(s)
| | - Caitlin C Klaeui
- Department of Animal Science, University of Minnesota, Saint Paul, MN 55108, USA
| | | | - Milena Saqui-Salces
- Department of Animal Science, University of Minnesota, Saint Paul, MN 55108, USA
| | - Kent M Reed
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN 55108, USA
| | | | | | - Kahina S Boukherroub
- Department of Animal Science, University of Minnesota, Saint Paul, MN 55108, USA.
| |
Collapse
|
5
|
Flesken-Nikitin A, Ralston CQ, Fu DJ, De Micheli AJ, Phuong DJ, Harlan BA, Ashe CS, Armstrong AP, McKellar DW, Ghuwalewala S, Ellenson LH, Schimenti JC, Cosgrove BD, Nikitin AY. Pre-ciliated tubal epithelial cells are prone to initiation of high-grade serous ovarian carcinoma. Nat Commun 2024; 15:8641. [PMID: 39366996 PMCID: PMC11452611 DOI: 10.1038/s41467-024-52984-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 09/27/2024] [Indexed: 10/06/2024] Open
Abstract
The distal region of the uterine (Fallopian) tube is commonly associated with high-grade serous carcinoma (HGSC), the predominant and most aggressive form of ovarian or extra-uterine cancer. Specific cell states and lineage dynamics of the adult tubal epithelium (TE) remain insufficiently understood, hindering efforts to determine the cell of origin for HGSC. Here, we report a comprehensive census of cell types and states of the mouse uterine tube. We show that distal TE cells expressing the stem/progenitor cell marker Slc1a3 can differentiate into both secretory (Ovgp1+) and ciliated (Fam183b+) cells. Inactivation of Trp53 and Rb1, whose pathways are commonly altered in HGSC, leads to elimination of targeted Slc1a3+ cells by apoptosis, thereby preventing their malignant transformation. In contrast, pre-ciliated cells (Krt5+, Prom1+, Trp73+) remain cancer-prone and give rise to serous tubal intraepithelial carcinomas and overt HGSC. These findings identify transitional pre-ciliated cells as a cancer-prone cell state and point to pre-ciliation mechanisms as diagnostic and therapeutic targets.
Collapse
Affiliation(s)
| | - Coulter Q Ralston
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Dah-Jiun Fu
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
| | - Andrea J De Micheli
- Department of Oncology and Children's Research Center, University Children's Hospital Zürich, Zürich, Switzerland
| | - Daryl J Phuong
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Blaine A Harlan
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
| | | | | | - David W McKellar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Sangeeta Ghuwalewala
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | | | - John C Schimenti
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Benjamin D Cosgrove
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | |
Collapse
|
6
|
Wesselman HM, Arceri L, Nguyen TK, Lara CM, Wingert RA. Genetic mechanisms of multiciliated cell development: from fate choice to differentiation in zebrafish and other models. FEBS J 2024; 291:4159-4192. [PMID: 37997009 DOI: 10.1111/febs.17012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/17/2023] [Accepted: 11/21/2023] [Indexed: 11/25/2023]
Abstract
Multiciliated cells (MCCS) form bundles of cilia and their activities are essential for the proper development and physiology of many organ systems. Not surprisingly, defects in MCCs have profound consequences and are associated with numerous disease states. Here, we discuss the current understanding of MCC formation, with a special focus on the genetic and molecular mechanisms of MCC fate choice and differentiation. Furthermore, we cast a spotlight on the use of zebrafish to study MCC ontogeny and several recent advances made in understanding MCCs using this vertebrate model to delineate mechanisms of MCC emergence in the developing kidney.
Collapse
Affiliation(s)
| | - Liana Arceri
- Department of Biological Sciences, University of Notre Dame, IN, USA
| | - Thanh Khoa Nguyen
- Department of Biological Sciences, University of Notre Dame, IN, USA
| | - Caroline M Lara
- Department of Biological Sciences, University of Notre Dame, IN, USA
| | - Rebecca A Wingert
- Department of Biological Sciences, University of Notre Dame, IN, USA
| |
Collapse
|
7
|
Crawford AJ, Forjaz A, Bons J, Bhorkar I, Roy T, Schell D, Queiroga V, Ren K, Kramer D, Huang W, Russo GC, Lee MH, Wu PH, Shih IM, Wang TL, Atkinson MA, Schilling B, Kiemen AL, Wirtz D. Combined assembloid modeling and 3D whole-organ mapping captures the microanatomy and function of the human fallopian tube. SCIENCE ADVANCES 2024; 10:eadp6285. [PMID: 39331707 PMCID: PMC11430475 DOI: 10.1126/sciadv.adp6285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/22/2024] [Indexed: 09/29/2024]
Abstract
The fallopian tubes play key roles in processes from pregnancy to ovarian cancer where three-dimensional (3D) cellular and extracellular interactions are important to their pathophysiology. Here, we develop a 3D multicompartment assembloid model of the fallopian tube that molecularly, functionally, and architecturally resembles the organ. Global label-free proteomics, innovative assays capturing physiological functions of the fallopian tube (i.e., oocyte transport), and whole-organ single-cell resolution mapping are used to validate these assembloids through a multifaceted platform with direct comparisons to fallopian tube tissue. These techniques converge at a unique combination of assembloid parameters with the highest similarity to the reference fallopian tube. This work establishes (i) an optimized model of the human fallopian tubes for in vitro studies of their pathophysiology and (ii) an iterative platform for customized 3D in vitro models of human organs that are molecularly, functionally, and microanatomically accurate by combining tunable assembloid and tissue mapping methods.
Collapse
Affiliation(s)
- Ashleigh J Crawford
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - André Forjaz
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Isha Bhorkar
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Triya Roy
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - David Schell
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Vasco Queiroga
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kehan Ren
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Donald Kramer
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biotechnology, Johns Hopkins Advanced Academic Programs, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Wilson Huang
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Gabriella C Russo
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Meng-Horng Lee
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Pei-Hsun Wu
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ie-Ming Shih
- Department of Gynecology and Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tian-Li Wang
- Department of Gynecology and Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mark A Atkinson
- Departments of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
- Departments of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | | | - Ashley L Kiemen
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Functional Anatomy and Evolution, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Denis Wirtz
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
8
|
Franko R, de Almeida Monteiro Melo Ferraz M. Exploring the potential of in vitro extracellular vesicle generation in reproductive biology. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70007. [PMID: 39238549 PMCID: PMC11375532 DOI: 10.1002/jex2.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/07/2024]
Abstract
The interest in the growing field of extracellular vesicle (EV) research highlights their significance in intercellular signalling and the selective transfer of biological information between donor and recipient cells. EV studies have provided valuable insights into intercellular communication mechanisms, signal identification and their involvement in disease states, offering potential avenues for manipulating pathological conditions, detecting biomarkers and developing drug-delivery systems. While our understanding of EV functions in reproductive tissues has significantly progressed, exploring their potential as biomarkers for infertility, therapeutic interventions and enhancements in assisted reproductive technologies remains to be investigated. This knowledge gap stems partly from the difficulties associated with large-scale EV production relevant to clinical applications. Most existing studies on EV production rely on conventional 2D cell culture systems, characterized by suboptimal EV yields and a failure to replicate in vivo conditions. This results in the generation of EVs that differ from their in vivo counterparts. Hence, this review firstly delves into the importance of EVs in reproduction to then expand on current techniques for in vitro EV production, specifically examining diverse methods of culture and the potential of bioengineering technologies to establish innovative systems for enhanced EV production.
Collapse
Affiliation(s)
- Roksan Franko
- Clinic of Ruminants, Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Oberschleißheim Germany
- Gene Center Ludwig-Maximilians-Universität München Munich Germany
| | - Marcia de Almeida Monteiro Melo Ferraz
- Clinic of Ruminants, Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Oberschleißheim Germany
- Gene Center Ludwig-Maximilians-Universität München Munich Germany
| |
Collapse
|
9
|
Desantis S, Cinone M, Lacitignola L, Laricchiuta P, Rossi R, Guaricci AC, Resta L, Albrizio M. Morphology, Glycan Pattern, Heat Shock Proteins, and Sex Steroid Receptors Expression in the Tubal Fimbria Epithelium of the Baboon Papio hamadryas during the Menstrual Cycle. Animals (Basel) 2024; 14:2321. [PMID: 39199855 PMCID: PMC11350877 DOI: 10.3390/ani14162321] [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: 07/06/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024] Open
Abstract
The oviductal fimbria is the first extraovarian anatomical structure that the cumulus-oocyte complex (COC) encounters, and is sensitive to sex hormone changes. The morphology, glycan pattern, expression of heat shock proteins (HSPs), estradiol receptor (ER), and progesterone receptor (PR) were investigated in the oviductal fimbria epithelium of the baboon (Papio hamadryas) during the menstrual cycle. The morphology was investigated by light and scanning electron microscopy; the glycopattern was characterized using conventional and lectin histochemistry; HSPs (60, -70, -90), ER, and PR were localized immunohistochemically. Well-differentiated ciliated and nonciliated cells were present only during the preovulatory phase. The nonciliated cells contained small apical protrusions and thin microvilli. During the preovulatory phase (1) the luminal surface of the fimbria displayed acidic glycans, complex N-glycans containing fucose, and oligolactosamine residues; (2) nonciliated cells expressed HSP60 and HSP90 in the apical blebs, HSP70 in the nucleus and cytoplasm, as well as nuclear ERα and PR; (3) ciliated cells showed HSP70 in the nucleus, cytoplasm, and cilia that also expressed HSP90 and PR. These results are related to the function of the fimbria where the early COC-oviduct crosstalk occurs and may represent a benchmark for translational studies of other primates.
Collapse
Affiliation(s)
- Salvatore Desantis
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy; (M.C.); (L.L.); (R.R.); (A.C.G.); (L.R.); (M.A.)
| | - Mario Cinone
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy; (M.C.); (L.L.); (R.R.); (A.C.G.); (L.R.); (M.A.)
| | - Luca Lacitignola
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy; (M.C.); (L.L.); (R.R.); (A.C.G.); (L.R.); (M.A.)
| | | | - Roberta Rossi
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy; (M.C.); (L.L.); (R.R.); (A.C.G.); (L.R.); (M.A.)
| | - Antonio Ciro Guaricci
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy; (M.C.); (L.L.); (R.R.); (A.C.G.); (L.R.); (M.A.)
| | - Leonardo Resta
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy; (M.C.); (L.L.); (R.R.); (A.C.G.); (L.R.); (M.A.)
| | - Maria Albrizio
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy; (M.C.); (L.L.); (R.R.); (A.C.G.); (L.R.); (M.A.)
| |
Collapse
|
10
|
Chehade H, Gogoi R, Adzibolosu NK, Galoforo S, Fehmi RA, Kheil M, Fox A, Kim S, Rattan R, Hou Z, Morris RT, Matherly LH, Mor G, Alvero AB. BRCA Status Dictates Wnt Responsiveness in Epithelial Ovarian Cancer. CANCER RESEARCH COMMUNICATIONS 2024; 4:2075-2088. [PMID: 39028933 PMCID: PMC11320024 DOI: 10.1158/2767-9764.crc-24-0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/17/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
The association of BRCA1 and BRCA2 mutations with increased risk for developing epithelial ovarian cancer is well established. However, the observed clinical differences, particularly the improved therapy response and patient survival in BRCA2-mutant patients, are unexplained. Our objective is to identify molecular pathways that are differentially regulated upon the loss of BRCA1 and BRCA2 functions in ovarian cancer. Transcriptomic and pathway analyses comparing BRCA1-mutant, BRCA2-mutant, and homologous recombination wild-type ovarian tumors showed differential regulation of the Wnt/β-catenin pathway. Using Wnt3A-treated BRCA1/2 wild-type, BRCA1-null, and BRCA2-null mouse ovarian cancer cells, we observed preferential activation of canonical Wnt/β-catenin signaling in BRCA1/2 wild-type ovarian cancer cells, whereas noncanonical Wnt/β-catenin signaling was preferentially activated in the BRCA1-null ovarian cancer cells. Interestingly, BRCA2-null mouse ovarian cancer cells demonstrated a unique response to Wnt3A with the preferential upregulation of the Wnt signaling inhibitor Axin2. In addition, decreased phosphorylation and enhanced stability of β-catenin were observed in BRCA2-null mouse ovarian cancer cells, which correlated with increased inhibitory phosphorylation on GSK3β. These findings open venues for the translation of these molecular observations into modalities that can impact patient survival. SIGNIFICANCE We show that BRCA1 and BRCA2 mutation statuses differentially impact the regulation of the Wnt/β-catenin signaling pathway, a major effector of cancer initiation and progression. Our findings provide a better understanding of molecular mechanisms that promote the known differential clinical profile in these patient populations.
Collapse
Affiliation(s)
- Hussein Chehade
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan.
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
| | - Radhika Gogoi
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
- Karmanos Cancer Institute, Detroit, Michigan.
| | - Nicholas K. Adzibolosu
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
| | - Sandra Galoforo
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
| | - Rouba-Ali Fehmi
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan.
- Karmanos Cancer Institute, Detroit, Michigan.
| | - Mira Kheil
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan.
- Karmanos Cancer Institute, Detroit, Michigan.
| | - Alexandra Fox
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
| | - Seongho Kim
- Karmanos Cancer Institute, Detroit, Michigan.
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
| | - Ramandeep Rattan
- Division of Gynecology Oncology, Department of Women’s Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, Michigan.
| | - Zhanjun Hou
- Karmanos Cancer Institute, Detroit, Michigan.
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
| | - Robert T. Morris
- Karmanos Cancer Institute, Detroit, Michigan.
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
| | - Larry H. Matherly
- Karmanos Cancer Institute, Detroit, Michigan.
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
| | - Gil Mor
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
- Karmanos Cancer Institute, Detroit, Michigan.
| | - Ayesha B. Alvero
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
- Karmanos Cancer Institute, Detroit, Michigan.
| |
Collapse
|
11
|
Seghers S, Le Compte M, Hendriks JMH, Van Schil P, Janssens A, Wener R, Komen N, Prenen H, Deben C. A systematic review of patient-derived tumor organoids generation from malignant effusions. Crit Rev Oncol Hematol 2024; 195:104285. [PMID: 38311013 DOI: 10.1016/j.critrevonc.2024.104285] [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: 12/07/2023] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/06/2024] Open
Abstract
This review assesses the possibility of utilizing malignant effusions (MEs) for generating patient-derived tumor organoids (PDTOs). Obtained through minimally invasive procedures MEs broaden the spectrum of organoid sources beyond resection specimens and tissue biopsies. A systematic search yielded 11 articles, detailing the successful generation of 190 ME-PDTOs (122 pleural effusions, 54 malignant ascites). Success rates ranged from 33% to 100%, with an average of 84% and median of 92%. A broad and easily applicable array of techniques can be employed, encompassing diverse collection methods, variable centrifugation speeds, and the inclusion of approaches like RBC lysis buffer or centrifuged ME supernatants supplementation, enhancing the versatility and accessibility of the methodology. ME-PDTOs were found to recapitulate primary tumor characteristics and were primarily used for drug screening applications. Thus, MEs are a reliable source for developing PDTOs, emphasizing the need for further research to maximize their potential, validate usage, and refine culturing processes.
Collapse
Affiliation(s)
- Sofie Seghers
- Department of Oncology, Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium.
| | - Maxim Le Compte
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Jeroen M H Hendriks
- Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium; Department of Thoracic and Vascular Surgery, Antwerp University Hospital, Edegem, Belgium; Antwerp ReSURG Group, Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), University of Antwerp, Wilrijk, Belgium
| | - Paul Van Schil
- Department of Thoracic and Vascular Surgery, Antwerp University Hospital, Edegem, Belgium; Antwerp ReSURG Group, Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), University of Antwerp, Wilrijk, Belgium
| | - Annelies Janssens
- Department of Thoracic Oncology Antwerp University Hospital, Edegem, Belgium
| | - Reinier Wener
- Department of Thoracic Oncology Antwerp University Hospital, Edegem, Belgium; Department of Pulmonary Diseases, Antwerp University Hospital, Edegem, Belgium
| | - Niels Komen
- Department of Abdominal Surgery, Antwerp University Hospital, Edegem, Belgium; Antwerp ReSURG Group, Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), University of Antwerp, Wilrijk, Belgium
| | - Hans Prenen
- Department of Oncology, Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium; Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Christophe Deben
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium; Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
12
|
Yu B, McCartney S, Strenk S, Valint DJ, Liu C, Haggerty CL, Fredricks DN. Vaginal Bacteria Elicit Acute Inflammatory Response in Fallopian Tube Organoids. Reprod Sci 2024; 31:505-513. [PMID: 37726587 PMCID: PMC11378751 DOI: 10.1007/s43032-023-01350-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/05/2023] [Indexed: 09/21/2023]
Abstract
To facilitate in vitro mechanistic studies in pelvic inflammatory disease and subsequent tubal factor infertility, we sought to establish patient tissue derived fallopian tube (FT) organoids and to study their inflammatory response to acute vaginal bacterial infection. FT tissues were obtained from four patients after salpingectomy for benign gynecological diseases. We introduced acute infection in the FT organoid culture system by inoculating the organoid culture media with two common vaginal bacterial species, Lactobacillus crispatus and Fannyhessea vaginae. The inflammatory response elicited in the organoids after acute bacterial infection was analyzed by the expression profile of 249 inflammatory genes. Compared to the negative controls that were not cultured with any bacteria, the organoids cultured with either bacterial species showed multiple differentially expressed inflammatory genes. Marked differences were noted between the Lactobacillus crispatus infected organoids and those infected by Fannyhessea vaginae. Genes from the C-X-C motif chemokine ligand (CXCL) family were highly upregulated in Fannyhessea vaginae infected organoids. Flow cytometry showed that immune cells quickly disappeared during the organoid culture, indicating the inflammatory response observed with bacterial culture was generated by the epithelial cells in the organoids. In summary, we have shown that patient tissue derived FT organoids respond to acute bacterial infection with upregulation of inflammatory genes specific to different vaginal bacterial species. FT organoids is a useful in vitro model system to study the host-pathogen interaction during bacterial infection.
Collapse
Affiliation(s)
- Bo Yu
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, 240 Pasteur Drive, Stanford, CA, 94305, USA.
- Stanford Maternal & Child Health Research Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
| | - Stephen McCartney
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Susan Strenk
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. North, E4-100, Seattle, WA, 98109, USA
| | - Daniel J Valint
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. North, E4-100, Seattle, WA, 98109, USA
| | - Congzhou Liu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. North, E4-100, Seattle, WA, 98109, USA
| | - Catherine L Haggerty
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - David N Fredricks
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. North, E4-100, Seattle, WA, 98109, USA.
- Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
13
|
Gabriel V, Zdyrski C, Sahoo DK, Ralston A, Wickham H, Bourgois-Mochel A, Ahmed B, Merodio MM, Paukner K, Piñeyro P, Kopper J, Rowe EW, Smith JD, Meyerholz D, Kol A, Viall A, Elbadawy M, Mochel JP, Allenspach K. Adult Animal Stem Cell-Derived Organoids in Biomedical Research and the One Health Paradigm. Int J Mol Sci 2024; 25:701. [PMID: 38255775 PMCID: PMC10815683 DOI: 10.3390/ijms25020701] [Citation(s) in RCA: 1] [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: 12/05/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Preclinical biomedical research is limited by the predictiveness of in vivo and in vitro models. While in vivo models offer the most complex system for experimentation, they are also limited by ethical, financial, and experimental constraints. In vitro models are simplified models that do not offer the same complexity as living animals but do offer financial affordability and more experimental freedom; therefore, they are commonly used. Traditional 2D cell lines cannot fully simulate the complexity of the epithelium of healthy organs and limit scientific progress. The One Health Initiative was established to consolidate human, animal, and environmental health while also tackling complex and multifactorial medical problems. Reverse translational research allows for the sharing of knowledge between clinical research in veterinary and human medicine. Recently, organoid technology has been developed to mimic the original organ's epithelial microstructure and function more reliably. While human and murine organoids are available, numerous other organoids have been derived from traditional veterinary animals and exotic species in the last decade. With these additional organoid models, species previously excluded from in vitro research are becoming accessible, therefore unlocking potential translational and reverse translational applications of animals with unique adaptations that overcome common problems in veterinary and human medicine.
Collapse
Affiliation(s)
- Vojtech Gabriel
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (H.W.); (B.A.); (J.P.M.)
| | | | - Dipak K. Sahoo
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA; (D.K.S.); (A.B.-M.); (J.K.)
| | - Abigail Ralston
- 3D Health Solutions Inc., Ames, IA 50010, USA; (C.Z.); (A.R.); (M.M.M.)
| | - Hannah Wickham
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (H.W.); (B.A.); (J.P.M.)
| | - Agnes Bourgois-Mochel
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA; (D.K.S.); (A.B.-M.); (J.K.)
| | - Basant Ahmed
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (H.W.); (B.A.); (J.P.M.)
| | - Maria M. Merodio
- 3D Health Solutions Inc., Ames, IA 50010, USA; (C.Z.); (A.R.); (M.M.M.)
| | - Karel Paukner
- Atherosclerosis Research Laboratory, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic;
| | - Pablo Piñeyro
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (P.P.); (J.D.S.)
| | - Jamie Kopper
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA; (D.K.S.); (A.B.-M.); (J.K.)
| | - Eric W. Rowe
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (H.W.); (B.A.); (J.P.M.)
| | - Jodi D. Smith
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (P.P.); (J.D.S.)
| | - David Meyerholz
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA;
| | - Amir Kol
- Department of Pathology, University of California, Davis, CA 94143, USA; (A.K.); (A.V.)
| | - Austin Viall
- Department of Pathology, University of California, Davis, CA 94143, USA; (A.K.); (A.V.)
| | - Mohamed Elbadawy
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30530, USA;
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt
| | - Jonathan P. Mochel
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (H.W.); (B.A.); (J.P.M.)
- 3D Health Solutions Inc., Ames, IA 50010, USA; (C.Z.); (A.R.); (M.M.M.)
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30530, USA;
| | - Karin Allenspach
- 3D Health Solutions Inc., Ames, IA 50010, USA; (C.Z.); (A.R.); (M.M.M.)
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA; (D.K.S.); (A.B.-M.); (J.K.)
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30530, USA;
| |
Collapse
|
14
|
Alsaadi A, Artibani M, Hu Z, Wietek N, Morotti M, Gonzalez LS, Alazzam M, Jiang J, Abdul B, Soleymani Majd H, Blazer LL, Adams J, Silvestri F, Sidhu SS, Brugge JS, Ahmed AA. Single-cell transcriptomics identifies a WNT7A-FZD5 signaling axis that maintains fallopian tube stem cells in patient-derived organoids. Cell Rep 2023; 42:113354. [PMID: 37917586 DOI: 10.1016/j.celrep.2023.113354] [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: 04/04/2023] [Revised: 07/31/2023] [Accepted: 10/11/2023] [Indexed: 11/04/2023] Open
Abstract
The study of fallopian tube (FT) function in health and disease has been hampered by limited knowledge of FT stem cells and lack of in vitro models of stem cell renewal and differentiation. Using optimized organoid culture conditions to address these limitations, we find that FT stem cell renewal is highly dependent on WNT/β-catenin signaling and engineer endogenous WNT/β-catenin signaling reporter organoids to biomark, isolate, and characterize these cells. Using functional approaches, as well as bulk and single-cell transcriptomics analyses, we show that an endogenous hormonally regulated WNT7A-FZD5 signaling axis is critical for stem cell renewal and that WNT/β-catenin pathway-activated cells form a distinct transcriptomic cluster of FT cells enriched in extracellular matrix (ECM) remodeling and integrin signaling pathways. Overall, we provide a deep characterization of FT stem cells and their molecular requirements for self-renewal, paving the way for mechanistic work investigating the role of stem cells in FT health and disease.
Collapse
Affiliation(s)
- Abdulkhaliq Alsaadi
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
| | - Mara Artibani
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Zhiyuan Hu
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Nina Wietek
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK
| | - Matteo Morotti
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK
| | - Laura Santana Gonzalez
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
| | - Moiad Alazzam
- Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK
| | - Jason Jiang
- Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK
| | - Beena Abdul
- Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK
| | - Hooman Soleymani Majd
- Medical Sciences Division, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Levi L Blazer
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Jarret Adams
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | | | - Sachdev S Sidhu
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Ahmed Ashour Ahmed
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK.
| |
Collapse
|
15
|
Gómez-Álvarez M, Agustina-Hernández M, Francés-Herrero E, Rodríguez-Eguren A, Bueno-Fernandez C, Cervelló I. Addressing Key Questions in Organoid Models: Who, Where, How, and Why? Int J Mol Sci 2023; 24:16014. [PMID: 37958996 PMCID: PMC10650475 DOI: 10.3390/ijms242116014] [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: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Organoids are three-dimensional cellular structures designed to recreate the biological characteristics of the body's native tissues and organs in vitro. There has been a recent surge in studies utilizing organoids due to their distinct advantages over traditional two-dimensional in vitro approaches. However, there is no consensus on how to define organoids. This literature review aims to clarify the concept of organoids and address the four fundamental questions pertaining to organoid models: (i) What constitutes organoids?-The cellular material. (ii) Where do organoids grow?-The extracellular scaffold. (iii) How are organoids maintained in vitro?-Via the culture media. (iv) Why are organoids suitable in vitro models?-They represent reproducible, stable, and scalable models for biological applications. Finally, this review provides an update on the organoid models employed within the female reproductive tract, underscoring their relevance in both basic biology and clinical applications.
Collapse
Affiliation(s)
- María Gómez-Álvarez
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
| | - Marcos Agustina-Hernández
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
| | - Emilio Francés-Herrero
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
- Department of Pediatrics, Obstetrics and Gynecology, Universitat de València, 46010 Valencia, Spain
| | - Adolfo Rodríguez-Eguren
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
| | - Clara Bueno-Fernandez
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
- Department of Pediatrics, Obstetrics and Gynecology, Universitat de València, 46010 Valencia, Spain
| | - Irene Cervelló
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
| |
Collapse
|
16
|
Menjivar NG, Gad A, Thompson RE, Meyers MA, Hollinshead FK, Tesfaye D. Bovine oviductal organoids: a multi-omics approach to capture the cellular and extracellular molecular response of the oviduct to heat stress. BMC Genomics 2023; 24:646. [PMID: 37891479 PMCID: PMC10605953 DOI: 10.1186/s12864-023-09746-y] [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: 01/13/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND The mammalian oviduct is a complex, fibromuscular organ known for its role in orchestrating a series of timely and dynamic changes to suitably support early embryogenesis. Climate change-induced heat stress (HS) is one of the largest single stressors compromising reproductive function in humans and farm animals via systemic changes in the redox status of the maternal environment, adversely affecting fertilization and early embryonic development. Oviductal organoids represent a unique 3-dimensional, biomimetic model to study the physiology of the oviduct and its subsequent impact on embryo development under various environmental conditions. RESULTS Our study is the first to demonstrate an innovative approach to understanding the cascade of molecular changes sustained by bovine oviductal organoids under HS and the subsequent maternal signals harnessed within their secreted extracellular vesicles (EVs). Transcriptomic analysis of oviductal organoids exposed to HS revealed 2,570 differentially expressed genes (1,222 up- and 1,348 downregulated), while EV-coupled miRNome analysis disclosed 18 miRNAs with significant differential expression (12 up- and 6 downregulated) in EVs from thermally stressed organoids compared to EVs released from organoids cultured under thermoneutral conditions. Genes activated in oviductal organoids in response to thermal stress, include: COX1, ACTB, CST6, TPT1, and HSPB1, while miR-1246, miR-148a, miR21-5p, miR-451, and miR-92a represent the top highly abundant EV-coupled miRNAs released in response to HS. Pathway analysis of genes enriched in organoids exposed to thermal stress showed the enrichment of endocrine resistance, cellular senescence, and notch signaling pathways. Similarly, EV-coupled miRNAs released from thermally stressed organoids showed their potential regulation of genes involved in cellular senescence, p53 signaling, and TGF-beta signaling pathways. CONCLUSIONS In conclusion, the cellular and extracellular response of bovine oviductal organoids to in vitro HS conditions reveal the prospective impact of environmental HS on the physiology of the oviduct and the probable subsequent impacts on oocyte fertilization and early embryo development. Future studies elucidating the potential impact of HS-associated EVs from oviductal organoids on oocyte fertilization and preimplantation embryo development, would justify the use of an organoid model to optimally understand the oviduct-embryo communication under suboptimal environments.
Collapse
Affiliation(s)
- Nico G Menjivar
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 3107 Rampart Rd, Fort Collins, CO, 80521, USA
| | - Ahmed Gad
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 3107 Rampart Rd, Fort Collins, CO, 80521, USA
- Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, 12613, Egypt
| | - Riley E Thompson
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Mindy A Meyers
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Fiona K Hollinshead
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Dawit Tesfaye
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 3107 Rampart Rd, Fort Collins, CO, 80521, USA.
| |
Collapse
|
17
|
Rizo JA, Davenport KM, Winuthayanon W, Spencer TE, Kelleher AM. Estrogen receptor alpha regulates uterine epithelial lineage specification and homeostasis. iScience 2023; 26:107568. [PMID: 37622003 PMCID: PMC10445454 DOI: 10.1016/j.isci.2023.107568] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/11/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
Postnatal development of the uterus involves specification of undifferentiated epithelium into uterine-type epithelium. That specification is regulated by stromal-epithelial interactions as well as intrinsic cell-specific transcription factors and gene regulatory networks. This study utilized mouse genetic models of Esr1 deletion, endometrial epithelial organoids (EEO), and organoid-stromal co-cultures to decipher the role of Esr1 in uterine epithelial development. Organoids derived from wild-type (WT) mice developed a normal single layer of columnar epithelium. In contrast, EEO from Esr1 null mice developed a multilayered stratified squamous type of epithelium with basal cells. Co-culturing Esr1 null epithelium with WT uterine stromal fibroblasts inhibited basal cell development. Of note, estrogen treatment of EEO-stromal co-cultures and Esr1 conditional knockout mice increased basal epithelial cell markers. Collectively, these findings suggest that Esr1 regulates uterine epithelium lineage plasticity and homeostasis and loss of ESR1 promotes altered luminal-to-basal differentiation driven by ESR1-mediated paracrine factors from the stroma.
Collapse
Affiliation(s)
- Jason A. Rizo
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | | | - Wipawee Winuthayanon
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO 65211, USA
| | - Thomas E. Spencer
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO 65211, USA
| | - Andrew M. Kelleher
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
18
|
Ford MJ, Harwalkar K, Kazemdarvish H, Yamanaka N, Yamanaka Y. CD133/Prom1 marks proximal mouse oviduct epithelial progenitors and adult epithelial cells with a low generative capacity. Biol Open 2023; 12:bio059963. [PMID: 37605939 PMCID: PMC10508696 DOI: 10.1242/bio.059963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/15/2023] [Indexed: 08/23/2023] Open
Abstract
The epithelium lining the oviduct or fallopian tube consists of multiciliated and secretory cells, which support fertilization and preimplantation development, however, its homeostasis remains poorly understood. CD133/Prom1 expression has been used as a marker to identify adult stem cell populations in various organs and often associated with cancer cells that have stem-like properties. Using an antibody targeted to CD133 and a Cre recombinase-based lineage tracing strategy, we found that CD133/Prom1 expression is not associated with a stem/progenitor population in the oviduct but marked predominantly multiciliated cells with a low generative capacity. Additionally, we have shown that CD133 is disparately localised along the oviduct during neonatal development, and that Prom1 expressing secretory cells in the ampulla rapidly transitioned to multiciliated cells and progressively migrated to the ridge of epithelial folds.
Collapse
Affiliation(s)
- Matthew J Ford
- Goodman Cancer Institute, Department of Human Genetics, McGill University, Montreal QC H3A 1A3, Canada
| | - Keerthana Harwalkar
- Goodman Cancer Institute, Department of Human Genetics, McGill University, Montreal QC H3A 1A3, Canada
| | - Hengameh Kazemdarvish
- Goodman Cancer Institute, Department of Human Genetics, McGill University, Montreal QC H3A 1A3, Canada
| | - Nobuko Yamanaka
- Goodman Cancer Institute, Department of Human Genetics, McGill University, Montreal QC H3A 1A3, Canada
| | - Yojiro Yamanaka
- Goodman Cancer Institute, Department of Human Genetics, McGill University, Montreal QC H3A 1A3, Canada
| |
Collapse
|
19
|
Crawford AJ, Forjaz A, Bhorkar I, Roy T, Schell D, Queiroga V, Ren K, Kramer D, Bons J, Huang W, Russo GC, Lee MH, Schilling B, Wu PH, Shih IM, Wang TL, Kiemen A, Wirtz D. Precision-engineered biomimetics: the human fallopian tube. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543923. [PMID: 37333379 PMCID: PMC10274705 DOI: 10.1101/2023.06.06.543923] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The fallopian tube has an essential role in several physiological and pathological processes from pregnancy to ovarian cancer. However, there are no biologically relevant models to study its pathophysiology. The state-of-the-art organoid model has been compared to two-dimensional tissue sections and molecularly assessed providing only cursory analyses of the model's accuracy. We developed a novel multi-compartment organoid model of the human fallopian tube that was meticulously tuned to reflect the compartmentalization and heterogeneity of the tissue's composition. We validated this organoid's molecular expression patterns, cilia-driven transport function, and structural accuracy through a highly iterative platform wherein organoids are compared to a three-dimensional, single-cell resolution reference map of a healthy, transplantation-quality human fallopian tube. This organoid model was precision-engineered to match the human microanatomy. One sentence summary Tunable organoid modeling and CODA architectural quantification in tandem help design a tissue-validated organoid model.
Collapse
|
20
|
Yu B, McCartney S, Strenk S, Valint D, Liu C, Haggerty C, Fredricks DN. Vaginal bacteria elicit acute inflammatory response in fallopian tube organoids: a model for pelvic inflammatory disease. RESEARCH SQUARE 2023:rs.3.rs-2891189. [PMID: 37293093 PMCID: PMC10246240 DOI: 10.21203/rs.3.rs-2891189/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Objective: To facilitate in vitro mechanistic studies in pelvic inflammatory disease (PID) and subsequent tubal factor infertility, as well as ovarian carcinogenesis, we sought to establish patient tissue derived fallopian tube (FT) organoids and to study their inflammatory response to acute vaginal bacterial infection. Design: Experimental study. Setting: Academic medical and researchcenter. Patients: FT tissues were obtained from four patients after salpingectomy for benign gynecological diseases. Interventions: We introduced acute infection in the FT organoid culture system by inoculating the organoid culture media with two common vaginal bacterial species, Lactobacillus crispatus and Fannyhesseavaginae . Main Outcome Measures: The inflammatory response elicited in the organoids after acute bacterial infection was analyzed by the expression profile of 249 inflammatory genes. Results: Compared to the negative controls that were not cultured with any bacteria, the organoids cultured with either bacterial species showed multiple differentially expressed inflammatory genes. Marked differences were noted between the Lactobacillus crispatus infected organoids and those infected by Fannyhessea vaginae . Genes from the C-X-C motif chemokine ligand (CXCL) family were highly upregulated in F. vaginae infected organoids. Flow cytometry showed that immune cells quickly disappeared during the organoid culture, indicating the inflammatory response observed with bacterial culture was generated by the epithelial cells in the organoids. Conclusion : Patient tissue derived FT organoids respond to acute bacterial infection with upregulation of inflammatory genes specific to different vaginal bacterial species. FT organoids is a useful model system to study the host-pathogen interaction during bacterial infection which may facilitate mechanistic investigations in PID and its contribution to tubal factor infertility and ovarian carcinogenesis.
Collapse
Affiliation(s)
- Bo Yu
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA
- Stanford Maternal & Child Health Research Institute, Stanford University School of Medicine, Stanford, CA
| | - Stephen McCartney
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA
| | - Susan Strenk
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Daniel Valint
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Congzhou Liu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - David N. Fredricks
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
21
|
Huo C, Zhang X, Gu Y, Wang D, Zhang S, Liu T, Li Y, He W. Organoids: Construction and Application in Gastric Cancer. Biomolecules 2023; 13:biom13050875. [PMID: 37238742 DOI: 10.3390/biom13050875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Gastric organoids are biological models constructed in vitro using stem cell culture and 3D cell culture techniques, which are the latest research hotspots. The proliferation of stem cells in vitro is the key to gastric organoid models, making the cell subsets within the models more similar to in vivo tissues. Meanwhile, the 3D culture technology also provides a more suitable microenvironment for the cells. Therefore, the gastric organoid models can largely restore the growth condition of cells in terms of morphology and function in vivo. As the most classic organoid models, patient-derived organoids use the patient's own tissues for in vitro culture. This kind of model is responsive to the 'disease information' of a specific patient and has great effect on evaluating the strategies of individualized treatment. Herein, we review the current literature on the establishment of organoid cultures, and also explore organoid translational applications.
Collapse
Affiliation(s)
- Chengdong Huo
- Department of the Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou 730030, China
- Department of Ophthalmology, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Xiaoxia Zhang
- Department of the Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou 730030, China
- Department of Ophthalmology, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Yanmei Gu
- Department of the Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Daijun Wang
- Department of the Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Shining Zhang
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou 730030, China
| | - Tao Liu
- Department of the Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou 730030, China
| | - Yumin Li
- Department of the Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou 730030, China
| | - Wenting He
- Department of the Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou 730030, China
| |
Collapse
|
22
|
Zeng J, Alvarez-Yela AC, Casarez E, Jiang Y, Wang L, Kelly BE, Jenkins T, Ke E, Atkins KA, Janes KA, Slack-Davis JK, Zong H. Dichotomous ovarian cancer-initiating potential of Pax8+ cells revealed by a mouse genetic mosaic model. iScience 2023; 26:106742. [PMID: 37207276 PMCID: PMC10189502 DOI: 10.1016/j.isci.2023.106742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/08/2023] [Accepted: 04/20/2023] [Indexed: 05/21/2023] Open
Abstract
Different cellular compartments within a tissue present distinct cancer-initiating capacities. Current approaches to dissect such heterogeneity require cell-type-specific genetic tools based on a well-understood lineage hierarchy, which are lacking for many tissues. Here, we circumvented this hurdle and revealed the dichotomous capacity of fallopian tube Pax8+ cells in initiating ovarian cancer, utilizing a mouse genetic system that stochastically generates rare GFP-labeled mutant cells. Through clonal analysis and spatial profiling, we determined that only clones founded by rare, stem/progenitor-like Pax8+ cells can expand on acquiring oncogenic mutations whereas vast majority of clones stall immediately. Furthermore, expanded mutant clones undergo further attrition: many turn quiescent shortly after the initial expansion, whereas others sustain proliferation and manifest a bias toward Pax8+ fate, underlying early pathogenesis. Our study showcases the power of genetic mosaic system-based clonal analysis for revealing cellular heterogeneity of cancer-initiating capacity in tissues with limited prior knowledge of lineage hierarchy.
Collapse
Affiliation(s)
- Jianhao Zeng
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | | | - Eli Casarez
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Ying Jiang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Lixin Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Brianna E. Kelly
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Taylor Jenkins
- Department of Pathology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Eugene Ke
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Kristen A. Atkins
- Department of Pathology, University of Virginia Health System, Charlottesville, VA 22908, USA
- University of Virginia Cancer Center, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Kevin A. Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- University of Virginia Cancer Center, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Jill K. Slack-Davis
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
- University of Virginia Cancer Center, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Hui Zong
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
- University of Virginia Cancer Center, University of Virginia Health System, Charlottesville, VA 22903, USA
| |
Collapse
|
23
|
Tomas E, Shepherd TG. Insights into high-grade serous carcinoma pathobiology using three-dimensional culture model systems. J Ovarian Res 2023; 16:70. [PMID: 37038202 PMCID: PMC10088149 DOI: 10.1186/s13048-023-01145-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/28/2023] [Indexed: 04/12/2023] Open
Abstract
Epithelial ovarian cancer (EOC) research has become more complex as researchers try to fully understand the metastatic process. Especially as we delve into the concept of tumour dormancy, where cells transition between proliferative and dormant states to survive during disease progression. Thus, the in vitro models used to conduct this research need to reflect this vast biological complexity. The innovation behind the many three-dimensional (3D) spheroid models has been refined to easily generate reproducible spheroids so that we may understand the various molecular signaling changes of cells during metastasis and determine therapeutic efficacy of treatments. This ingenuity was then used to develop the 3D ex vivo patient-derived organoid model, as well as multiple co-culture model systems for EOC research. Although, researchers need to continue to push the boundaries of these current models for in vitro and even in vivo work in the future. In this review, we describe the 3D models already in use, where these models can be developed further and how we can use these models to gain the most knowledge on EOC pathogenesis and discover new targeted therapies.
Collapse
Affiliation(s)
- Emily Tomas
- London Regional Cancer Program, The Mary & John Knight Translational Ovarian Cancer Research Unit, 790 Commissioners Rd. E. Room A4-836, London, ON, N6A 4L6, Canada
- Department of Anatomy & Cell Biology, Western University, London, ON, Canada
| | - Trevor G Shepherd
- London Regional Cancer Program, The Mary & John Knight Translational Ovarian Cancer Research Unit, 790 Commissioners Rd. E. Room A4-836, London, ON, N6A 4L6, Canada.
- Department of Anatomy & Cell Biology, Western University, London, ON, Canada.
- Department of Obstetrics & Gynaecology, Western University, London, ON, Canada.
- Department of Oncology, Western University, London, ON, Canada.
| |
Collapse
|
24
|
Yu B, Nguyen EB, McCartney S, Strenk S, Valint D, Liu C, Haggerty C, Fredricks DN. Vaginal bacteria elicit acute inflammatory response in fallopian tube organoids: a model for pelvic inflammatory disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527402. [PMID: 36798329 PMCID: PMC9934550 DOI: 10.1101/2023.02.06.527402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Objective To facilitate in vitro mechanistic studies in pelvic inflammatory disease (PID) and subsequent tubal factor infertility, as well as ovarian carcinogenesis, we sought to establish patient tissue derived fallopian tube (FT) organoids and to study their inflammatory response to acute vaginal bacterial infection. Design Experimental study. Setting Academic medical and research center. Patients FT tissues were obtained from four patients after salpingectomy for benign gynecological diseases. Interventions We introduced acute infection in the FT organoid culture system by inoculating the organoid culture media with two common vaginal bacterial species, Lactobacillus crispatus and Fannyhessea vaginae . Main Outcome Measures The inflammatory response elicited in the organoids after acute bacterial infection was analyzed by the expression profile of 249 inflammatory genes. Results Compared to the negative controls that were not cultured with any bacteria, the organoids cultured with either bacterial species showed multiple differentially expressed inflammatory genes. Marked differences were noted between the Lactobacillus crispatus infected organoids and those infected by Fannyhessea vaginae . Genes from the C-X-C motif chemokine ligand (CXCL) family were highly upregulated in F. vaginae infected organoids. Flow cytometry showed that immune cells quickly disappeared during the organoid culture, indicating the inflammatory response observed with bacterial culture was generated by the epithelial cells in the organoids. Conclusion Patient tissue derived FT organoids respond to acute bacterial infection with upregulation of inflammatory genes specific to different vaginal bacterial species. FT organoids is a useful model system to study the host-pathogen interaction during bacterial infection which may facilitate mechanistic investigations in PID and its contribution to tubal factor infertility and ovarian carcinogensis.
Collapse
|
25
|
Generation and cryopreservation of feline oviductal organoids. Theriogenology 2023; 196:167-173. [PMID: 36423511 DOI: 10.1016/j.theriogenology.2022.11.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022]
Abstract
Next-generation in vitro culture model systems are needed to study the reproductive pathologies that affect domestic animals. These 3D culture models more closely mimic normal physiological function to allow a greater understanding of reproductive pathology and to trial therapeutics without the welfare concerns and the increased time and cost associated with live animal research. Recent advances with in vitro cell culture systems utilizing human and laboratory animal tissues have been reported, but implementation of these technologies in veterinary species has been slower. Organoids are a physiologically representative 3D cell culture system that can be maintained long-term. By combining organoid culture with cryopreservation, a long-term, experimental model can be available for year-round application, thus bypassing seasonality and reproductive tract availability restrictions. Here we report the generation and cryopreservation of feline oviductal organoids for the first time. Optimal culture medium for the generation of feline oviductal organoids was established, and organoids were successfully cryopreserved using three different freezing media with organoids from each treatment demonstrating comparable viability, growth rate, and protein expression after thawing and culture. Feline oviductal organoids may facilitate an in vivo-like environment that, in conjunction with co-culture for in vitro maturation and in vitro fertilization, may positively influence in vitro gamete and embryo development, embryo quality, and pregnancy rates after embryo transfer in domestic and nondomestic felids. Furthermore, readily available cryopreserved feline oviductal organoids will facilitate this co-culture, which is of particular importance to endangered felid breeding programs where tissue and gamete samples are often opportunistically obtained with little or no notice.
Collapse
|
26
|
Kumar A, Cai S, Allam M, Henderson S, Ozbeyler M, Saiontz L, Coskun AF. Single-Cell and Spatial Analysis of Emergent Organoid Platforms. Methods Mol Biol 2023; 2660:311-344. [PMID: 37191807 DOI: 10.1007/978-1-0716-3163-8_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Organoids have emerged as a promising advancement of the two-dimensional (2D) culture systems to improve studies in organogenesis, drug discovery, precision medicine, and regenerative medicine applications. Organoids can self-organize as three-dimensional (3D) tissues derived from stem cells and patient tissues to resemble organs. This chapter presents growth strategies, molecular screening methods, and emerging issues of the organoid platforms. Single-cell and spatial analysis resolve organoid heterogeneity to obtain information about the structural and molecular cellular states. Culture media diversity and varying lab-to-lab practices have resulted in organoid-to-organoid variability in morphology and cell compositions. An essential resource is an organoid atlas that can catalog protocols and standardize data analysis for different organoid types. Molecular profiling of individual cells in organoids and data organization of the organoid landscape will impact biomedical applications from basic science to translational use.
Collapse
Affiliation(s)
- Aditi Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Shuangyi Cai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Mayar Allam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Samuel Henderson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Melissa Ozbeyler
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Lilly Saiontz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, , Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
27
|
Chang YH, Wu KC, Harnod T, Ding DC. Comparison of the Cost and Effect of Combined Conditioned Medium and Conventional Medium for Fallopian Tube Organoid Cultures. Cell Transplant 2023; 32:9636897231160216. [PMID: 36919683 PMCID: PMC10021093 DOI: 10.1177/09636897231160216] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Fallopian tube epithelial cells (FTEC) are thought to be the cell of origin of high-grade serous ovarian carcinoma. FTEC organoids can be used as research models for the disease. Nevertheless, culturing organoids requires a medium supplemented with several expensive growth factors. We proposed that a combined conditioned medium based on the composition of the fallopian tubes, including epithelial, stromal, and endothelial cells could enhance FTEC organoid formation. We derived two primary culture cell lines from the fimbria portion of the fallopian tubes. The organoids were split into conventional or combined medium groups based on what medium they were grown in and compared. The number and size of the organoids were evaluated. Quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC) were used to evaluate gene and protein expression (PAX8, FOXJ1, beta-catenin, and stemness genes). Enzyme-linked immunosorbent assay was used to measure Wnt3a and RSPO1 in both mediums. DKK1 and LiCl were added to the mediums to evaluate their influence on beta-catenin signaling. The growth factor in the combined medium was evaluated by the growth factor array. We found that the conventional medium was better for organoids regarding proliferation (number and size). In addition, WNT3A and RSPO1 concentrations were too low in the combined medium and needed to be added making the cost equivalent to the conventional medium. However, the organoid formation rate was 100% in both groups. Furthermore, the combined medium group had higher PAX8 and stemness gene expression (OLFM4, SSEA4, LGR5, B3GALT5) when compared with the conventional medium group. Wnt signaling was evident in the organoids grown in the conventional medium but not in the combined medium. PLGF, IGFBP6, VEGF, bFGF, and SCFR were found to be enriched in the combined medium. In conclusion, the combined medium could successfully culture organoids and enhance PAX8 and stemness gene expression. However, the conventional medium was a better medium for organoid proliferation. The expense of both mediums was comparable. The benefit of using a combined medium requires further exploration.
Collapse
Affiliation(s)
- Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien.,Tzu Chi University, Hualien
| | - Kun-Chi Wu
- Tzu Chi University, Hualien.,Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien
| | - Tomor Harnod
- Tzu Chi University, Hualien.,Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien
| | - Dah-Ching Ding
- Tzu Chi University, Hualien.,Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien.,Institute of Medical Sciences, Collagen of Medicine, Tzu Chi University, Hualien
| |
Collapse
|
28
|
Qin G, Park ES, Chen X, Han S, Xiang D, Ren F, Liu G, Chen H, Yuan GC, Li Z. Distinct niche structures and intrinsic programs of fallopian tube and ovarian surface epithelial cells. iScience 2022; 26:105861. [PMID: 36624845 PMCID: PMC9823228 DOI: 10.1016/j.isci.2022.105861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022] Open
Abstract
Epithelial ovarian cancer (EOC) can originate from either fallopian tube epithelial (FTE) or ovarian surface epithelial (OSE) cells, but with different latencies and disease outcomes. To address the basis of these differences, we performed single cell RNA-sequencing of mouse cells isolated from the distal half of fallopian tube (FT) and surface layer of ovary. We find at the molecular level, FTE secretory stem/progenitor cells and OSE cells resemble mammary luminal progenitors and basal cells, respectively. An FT stromal subpopulation, enriched with Pdgfra + and Esr1 + cells, expresses multiple secreted factor (e.g., IGF1) and Hedgehog pathway genes and may serve as a niche for FTE cells. In contrast, Lgr5 + OSE cells express similar genes largely by themselves, raising a possibility that they serve as their own niche. The differences in intrinsic expression programs and niche organizations of FTE and OSE cells may contribute to their different courses toward the development of EOCs.
Collapse
Affiliation(s)
- Guyu Qin
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Eun-Sil Park
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Xueqing Chen
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Sen Han
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Dongxi Xiang
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Fang Ren
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Gang Liu
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Huidong Chen
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, MA 02215, USA
| | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, MA 02215, USA
| | - Zhe Li
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA,Corresponding author
| |
Collapse
|
29
|
Psilopatis I, Sykaras AG, Mandrakis G, Vrettou K, Theocharis S. Patient-Derived Organoids: The Beginning of a New Era in Ovarian Cancer Disease Modeling and Drug Sensitivity Testing. Biomedicines 2022; 11:1. [PMID: 36672509 PMCID: PMC9855526 DOI: 10.3390/biomedicines11010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer (OC) is the leading cause of death from gynecological malignancies. Despite great advances in treatment strategies, therapeutic resistance and the gap between preclinical data and actual clinical efficacy justify the necessity of developing novel models for investigating OC. Organoids represent revolutionary three-dimensional cell culture models, deriving from stem cells and reflecting the primary tissue's biology and pathology. The aim of the current review is to study the current status of mouse- and patient-derived organoids, as well as their potential to model carcinogenesis and perform drug screenings for OC. Herein, we describe the role of organoids in the assessment of high-grade serous OC (HGSOC) cells-of-origin, illustrate their use as promising preclinical OC models and highlight the advantages of organoid technology in terms of disease modelling and drug sensitivity testing.
Collapse
Affiliation(s)
- Iason Psilopatis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Bld 10, Goudi, 11527 Athens, Greece
- Department of Gynecology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt—Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Alexandros G. Sykaras
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Bld 10, Goudi, 11527 Athens, Greece
- Department of Cytopathology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Georgios Mandrakis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Bld 10, Goudi, 11527 Athens, Greece
| | - Kleio Vrettou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Bld 10, Goudi, 11527 Athens, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Bld 10, Goudi, 11527 Athens, Greece
| |
Collapse
|
30
|
Di Berardino C, Peserico A, Capacchietti G, Zappacosta A, Bernabò N, Russo V, Mauro A, El Khatib M, Gonnella F, Konstantinidou F, Stuppia L, Gatta V, Barboni B. High-Fat Diet and Female Fertility across Lifespan: A Comparative Lesson from Mammal Models. Nutrients 2022; 14:nu14204341. [PMID: 36297035 PMCID: PMC9610022 DOI: 10.3390/nu14204341] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022] Open
Abstract
Female reproduction focuses mainly on achieving fully grown follicles and competent oocytes to be successfully fertilized, as well as on nourishing the developing offspring once pregnancy occurs. Current evidence demonstrates that obesity and/or high-fat diet regimes can perturbate these processes, leading to female infertility and transgenerational disorders. Since the mechanisms and reproductive processes involved are not yet fully clarified, the present review is designed as a systematic and comparative survey of the available literature. The available data demonstrate the adverse influences of obesity on diverse reproductive processes, such as folliculogenesis, oogenesis, and embryo development/implant. The negative reproductive impact may be attributed to a direct action on reproductive somatic and germinal compartments and/or to an indirect influence mediated by the endocrine, metabolic, and immune axis control systems. Overall, the present review highlights the fragmentation of the current information limiting the comprehension of the reproductive impact of a high-fat diet. Based on the incidence and prevalence of obesity in the Western countries, this topic becomes a research challenge to increase self-awareness of dietary reproductive risk to propose solid and rigorous preventive dietary regimes, as well as to develop targeted pharmacological interventions.
Collapse
Affiliation(s)
- Chiara Di Berardino
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Alessia Peserico
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Correspondence:
| | - Giulia Capacchietti
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Alex Zappacosta
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council, A. Buzzati-Traverso Campus, via E. Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy
| | - Valentina Russo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Annunziata Mauro
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Mohammad El Khatib
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Francesca Gonnella
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Fani Konstantinidou
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Liborio Stuppia
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Barbara Barboni
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
31
|
Thompson RE, Meyers MA, Veeramachaneni DNR, Pukazhenthi BS, Hollinshead FK. Equine Oviductal Organoid Generation and Cryopreservation. Methods Protoc 2022; 5:mps5030051. [PMID: 35736552 PMCID: PMC9230449 DOI: 10.3390/mps5030051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022] Open
Abstract
Organoids are a type of three-dimensional (3D) cell culture that more closely mimic the in vivo environment and can be maintained in the long term. To date, oviductal organoids have only been reported in laboratory mice, women, and cattle. Equine oviductal organoids were generated and cultured for 42 days (including 3 passages and freeze-thawing at passage 1). Consistent with the reports in mouse and human oviductal organoids, the equine oviductal organoids revealed round cell clusters with a central lumen. Developing a 3D model of the mare oviduct may allow for an increased understanding of their normal physiology, including hormonal regulation. These organoids may provide an environment that mimics the in vivo equine oviduct and facilitate improved in vitro embryo production in equids.
Collapse
Affiliation(s)
- Riley E. Thompson
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (M.A.M.); (F.K.H.)
- Correspondence:
| | - Mindy A. Meyers
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (M.A.M.); (F.K.H.)
| | | | - Budhan S. Pukazhenthi
- Center for Species Survival, Smithsonian National Zoo and Conservation Biology Institute, Front Royal, VA 22630, USA;
| | - Fiona K. Hollinshead
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (M.A.M.); (F.K.H.)
| |
Collapse
|
32
|
Protocol to generate mouse oviduct epithelial organoids for viral transduction and whole-mount 3D imaging. STAR Protoc 2022; 3:101164. [PMID: 35199031 PMCID: PMC8844721 DOI: 10.1016/j.xpro.2022.101164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Epithelial cells lining the oviduct/fallopian tube are essential in reproduction and have been identified as the cell-of-origin in high-grade serous ovarian carcinoma (HGSOC). This protocol describes the generation of organoids from mouse oviduct epithelial cells, providing a powerful in vitro tool to study epithelial homeostasis and malignant transformation. We also outline a protocol for whole-mount immunofluorescence and 3D confocal imaging. In addition, we describe approaches of viral transduction to investigate gene function in organoid development and epithelial cell behavior. For complete details on the use and execution of this profile, please refer to Ford et al. (2021). A protocol for the isolation and organoid culture of mouse oviduct epithelial cells A procedure for organoid isolation, fixation, and whole mount immunofluorescence Organoids retain lineage specific markers and differentiation capacity Viral transduction methods to study gene function in disease and homeostasis
Collapse
|
33
|
Thompson RE, Bouma GJ, Hollinshead FK. The Roles of Extracellular Vesicles and Organoid Models in Female Reproductive Physiology. Int J Mol Sci 2022; 23:ijms23063186. [PMID: 35328607 PMCID: PMC8954697 DOI: 10.3390/ijms23063186] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/14/2022] [Accepted: 03/14/2022] [Indexed: 02/06/2023] Open
Abstract
Culture model systems that can recapitulate the anatomy and physiology of reproductive organs, such as three-dimensional (3D) organoid culture systems, limit the cost and welfare concerns associated with a research animal colony and provide alternative approaches to study specific processes in humans and animals. These 3D models facilitate a greater understanding of the physiological role of individual cell types and their interactions than can be accomplished with traditional monolayer culture systems. Furthermore, 3D culture systems allow for the examination of specific cellular, molecular, or hormonal interactions, without confounding factors that occur with in vivo models, and provide a powerful approach to study physiological and pathological reproductive conditions. The goal of this paper is to review and compare organoid culture systems to other in vitro cell culture models, currently used to study female reproductive physiology, with an emphasis on the role of extracellular vesicle interactions. The critical role of extracellular vesicles for intercellular communication in physiological processes, including reproduction, has been well documented, and an overview of the roles of extracellular vesicles in organoid systems will be provided. Finally, we will propose future directions for understanding the role of extracellular vesicles in normal and pathological conditions of reproductive organs, utilizing 3D organoid culture systems.
Collapse
|
34
|
Liu C, Pei M, Li Q, Zhang Y. Decellularized extracellular matrix mediates tissue construction and regeneration. Front Med 2022; 16:56-82. [PMID: 34962624 PMCID: PMC8976706 DOI: 10.1007/s11684-021-0900-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023]
Abstract
Contributing to organ formation and tissue regeneration, extracellular matrix (ECM) constituents provide tissue with three-dimensional (3D) structural integrity and cellular-function regulation. Containing the crucial traits of the cellular microenvironment, ECM substitutes mediate cell-matrix interactions to prompt stem-cell proliferation and differentiation for 3D organoid construction in vitro or tissue regeneration in vivo. However, these ECMs are often applied generically and have yet to be extensively developed for specific cell types in 3D cultures. Cultured cells also produce rich ECM, particularly stromal cells. Cellular ECM improves 3D culture development in vitro and tissue remodeling during wound healing after implantation into the host as well. Gaining better insight into ECM derived from either tissue or cells that regulate 3D tissue reconstruction or organ regeneration helps us to select, produce, and implant the most suitable ECM and thus promote 3D organoid culture and tissue remodeling for in vivo regeneration. Overall, the decellularization methodologies and tissue/cell-derived ECM as scaffolds or cellular-growth supplements used in cell propagation and differentiation for 3D tissue culture in vitro are discussed. Moreover, current preclinical applications by which ECM components modulate the wound-healing process are reviewed.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27109, USA.
| |
Collapse
|
35
|
Ford MJ, Yamanaka Y. Reprogramming Mouse Oviduct Epithelial Cells Using In Vivo Electroporation and CRISPR/Cas9-Mediated Genetic Manipulation. Methods Mol Biol 2022; 2429:367-377. [PMID: 35507174 DOI: 10.1007/978-1-0716-1979-7_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Advances in gene editing tools such as CRISPR/Cas9 have made precise in vivo gene editing possible, opening up avenues of research into somatic cell reprograming to study adult stem cells, homeostasis, and malignant transformation. Here we describe a method for CRISPR/Cas9 mediated in vivo gene editing, in combination with Cre-based lineage tracing via electroporation in the mouse oviduct. This method facilitates the delivery of multiple plasmids into oviduct epithelial cells, sufficient for studying homeostasis and generation of high-grade serous ovarian cancer (HGSOC) models.
Collapse
Affiliation(s)
- Matthew J Ford
- Department of Human Genetics, Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Yojiro Yamanaka
- Department of Human Genetics, Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
36
|
Francés-Herrero E, Lopez R, Hellström M, de Miguel-Gómez L, Herraiz S, Brännström M, Pellicer A, Cervelló I. OUP accepted manuscript. Hum Reprod Update 2022; 28:798-837. [PMID: 35652272 PMCID: PMC9629485 DOI: 10.1093/humupd/dmac025] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 04/13/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND To provide the optimal milieu for implantation and fetal development, the female reproductive system must orchestrate uterine dynamics with the appropriate hormones produced by the ovaries. Mature oocytes may be fertilized in the fallopian tubes, and the resulting zygote is transported toward the uterus, where it can implant and continue developing. The cervix acts as a physical barrier to protect the fetus throughout pregnancy, and the vagina acts as a birth canal (involving uterine and cervix mechanisms) and facilitates copulation. Fertility can be compromised by pathologies that affect any of these organs or processes, and therefore, being able to accurately model them or restore their function is of paramount importance in applied and translational research. However, innate differences in human and animal model reproductive tracts, and the static nature of 2D cell/tissue culture techniques, necessitate continued research and development of dynamic and more complex in vitro platforms, ex vivo approaches and in vivo therapies to study and support reproductive biology. To meet this need, bioengineering is propelling the research on female reproduction into a new dimension through a wide range of potential applications and preclinical models, and the burgeoning number and variety of studies makes for a rapidly changing state of the field. OBJECTIVE AND RATIONALE This review aims to summarize the mounting evidence on bioengineering strategies, platforms and therapies currently available and under development in the context of female reproductive medicine, in order to further understand female reproductive biology and provide new options for fertility restoration. Specifically, techniques used in, or for, the uterus (endometrium and myometrium), ovary, fallopian tubes, cervix and vagina will be discussed. SEARCH METHODS A systematic search of full-text articles available in PubMed and Embase databases was conducted to identify relevant studies published between January 2000 and September 2021. The search terms included: bioengineering, reproduction, artificial, biomaterial, microfluidic, bioprinting, organoid, hydrogel, scaffold, uterus, endometrium, ovary, fallopian tubes, oviduct, cervix, vagina, endometriosis, adenomyosis, uterine fibroids, chlamydia, Asherman’s syndrome, intrauterine adhesions, uterine polyps, polycystic ovary syndrome and primary ovarian insufficiency. Additional studies were identified by manually searching the references of the selected articles and of complementary reviews. Eligibility criteria included original, rigorous and accessible peer-reviewed work, published in English, on female reproductive bioengineering techniques in preclinical (in vitro/in vivo/ex vivo) and/or clinical testing phases. OUTCOMES Out of the 10 390 records identified, 312 studies were included for systematic review. Owing to inconsistencies in the study measurements and designs, the findings were assessed qualitatively rather than by meta-analysis. Hydrogels and scaffolds were commonly applied in various bioengineering-related studies of the female reproductive tract. Emerging technologies, such as organoids and bioprinting, offered personalized diagnoses and alternative treatment options, respectively. Promising microfluidic systems combining various bioengineering approaches have also shown translational value. WIDER IMPLICATIONS The complexity of the molecular, endocrine and tissue-level interactions regulating female reproduction present challenges for bioengineering approaches to replace female reproductive organs. However, interdisciplinary work is providing valuable insight into the physicochemical properties necessary for reproductive biological processes to occur. Defining the landscape of reproductive bioengineering technologies currently available and under development for women can provide alternative models for toxicology/drug testing, ex vivo fertility options, clinical therapies and a basis for future organ regeneration studies.
Collapse
Affiliation(s)
| | | | - Mats Hellström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lucía de Miguel-Gómez
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain
- Fundación IVI, IVI-RMA Global, Valencia, Spain
| | - Sonia Herraiz
- Fundación IVI, IVI-RMA Global, Valencia, Spain
- Reproductive Medicine Research Group, IIS La Fe, Valencia, Spain
| | - Mats Brännström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Stockholm IVF-EUGIN, Stockholm, Sweden
| | - Antonio Pellicer
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain
- IVI Roma Parioli, IVI-RMA Global, Rome, Italy
| | | |
Collapse
|
37
|
Lin Y, Wei Y, Jiang M, Tang X, Huang F, Yang X. Organoid culture of mouse fallopian tube epithelial stem cells with a thermo-reversible gelation polymer. Tissue Cell 2021; 73:101622. [PMID: 34454367 DOI: 10.1016/j.tice.2021.101622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023]
Abstract
In this study, a three-dimensional (3D) thermo-reversible gelation polymer (TGP) culture system was established for organoid culture of mouse fallopian tube (FT) epithelial stem cells (FTESCs) without cell isolation. FT tissues from 6- to 8-week-old ICR mice were digested with collagenase, and whole FT cells (FTCs) were inoculated into the TGP. After 6 days of culture, many spheres in the TGP formed. Some cells in the spheres were positive for 5-ethynyl-2'-deoxyuridine (EdU), a marker of cell proliferation. Furthermore, all the spheres that formed in the TGP were also labelled for EpCAM and LGR5. Some cells in the spheres were stained for PAX8, a secretory cell marker, and fewer cells were labelled with TUBB4, a ciliated cell marker. These results indicate that the 3D TGP culture system is a useful tool for organoid culture of FTESCs in vitro.
Collapse
Affiliation(s)
- YunXiu Lin
- Department of Histology and Embryology, School of Basic Medicine, Fujian Medical University, PR China.
| | - YuZhen Wei
- Department of Histology and Embryology, School of Basic Medicine, Fujian Medical University, PR China.
| | - MingZhu Jiang
- School of Clinical Medicine, Fujian Medical University, PR China.
| | - Xuan Tang
- School of Clinical Medicine, Fujian Medical University, PR China.
| | - Feng Huang
- Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medicine, Fujian Medical University, PR China.
| | - XinZhi Yang
- Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medicine, Fujian Medical University, PR China.
| |
Collapse
|
38
|
Ford MJ, Harwalkar K, Pacis AS, Maunsell H, Wang YC, Badescu D, Teng K, Yamanaka N, Bouchard M, Ragoussis J, Yamanaka Y. Oviduct epithelial cells constitute two developmentally distinct lineages that are spatially separated along the distal-proximal axis. Cell Rep 2021; 36:109677. [PMID: 34496237 DOI: 10.1016/j.celrep.2021.109677] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 05/25/2021] [Accepted: 08/17/2021] [Indexed: 01/02/2023] Open
Abstract
Owing to technical advances in single-cell biology, the appreciation of cellular heterogeneity has increased, which has aided our understanding of organ function, homeostasis, and disease progression. The oviduct (also known as the fallopian tube) is the distalmost portion of the female reproductive tract. It is essential for reproduction and the proposed origin of high-grade serous ovarian carcinoma (HGSOC). In mammals, the oviduct is morphologically segmented along the ovary-uterus axis into four evolutionally conserved regions. It is unclear, however, if there is a diversification of epithelial cell characteristics between these regions. In this study, we identify transcriptionally distinct populations of secretory and multiciliated cells restricted to the distal and proximal regions of the oviduct. We demonstrate that distal and proximal populations are distinct lineages specified early in Müllerian duct development and are maintained separately. These results aid our understanding of epithelial development, homeostasis, and initiation of disease from the oviduct.
Collapse
Affiliation(s)
- Matthew J Ford
- Rosalind and Morris Goodman Cancer Research Institute, Department of Human Genetics, McGill University, Montreal, QC H3A 1A3, Canada
| | - Keerthana Harwalkar
- Rosalind and Morris Goodman Cancer Research Institute, Department of Human Genetics, McGill University, Montreal, QC H3A 1A3, Canada
| | - Alain S Pacis
- Canadian Centre for Computational Genomics (C3G), Genome Quebec Innovation Centre, McGill University, Montreal, QC H3A 1A4, Canada
| | - Helen Maunsell
- Rosalind and Morris Goodman Cancer Research Institute, Department of Human Genetics, McGill University, Montreal, QC H3A 1A3, Canada
| | - Yu Chang Wang
- Department of Human Genetics, McGill University, Montreal, QC H3A OC7, Canada; McGill University and Genome Centre, Montreal, QC H3A 1A4, Canada
| | - Dunarel Badescu
- Department of Human Genetics, McGill University, Montreal, QC H3A OC7, Canada; McGill University and Genome Centre, Montreal, QC H3A 1A4, Canada
| | - Katie Teng
- Rosalind and Morris Goodman Cancer Research Institute, Department of Human Genetics, McGill University, Montreal, QC H3A 1A3, Canada
| | - Nobuko Yamanaka
- Rosalind and Morris Goodman Cancer Research Institute, Department of Human Genetics, McGill University, Montreal, QC H3A 1A3, Canada
| | - Maxime Bouchard
- Rosalind and Morris Goodman Cancer Research Institute and Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada
| | - Jiannis Ragoussis
- Department of Human Genetics, McGill University, Montreal, QC H3A OC7, Canada; McGill University and Genome Centre, Montreal, QC H3A 1A4, Canada; Department of Bioengineering, McGill University, Montreal, QC H3A 0C3, Canada
| | - Yojiro Yamanaka
- Rosalind and Morris Goodman Cancer Research Institute, Department of Human Genetics, McGill University, Montreal, QC H3A 1A3, Canada.
| |
Collapse
|
39
|
Sağraç D, Şişli HB, Şenkal S, Hayal TB, Şahin F, Doğan A. Organoids in Tissue Transplantation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1347:45-64. [PMID: 34164796 DOI: 10.1007/5584_2021_647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Improvements in stem cell-based research and genetic modification tools enable stem cell-based tissue regeneration applications in clinical therapies. Although inadequate cell numbers in culture, invasive isolation procedures, and poor survival rates after transplantation remain as major challenges, cell-based therapies are useful tools for tissue regeneration.Organoids hold a great promise for tissue regeneration, organ and disease modeling, drug testing, development, and genetic profiling studies. Establishment of 3D cell culture systems eliminates the disadvantages of 2D models in terms of cell adaptation and tissue structure and function. Organoids possess the capacity to mimic the specific features of tissue architecture, cell-type composition, and the functionality of real organs while preserving the advantages of simplified and easily accessible cell culture models. Thus, organoid technology might emerge as an alternative to cell and tissue transplantation. Although transplantation of various organoids in animal models has been demonstrated, liöitations related to vascularized structure formation, cell viability and functionality remain as obstacles in organoid-based transplantation therapies. Clinical applications of organoid-based transplantations might be possible in the near future, when limitations related to cell viability and tissue integration are solved. In this review, the literature was analyzed and discussed to explore the current status of organoid-based transplantation studies.
Collapse
Affiliation(s)
- Derya Sağraç
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Hatice Burcu Şişli
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Selinay Şenkal
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Taha Bartu Hayal
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Ayşegül Doğan
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
40
|
Organ-on-a-chip technology for the study of the female reproductive system. Adv Drug Deliv Rev 2021; 173:461-478. [PMID: 33831478 DOI: 10.1016/j.addr.2021.03.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/11/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022]
Abstract
Over the past decade, organs-on-a-chip and microphysiological systems have emerged as a disruptive in vitro technology for biopharmaceutical applications. By enabling new capabilities to engineer physiological living tissues and organ units in the precisely controlled environment of microfabricated devices, these systems offer great promise to advance the frontiers of basic and translational research in biomedical sciences. Here, we review an emerging body of interdisciplinary work directed towards harnessing the power of organ-on-a-chip technology for reproductive biology and medicine. The focus of this topical review is to provide an overview of recent progress in the development of microengineered female reproductive organ models with relevance to drug delivery and discovery. We introduce the engineering design of these advanced in vitro systems and examine their applications in the study of pregnancy, infertility, and reproductive diseases. We also present two case studies that use organ-on-a-chip design principles to model placental drug transport and hormonally regulated crosstalk between multiple female reproductive organs. Finally, we discuss challenges and opportunities for the advancement of reproductive organ-on-a-chip technology.
Collapse
|
41
|
Heremans R, Jan Z, Timmerman D, Vankelecom H. Organoids of the Female Reproductive Tract: Innovative Tools to Study Desired to Unwelcome Processes. Front Cell Dev Biol 2021; 9:661472. [PMID: 33959613 PMCID: PMC8093793 DOI: 10.3389/fcell.2021.661472] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
The pelviperineal organs of the female reproductive tract form an essential cornerstone of human procreation. The system comprises the ectodermal external genitalia, the Müllerian upper-vaginal, cervical, endometrial and oviductal derivatives, and the endodermal ovaries. Each of these organs presents with a unique course of biological development as well as of malignant degeneration. For many decades, various preclinical in vitro models have been employed to study female reproductive organ (patho-)biology, however, facing important shortcomings of limited expandability, loss of representativeness and inadequate translatability to the clinic. The recent emergence of 3D organoid models has propelled the field forward by generating powerful research tools that in vitro replicate healthy as well as diseased human tissues and are amenable to state-of-the-art experimental interventions. Here, we in detail review organoid modeling of the different female reproductive organs from healthy and tumorigenic backgrounds, and project perspectives for both scientists and clinicians.
Collapse
Affiliation(s)
- Ruben Heremans
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven (University of Leuven), Leuven, Belgium
- Cluster Woman and Child, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynecology, University Hospitals, KU Leuven, Leuven, Belgium
| | - Ziga Jan
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven (University of Leuven), Leuven, Belgium
- Cluster Woman and Child, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Gynecology, Klinikum Klagenfurt, Klagenfurt, Austria
| | - Dirk Timmerman
- Cluster Woman and Child, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynecology, University Hospitals, KU Leuven, Leuven, Belgium
| | - Hugo Vankelecom
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven (University of Leuven), Leuven, Belgium
| |
Collapse
|
42
|
Chumduri C, Turco MY. Organoids of the female reproductive tract. J Mol Med (Berl) 2021; 99:531-553. [PMID: 33580825 PMCID: PMC8026429 DOI: 10.1007/s00109-020-02028-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023]
Abstract
Healthy functioning of the female reproductive tract (FRT) depends on balanced and dynamic regulation by hormones during the menstrual cycle, pregnancy and childbirth. The mucosal epithelial lining of different regions of the FRT-ovaries, fallopian tubes, uterus, cervix and vagina-facilitates the selective transport of gametes and successful transfer of the zygote to the uterus where it implants and pregnancy takes place. It also prevents pathogen entry. Recent developments in three-dimensional (3D) organoid systems from the FRT now provide crucial experimental models that recapitulate the cellular heterogeneity and physiological, anatomical and functional properties of the organ in vitro. In this review, we summarise the state of the art on organoids generated from different regions of the FRT. We discuss the potential applications of these powerful in vitro models to study normal physiology, fertility, infections, diseases, drug discovery and personalised medicine.
Collapse
Affiliation(s)
- Cindrilla Chumduri
- Department of Microbiology, University of Würzburg, Biocenter, Würzburg, Germany.
- Max Planck Institute for Infection Biology, Berlin, Germany.
| | - Margherita Y Turco
- Department of Pathology, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, Cambridge, UK.
| |
Collapse
|
43
|
Bourdon G, Cadoret V, Charpigny G, Couturier-Tarrade A, Dalbies-Tran R, Flores MJ, Froment P, Raliou M, Reynaud K, Saint-Dizier M, Jouneau A. Progress and challenges in developing organoids in farm animal species for the study of reproduction and their applications to reproductive biotechnologies. Vet Res 2021; 52:42. [PMID: 33691745 PMCID: PMC7944619 DOI: 10.1186/s13567-020-00891-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
Within the past decades, major progress has been accomplished in isolating germ/stem/pluripotent cells, in refining culture medium and conditions and in establishing 3-dimensional culture systems, towards developing organoids for organs involved in reproduction in mice and to some extent in humans. Haploid male germ cells were generated in vitro from primordial germ cells. So were oocytes, with additional support from ovarian cells and subsequent follicle culture. Going on with the female reproductive tract, spherical oviduct organoids were obtained from adult stem/progenitor cells. Multicellular endometrial structures mimicking functional uterine glands were derived from endometrial cells. Trophoblastic stem cells were induced to form 3-dimensional syncytial-like structures and exhibited invasive properties, a crucial point for placentation. Finally, considering the embryo itself, pluripotent embryonic cells together with additional extra-embryonic cells, could self-organize into a blastoid, and eventually into a post-implantation-like embryo. Most of these accomplishments have yet to be reached in farm animals, but much effort is devoted towards this goal. Here, we review the progress and discuss the specific challenges of developing organoids for the study of reproductive biology in these species. We consider the use of such organoids in basic research to delineate the physiological mechanisms involved at each step of the reproductive process, or to understand how they are altered by environmental factors relevant to animal breeding. We evaluate their potential in reproduction of animals with a high genetic value, from a breeding point of view or in the context of preserving local breeds with limited headcounts.
Collapse
Affiliation(s)
- Guillaume Bourdon
- INRAE, CNRS, Université de Tours, IFCE, PRC, 37380, Nouzilly, France
| | - Véronique Cadoret
- INRAE, CNRS, Université de Tours, IFCE, PRC, 37380, Nouzilly, France
- CHU Bretonneau, Médecine et Biologie de la Reproduction-CECOS, 37044, Tours, France
| | - Gilles Charpigny
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire D'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Anne Couturier-Tarrade
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire D'Alfort, BREED, 94700, Maisons-Alfort, France
| | | | - Maria-José Flores
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire D'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Pascal Froment
- INRAE, CNRS, Université de Tours, IFCE, PRC, 37380, Nouzilly, France
| | - Mariam Raliou
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire D'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Karine Reynaud
- INRAE, CNRS, Université de Tours, IFCE, PRC, 37380, Nouzilly, France
| | - Marie Saint-Dizier
- INRAE, CNRS, Université de Tours, IFCE, PRC, 37380, Nouzilly, France
- Faculty of Sciences and Techniques, University of Tours, 37200, Tours, France
| | - Alice Jouneau
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France.
- Ecole Nationale Vétérinaire D'Alfort, BREED, 94700, Maisons-Alfort, France.
| |
Collapse
|
44
|
Ding DC, Chang YH, Wu KC, Harnod T. The organoid: A research model for ovarian cancer. Tzu Chi Med J 2021; 34:255-260. [PMID: 35912056 PMCID: PMC9333109 DOI: 10.4103/tcmj.tcmj_63_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 11/29/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is a heterogeneous disease with a variety of distinct clinical and molecular characteristics. The currently available and common research models for EOC include tumor cell lines and patient-derived xenografts. However, these models have certain shortcomings: establishing a cell line is time-consuming, loss of genetic traits after long-term culture is a possibility, and investment is required in terms of animal care facilities. Therefore, better research models are required. Organoid technology was originally developed from colorectal cancer. Tumor organoid is a three-dimensional culture system and can help accurately recapture the tumor phenotype from the original tumor. Tumor organoid systems can overcome the above-mentioned shortcomings of the currently available research models. The organoid model can be used for culturing ovarian cancer subtypes, screening drugs, assessing genomes, and establishing biobanks. However, the currently available organoid models can only culture one type of cells, epithelial cells. Therefore, an organoid-on-a-chip device can be developed in the future to provide a microenvironment for cell–cell, cell–matrix, and cell–media interactions. Thus, organoid models can be used in ovarian cancer research and can generate a simulated in vivo system, enabling studies on the heterogeneity of ovarian cancer.
Collapse
|
45
|
Examination of expression patterns of WNT signaling in the human fallopian tubes. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
46
|
Sheta R, Bachvarova M, Plante M, Renaud MC, Sebastianelli A, Gregoire J, Navarro JM, Perez RB, Masson JY, Bachvarov D. Development of a 3D functional assay and identification of biomarkers, predictive for response of high-grade serous ovarian cancer (HGSOC) patients to poly-ADP ribose polymerase inhibitors (PARPis): targeted therapy. J Transl Med 2020; 18:439. [PMID: 33213473 PMCID: PMC7678187 DOI: 10.1186/s12967-020-02613-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 11/06/2020] [Indexed: 11/13/2022] Open
Abstract
Background Poly(ADP-ribose) polymerase inhibitors (PARPis) specifically target homologous recombination deficiency (HRD) cells and display good therapeutic effect in women with advanced-stage BRCA1/2-mutated breast and epithelial ovarian cancer (EOC). However, about 50% of high grade serous ovarian cancers (HGSOC) present with HRD due to epigenetic BRCA1 inactivation, as well as genetic/epigenetic inactivation(s) of other HR genes, a feature known as “BRCAness”. Therefore, there is a potential for extending the use of PARPis to these patients if HR status can be identified. Methods We have developed a 3D (spheroid) functional assay to assess the sensitivity of two PARPis (niraparib and olaparib) in ascites-derived primary cell cultures (AsPCs) from HGSOC patients. A method for AsPCs preparation was established based on a matrix (agarose), allowing for easy isolation and successive propagation of monolayer and 3D AsPCs. Based on this method, we performed cytotoxicity assays on 42 AsPCs grown both as monolayers and spheroids. Results The response to PARPis treatment in monolayer AsPCs, was significantly higher, compared to 3D AsPCs, as 88% and 52% of the monolayer AsPCs displayed sensitivity to niraparib and olaparib respectively, while 66% of the 3D AsPCs were sensitive to niraparib and 38% to olaparib, the latter being more consistent with previous estimates of HRD (40%–60%) in EOC. Moreover, niraparib displayed a significantly stronger cytotoxic effect in both in 3D and monolayer AsPCs, which was confirmed by consecutive analyses of the HR pathway activity (γH2AX foci formation) in PARPis-sensitive and resistant AsPCs. Global gene expression comparison of 6 PARPi-resistant and 6 PARPi-sensitive 3D AsPCs was indicative for the predominant downregulation of numerous genes and networks with previously demonstrated roles in EOC chemoresistance, suggesting that the PARPis-sensitive AsPCs could display enhanced sensitivity to other chemotherapeutic drugs, commonly applied in cancer management. Microarray data validation identified 24 potential gene biomarkers associated with PARPis sensitivity. The differential expression of 7 selected biomarkers was consecutively confirmed by immunohistochemistry in matched EOC tumor samples. Conclusion The application of this assay and the potential biomarkers with possible predictive significance to PARPis therapy of EOC patients now need testing in the setting of a clinical trial.
Collapse
Affiliation(s)
- Razan Sheta
- Department of Molecular Medicine, Université Laval, Québec, QC, G1V 0A6, Canada.,Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada
| | - Magdalena Bachvarova
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada
| | - Marie Plante
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Marie-Claude Renaud
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Alexandra Sebastianelli
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Jean Gregoire
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Jamilet Miranda Navarro
- Bioinformatics Department, Center for Genetic Engineering and Biotechnology, 10600, Havana, CP, Cuba
| | - Ricardo Bringas Perez
- Bioinformatics Department, Center for Genetic Engineering and Biotechnology, 10600, Havana, CP, Cuba
| | - Jean-Yves Masson
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Molecular Biology, Medical Biochemistry, and Pathology, Laval University Cancer Research Center, Québec, QC, G1V 0A6, Canada
| | - Dimcho Bachvarov
- Department of Molecular Medicine, Université Laval, Québec, QC, G1V 0A6, Canada. .,Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.
| |
Collapse
|
47
|
Zhu M, Iwano T, Takeda S. Fallopian Tube Basal Stem Cells Reproducing the Epithelial Sheets In Vitro-Stem Cell of Fallopian Epithelium. Biomolecules 2020; 10:biom10091270. [PMID: 32899226 PMCID: PMC7565394 DOI: 10.3390/biom10091270] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/30/2020] [Accepted: 08/31/2020] [Indexed: 12/30/2022] Open
Abstract
The fallopian tube (FT) is an important reproductive organ in females. The luminal epithelium of the FT is composed of highly polarized secretory and ciliated cells. Recently, accumulating lines of evidence have suggested that the origin of high-grade serous ovarian carcinoma (HGSC) is fallopian tube epithelial cells (FTECs). Due to the lack of a high-fidelity model for FTECs in vitro, homeostasis, differentiation, as well as the transformation of FTECs are still enigmatic. In this study, we optimized the culture condition for the stable expansion of basal stem cells, as well as inducing differentiation of basal cells into polarized secretory and ciliated cells in the air-liquid interface (ALI) condition suitable for long-term culture. This storable culture method of FTECs provides a versatile platform for studying differentiation mechanisms, intercellular communication, and transformation to HGSC, as well as the physiological function of the FT in vitro.
Collapse
Affiliation(s)
| | - Tomohiko Iwano
- Correspondence: (T.I.); (S.T.); Tel.: +81-55-273-9471 (T.I.); +81-55-273-6726 (S.T.); Fax: +81-55-273-9473 (T.I. & S.T.)
| | - Sen Takeda
- Correspondence: (T.I.); (S.T.); Tel.: +81-55-273-9471 (T.I.); +81-55-273-6726 (S.T.); Fax: +81-55-273-9473 (T.I. & S.T.)
| |
Collapse
|
48
|
Cochrane DR, Campbell KR, Greening K, Ho GC, Hopkins J, Bui M, Douglas JM, Sharlandjieva V, Munzur AD, Lai D, DeGrood M, Gibbard EW, Leung S, Boyd N, Cheng AS, Chow C, Lim JL, Farnell DA, Kommoss S, Kommoss F, Roth A, Hoang L, McAlpine JN, Shah SP, Huntsman DG. Single cell transcriptomes of normal endometrial derived organoids uncover novel cell type markers and cryptic differentiation of primary tumours. J Pathol 2020; 252:201-214. [PMID: 32686114 DOI: 10.1002/path.5511] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/11/2020] [Accepted: 07/14/2020] [Indexed: 01/04/2023]
Abstract
Endometrial carcinoma, the most common gynaecological cancer, develops from endometrial epithelium which is composed of secretory and ciliated cells. Pathologic classification is unreliable and there is a need for prognostic tools. We used single cell sequencing to study organoid model systems derived from normal endometrial endometrium to discover novel markers specific for endometrial ciliated or secretory cells. A marker of secretory cells (MPST) and several markers of ciliated cells (FAM92B, WDR16, and DYDC2) were validated by immunohistochemistry on organoids and tissue sections. We performed single cell sequencing on endometrial and ovarian tumours and found both secretory-like and ciliated-like tumour cells. We found that ciliated cell markers (DYDC2, CTH, FOXJ1, and p73) and the secretory cell marker MPST were expressed in endometrial tumours and positively correlated with disease-specific and overall survival of endometrial cancer patients. These findings suggest that expression of differentiation markers in tumours correlates with less aggressive disease, as would be expected for tumours that retain differentiation capacity, albeit cryptic in the case of ciliated cells. These markers could be used to improve the risk stratification of endometrial cancer patients, thereby improving their management. We further assessed whether consideration of MPST expression could refine the ProMiSE molecular classification system for endometrial tumours. We found that higher expression levels of MPST could be used to refine stratification of three of the four ProMiSE molecular subgroups, and that any level of MPST expression was able to significantly refine risk stratification of the copy number high subgroup which has the worst prognosis. Taken together, this shows that single cell sequencing of putative cells of origin has the potential to uncover novel biomarkers that could be used to guide management of cancers. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Dawn R Cochrane
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | - Kieran R Campbell
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | - Kendall Greening
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Germain C Ho
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - James Hopkins
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | - Minh Bui
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | - J Maxwell Douglas
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | | | - Aslı D Munzur
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | - Daniel Lai
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | - Maya DeGrood
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Evan W Gibbard
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Samuel Leung
- Genetic Pathology Evaluation Centre, Vancouver General Hospital, Vancouver, BC, Canada
| | - Niki Boyd
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | - Angela S Cheng
- Genetic Pathology Evaluation Centre, Vancouver General Hospital, Vancouver, BC, Canada
| | - Christine Chow
- Genetic Pathology Evaluation Centre, Vancouver General Hospital, Vancouver, BC, Canada
| | - Jamie Lp Lim
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - David A Farnell
- Department of Anatomical Pathology, Vancouver General Hospital, Vancouver, BC, Canada
| | - Stefan Kommoss
- Department of Obstetrics and Gynecology, University of Tübingen, Tübingen, Germany
| | - Friedrich Kommoss
- Institute of Pathology, Medizin Campus Bodensee, Friedrichshafen, Germany
| | - Andrew Roth
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | - Lien Hoang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jessica N McAlpine
- Department of Gynecology and Obstetrics, University of British Columbia, Vancouver, BC, Canada
| | - Sohrab P Shah
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - David G Huntsman
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
49
|
Heidari-Khoei H, Esfandiari F, Hajari MA, Ghorbaninejad Z, Piryaei A, Baharvand H. Organoid technology in female reproductive biomedicine. Reprod Biol Endocrinol 2020; 18:64. [PMID: 32552764 PMCID: PMC7301968 DOI: 10.1186/s12958-020-00621-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Recent developments in organoid technology are revolutionizing our knowledge about the biology, physiology, and function of various organs. Female reproductive biology and medicine also benefit from this technology. Organoids recapitulate features of different reproductive organs including the uterus, fallopian tubes, and ovaries, as well as trophoblasts. The genetic stability of organoids and long-lasting commitment to their tissue of origin during long-term culture makes them attractive substitutes for animal and in vitro models. Despite current limitations, organoids offer a promising platform to address fundamental questions regarding the reproductive system's physiology and pathology. They provide a human source to harness stem cells for regenerative medicine, heal damaged epithelia in specific diseases, and study biological processes in healthy and pathological conditions. The combination of male and female reproductive organoids with other technologies, such as microfluidics technology, would enable scientists to create a multi-organoid-on-a-chip platform for the next step to human-on-a-chip platforms for clinical applications, drug discovery, and toxicology studies. The present review discusses recent advances in producing organoid models of reproductive organs and highlights their applications, as well as technical challenges and future directions.
Collapse
Affiliation(s)
- Heidar Heidari-Khoei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, 1665659911, Iran
| | - Fereshteh Esfandiari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, 1665659911, Iran
| | - Mohammad Amin Hajari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, 1665659911, Iran
| | - Zeynab Ghorbaninejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, 1665659911, Iran
| | - Abbas Piryaei
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19395-4719, Tehran, Iran.
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, 1665659911, Iran.
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
50
|
WNT and inflammatory signaling distinguish human Fallopian tube epithelial cell populations. Sci Rep 2020; 10:9837. [PMID: 32555344 PMCID: PMC7300082 DOI: 10.1038/s41598-020-66556-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/21/2020] [Indexed: 12/18/2022] Open
Abstract
Many high-grade serous carcinomas (HGSCs) likely originate in the distal region of the Fallopian tube’s epithelium (TE) before metastasizing to the ovary. Unfortunately, molecular mechanisms promoting malignancy in the distal TE are obfuscated, largely due to limited primary human TE gene expression data. Here we report an in depth bioinformatic characterization of 34 primary TE mRNA-seq samples. These samples were prepared from proximal and distal TE regions of 12 normal Fallopian tubes. Samples were segregated based on their aldehyde dehydrogenase (ALDH) activity. Distal cells form organoids with higher frequency and larger size during serial organoid formation assays when compared to proximal cells. Consistent with enrichment for stem/progenitor cells, ALDH+ cells have greater WNT signaling. Comparative evaluation of proximal and distal TE cell population’s shows heightened inflammatory signaling in distal differentiated (ALDH−) TE. Furthermore, comparisons of proximal and distal TE cell populations finds that the distal ALDH+ TE cells exhibit pronounced expression of gene sets characteristic of HGSC sub-types. Overall, our study indicates increased organoid forming capacity, WNT/inflammatory signaling, and HGSC signatures underlie differences between distal and proximal regions of the human TE. These findings provide the basis for further mechanistic studies of distal TE susceptibility to the malignant transformation.
Collapse
|