Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Biofabrication ; 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38986466

ABSTRACT

This study presents a biphasic approach to overcome the limitations of current testicular organoid (TO) cultures, including histological heterogeneity, germ cell loss and absence of spermatogenesis. Agarose microwells were utilized to create TOs from prepubertal C57BL/6J testicular cells. First emphasis was on improving germ cell survival during the initial 2-week reorganization phase by comparing α-MEM + 10% KSR medium, known to support TO generation in mice, to three optimized media (1-3). Cell densities and culture dynamics were also tested to recreate histological resemblance to testes. After optimizing germ cell survival and cell organization, the effect of growth factors and immunomodulation through CD45+ immune cell depletion or dexamethasone (DEX) supplementation were assessed for enhancing spermatogenesis during the subsequent differentiation phase. Testicular cells self-reorganized into organoids resembling the testicular anatomical unit, characterized by one tubule-like structure surrounded by interstitium. Media 1 3 proved superior for organoid growth during the reorganization phase, with TOs in medium 3 exhibiting germ cell numbers (7.4 ± 4.8%) comparable to controls (9.3 ± 5.3%). Additionally, 37 ± 30% demonstrated organized histology from 32 × 103 cells under static conditions. Switching to α-MEM + 10% KSR during the differentiation phase increased formation efficiency to 85 ± 7%, along with elevated germ cell numbers, testosterone production (3.1 ± 0.9 ng/mL) and generation of γH2AX+ spermatid-like cells (steps 8-11, 1.2 ± 2.2% of the total). Adding differentiation factors to the α-MEM increased spermatid-like cell numbers to 2.9 ± 5.9%, confirmed through positive staining for CREM, TP1, and PNA. Although, these remained diploid with irregular nuclear maturation. DEX supplementation had no additional effect, and immune cell depletion adversely impacted TO formation. The manipulability of TOs offers advantages in studying male infertility and exploring therapies, with scalability enabling high-throughput chemical screening and reducing animal usage in reproductive toxicity and drug discovery studies.

3.
Methods Mol Biol ; 2770: 135-149, 2024.
Article in English | MEDLINE | ID: mdl-38351452

ABSTRACT

Testes have a complex architecture that is compartmentalized into seminiferous tubules with a diameter of approximatively 200 µm in which the germ cells differentiate, surrounded by a basement membrane and interstitium. 3D bioprinting might be used to recreate the compartmentalized testicular architecture in vitro. Directed by a software program, pneumatic microextrusion printers can deposit 3D layers of hydrogel-encapsulated interstitial cells in a controlled manner by applying pressure. Once macroporous-shaped scaffolds resembling seminiferous tubules have been bioprinted with interstitial cells, the epithelial cell fraction can be seeded in the macropores to resemble the in vivo testicular architecture. Moreover, macropores can serve as a delimitation for all testicular cells to reorganize and improve the supply of nutrients to cells through the 3D constructs.


Subject(s)
Bioprinting , Spermatogenesis , Male , Animals , Mice , Testis , Seminiferous Tubules , Tissue Scaffolds , Leydig Cells , Hydrogels , Tissue Engineering , Printing, Three-Dimensional
4.
Biol Open ; 13(1)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38180243

ABSTRACT

The 15th Network of Young Researchers in Andrology (NYRA) meeting, held at the Palace de Caux, Switzerland, served as a valuable platform to disseminate cutting-edge research and facilitate interactions among early-career researchers and trainees in andrology from around the world. Preceding the 22nd European Testis Workshop, the 2-day event brought together participants from a variety of countries to discuss a range of topics pertaining to men's reproductive health and biology. Specific focuses included piRNAs in mammalian reproduction, biomolecules enhancing sperm physiology, advances in in vitro spermatogenesis, reproductive strategies across species, and career development. A dedicated 'scientific speed-dating' social event also stood out, encouraging cross-disciplinary collaborations and strengthening ties within the scientific community. The high participation rate of the meeting highlighted its value in connecting the andrology community. Finally, the announcement of NYRA's merger with the European Academy of Andrology (EAA) marked a pivotal moment, enabling NYRA to support young researchers while collaborating with the EAA to advance andrology research. The 15th NYRA meeting played a crucial role in enhancing knowledge dissemination and andrology research, empowering young researchers, and addressing key challenges in male infertility.


Subject(s)
Andrology , Animals , Humans , Male , Reproductive Health , Semen , Reproduction , Power, Psychological , Mammals
5.
Biol Open ; 12(5)2023 05 15.
Article in English | MEDLINE | ID: mdl-37249510

ABSTRACT

The 14th Network of Young Researchers in Andrology (NYRA) meeting was a 2-day conference held at the University of East Anglia in Norwich, UK, organized by the NYRA. The meeting brought together researchers and experts to discuss and exchange ideas on male infertility and spermatogenesis. The meeting covered a wide range of topics related to male germline research, including the impact of mutations in the male germline on future generations, the use of innovative sequencing tools for the study of male infertility, and the intricate germline epigenome. The impact of aging on spermatogenesis was also discussed, with a focus on the increased DNA fragmentation rates, changes in DNA methylation patterns, and longer telomeres associated with aging sperm. Additionally, progress on fertility preservation options for children undergoing gonadotoxic cancer treatments was presented. The meeting also featured workshops on leadership and career development strategies in science, providing a valuable opportunity for young researchers to learn from experts in the field and exchange ideas with their peers. Overall, the meeting provided a platform for researchers to discuss the latest developments in male germline research, highlighting the importance of empowering young researchers to tackle male reproductive health.


Subject(s)
Andrology , Fertility Preservation , Infertility, Male , Child , Male , Humans , Reproductive Health , Semen , Infertility, Male/genetics , Infertility, Male/therapy
6.
Front Physiol ; 12: 757565, 2021.
Article in English | MEDLINE | ID: mdl-35002756

ABSTRACT

Short-term germ cell survival and central tissue degeneration limit organoid cultures. Here, testicular organoids (TOs) were generated from two different mouse strains in 3D printed one-layer scaffolds (1LS) at the air-medium interface displaying tubule-like structures and Leydig cell functionality supporting long-term survival and differentiation of germ cells to the meiotic phase. Chimeric TOs, consisting of a mixture of primary testicular cells and EGFP+ germline stem (GS) cells, were cultured in two-layer scaffolds (2LSs) for better entrapment. They showed an improved spheroidal morphology consisting of one intact tubule-like structure and surrounding interstitium, representing the functional unit of a testis. However, GS cells did not survive long-term culture. Consequently, further optimization of the culture medium is required to enhance the maintenance and differentiation of germ cells. The opportunities TOs offer to manipulate somatic and germ cells are essential for the study of male infertility and the search for potential therapies.

7.
Andrology ; 8(4): 879-891, 2020 07.
Article in English | MEDLINE | ID: mdl-31823507

ABSTRACT

BACKGROUND: The testicular organoid concept has recently been introduced in tissue engineering to refer to testicular cell organizations modeling testicular architecture and function. The testicular organoid approach gives control over which and how cells reaggregate, which is not possible in organotypic cultures, thereby extending the applicability of in-vitro spermatogenesis (IVS) systems. However, it remains unclear which culture method and medium allow reassociation of testicular cells into a functional testicular surrogate in-vitro. OBJECTIVE: The aim of this paper is to review the different strategies that have been used in an attempt to create testicular organoids and generate spermatozoa. We want to provide an up-to-date list on culture methodologies and media compositions that have been used and determine their role in regulating tubulogenesis and differentiation of testicular cells. SEARCH METHOD: A literature search was conducted in PubMed, Web of Science, and Scopus to select studies reporting the reorganization of testicular cell suspensions in-vitro, using the keywords: three-dimensional culture, in-vitro spermatogenesis, testicular organoid, testicular scaffold, and tubulogenesis. Papers published before the August 1, 2019, were selected. OUTCOME: Only a limited number of studies have concentrated on recreating the testicular architecture in-vitro. While some advances have been made in the testicular organoid research in terms of cellular reorganization, none of the described culture systems is adequate for the reproduction of both the testicular architecture and IVS. CONCLUSION: Further improvements in culture methodology and medium composition have to be made before being able to provide both testicular tubulogenesis and spermatogenesis in-vitro.


Subject(s)
Cell Culture Techniques , Organoids/cytology , Spermatogenesis/physiology , Testis/cytology , Animals , Cell Differentiation/physiology , Humans , Male , Spermatozoa/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...