Bioengineering Technologies to Advance Reproductive Health

A special issue of Bioengineering (ISSN 2306-5354). This special issue belongs to the section "Regenerative Engineering".

Deadline for manuscript submissions: 31 May 2024 | Viewed by 12423

Special Issue Editor


E-Mail Website
Guest Editor
1. Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
2. Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
Interests: reproductive biology; biofabrication; stem cells; fertility preservation; endocrinology; regenerative medicine; gonadal development

Special Issue Information

Dear Colleagues,

Over the last several years, engineering technologies and bioengineered systems have been used to investigate the physical properties of reproductive tissues, study the dynamics of gametogenesis, and integrate multitissue systems. The purpose of this Special Issue is to identify recent advancements in reproductive biology and regenerative medicine through bioengineering. Topics for publication include the use of engineered microenvironments or materials to interrogate the molecular mechanisms or physiology of reproductive organs or cells, the use of microfluidics to integrate multiple tissues and support gamete quality selection or fertilization, and the use of engineering technologies to characterize the physical properties of reproductive tissues.

Original research submissions are highly encouraged, and review articles that focus on research from the last five years as well as conceptualize the promise of future contributions to understanding reproductive biology and supporting future innovations in regenerative medicine will also be considered.

Dr. Monica M Laronda
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Bioengineering is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • engineered microenvironment
  • scaffolds
  • reproductive biology
  • fertilization
  • pregnancy
  • microfluidics
  • tissue engineering
  • biofabrication
  • regenerative medicine

Published Papers (6 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

15 pages, 1326 KiB  
Article
Generation of Tailored Extracellular Matrix Hydrogels for the Study of In Vitro Folliculogenesis in Response to Matrisome-Dependent Biochemical Cues
by Hannah B. McDowell, Kathryn L. McElhinney, Elizabeth L. Tsui and Monica M. Laronda
Bioengineering 2024, 11(6), 543; https://doi.org/10.3390/bioengineering11060543 - 25 May 2024
Viewed by 344
Abstract
While ovarian tissue cryopreservation (OTC) is an important fertility preservation option, it has its limitations. Improving OTC and ovarian tissue transplantation (OTT) must include extending the function of reimplanted tissue by reducing the extensive activation of primordial follicles (PMFs) and eliminating the risk [...] Read more.
While ovarian tissue cryopreservation (OTC) is an important fertility preservation option, it has its limitations. Improving OTC and ovarian tissue transplantation (OTT) must include extending the function of reimplanted tissue by reducing the extensive activation of primordial follicles (PMFs) and eliminating the risk of reimplanting malignant cells. To develop a more effective OTT, we must understand the effects of the ovarian microenvironment on folliculogenesis. Here, we describe a method for producing decellularized extracellular matrix (dECM) hydrogels that reflect the protein composition of the ovary. These ovarian dECM hydrogels were engineered to assess the effects of ECM on in vitro follicle growth, and we developed a novel method for selectively removing proteins of interest from dECM hydrogels. Finally, we validated the depletion of these proteins and successfully cultured murine follicles encapsulated in the compartment-specific ovarian dECM hydrogels and these same hydrogels depleted of EMILIN1. These are the first, optically clear, tailored tissue-specific hydrogels that support follicle survival and growth comparable to the “gold standard” alginate hydrogels. Furthermore, depleted hydrogels can serve as a novel tool for many tissue types to evaluate the impact of specific ECM proteins on cellular and molecular behavior. Full article
(This article belongs to the Special Issue Bioengineering Technologies to Advance Reproductive Health)
Show Figures

Graphical abstract

18 pages, 9581 KiB  
Article
Comparative Tensile Properties and Collagen Patterns in Domestic Cat (Felis catus) and Dog (Canis lupus familiaris) Ovarian Cortical Tissues
by Jennifer B. Nagashima, Shoshana Zenilman, April Raab, Helim Aranda-Espinoza and Nucharin Songsasen
Bioengineering 2023, 10(11), 1285; https://doi.org/10.3390/bioengineering10111285 - 4 Nov 2023
Viewed by 1261
Abstract
The importance of the ovarian extracellular environment and tissue rigidity on follicle survival and development has gained attention in recent years. Our laboratory has anecdotally observed differences in the rigidity of domestic cat and dog ovarian cortical tissues, which have been postulated to [...] Read more.
The importance of the ovarian extracellular environment and tissue rigidity on follicle survival and development has gained attention in recent years. Our laboratory has anecdotally observed differences in the rigidity of domestic cat and dog ovarian cortical tissues, which have been postulated to underlie the differences in in vitro culture responses between the species, wherein cat ovarian tissues display higher survival in extended incubation. Here, the tensile strengths of cat and dog ovarian cortical tissues were compared via micropipette aspiration. The underlying collagen patterns, including fiber length, thickness, alignment, curvature, branch points and end points, and overall tissue lacunary and high-density matrix (HDM) were quantified via picrosirius red staining and TWOMBLI analysis. Finally, we explored the potential of MMP (−1 and −9) and TIMP1 supplementation in modulating tissue rigidity, collagen structure, and follicle activation in vitro. No differences in stiffness were observed between cat or dog cortical tissues, or pre- versus post-pubertal status. Cat ovarian collagen was characterized by an increased number of branch points, thinner fibers, and lower HDM compared with dog ovarian collagen, and cat tissues exposed to MMP9 in vitro displayed a reduced Young’s modulus. Yet, MMP exposure had a minor impact on follicle development in vitro in either species. This study contributes to our growing understanding of the interactions among the physical properties of the ovarian microenvironment, collagen patterns, and follicle development in vitro. Full article
(This article belongs to the Special Issue Bioengineering Technologies to Advance Reproductive Health)
Show Figures

Figure 1

18 pages, 5263 KiB  
Article
Encapsulated Allografts Preclude Host Sensitization and Promote Ovarian Endocrine Function in Ovariectomized Young Rhesus Monkeys and Sensitized Mice
by James R. Day, Colleen L. Flanagan, Anu David, Dennis J. Hartigan-O’Connor, Mayara Garcia de Mattos Barbosa, Michele L. Martinez, Charles Lee, Jenna Barnes, Evan Farkash, Mary Zelinski, Alice Tarantal, Marilia Cascalho and Ariella Shikanov
Bioengineering 2023, 10(5), 550; https://doi.org/10.3390/bioengineering10050550 - 3 May 2023
Cited by 2 | Viewed by 1736
Abstract
Transplantation of allogeneic donor ovarian tissue holds great potential for female cancer survivors who often experience premature ovarian insufficiency. To avoid complications associated with immune suppression and to protect transplanted ovarian allografts from immune-mediated injury, we have developed an immunoisolating hydrogel-based capsule that [...] Read more.
Transplantation of allogeneic donor ovarian tissue holds great potential for female cancer survivors who often experience premature ovarian insufficiency. To avoid complications associated with immune suppression and to protect transplanted ovarian allografts from immune-mediated injury, we have developed an immunoisolating hydrogel-based capsule that supports the function of ovarian allografts without triggering an immune response. Encapsulated ovarian allografts implanted in naïve ovariectomized BALB/c mice responded to the circulating gonadotropins and maintained function for 4 months, as evident by regular estrous cycles and the presence of antral follicles in the retrieved grafts. In contrast to non-encapsulated controls, repeated implantations of encapsulated mouse ovarian allografts did not sensitize naïve BALB/c mice, which was confirmed with undetectable levels of alloantibodies. Further, encapsulated allografts implanted in hosts previously sensitized by the implantation of non-encapsulated allografts restored estrous cycles similarly to our results in naïve recipients. Next, we tested the translational potential and efficiency of the immune-isolating capsule in a rhesus monkey model by implanting encapsulated ovarian auto- and allografts in young ovariectomized animals. The encapsulated ovarian grafts survived and restored basal levels of urinary estrone conjugate and pregnanediol 3-glucuronide during the 4- and 5-month observation periods. We demonstrate, for the first time, that encapsulated ovarian allografts functioned for months in young rhesus monkeys and sensitized mice, while the immunoisolating capsule prevented sensitization and protected the allograft from rejection. Full article
(This article belongs to the Special Issue Bioengineering Technologies to Advance Reproductive Health)
Show Figures

Figure 1

21 pages, 2443 KiB  
Article
Proteomic Profiling of Fallopian Tube-Derived Extracellular Vesicles Using a Microfluidic Tissue-on-Chip System
by Didi Zha, Sagar Rayamajhi, Jared Sipes, Angela Russo, Harsh B. Pathak, Kailiang Li, Mihaela E. Sardiu, Leonidas E. Bantis, Amrita Mitra, Rajni V. Puri, Camille V. Trinidad, Brian P. Cain, Brett C. Isenberg, Jonathan Coppeta, Shannon MacLaughlan, Andrew K. Godwin and Joanna E. Burdette
Bioengineering 2023, 10(4), 423; https://doi.org/10.3390/bioengineering10040423 - 27 Mar 2023
Cited by 4 | Viewed by 4352
Abstract
The human fallopian tube epithelium (hFTE) is the site of fertilization, early embryo development, and the origin of most high-grade serous ovarian cancers (HGSOCs). Little is known about the content and functions of hFTE-derived small extracellular vesicles (sEVs) due to the limitations of [...] Read more.
The human fallopian tube epithelium (hFTE) is the site of fertilization, early embryo development, and the origin of most high-grade serous ovarian cancers (HGSOCs). Little is known about the content and functions of hFTE-derived small extracellular vesicles (sEVs) due to the limitations of biomaterials and proper culture methods. We have established a microfluidic platform to culture hFTE for EV collection with adequate yield for mass spectrometry-based proteomic profiling, and reported 295 common hFTE sEV proteins for the first time. These proteins are associated with exocytosis, neutrophil degranulation, and wound healing, and some are crucial for fertilization processes. In addition, by correlating sEV protein profiles with hFTE tissue transcripts characterized using GeoMx® Cancer Transcriptome Atlas, spatial transcriptomics analysis revealed cell-type-specific transcripts of hFTE that encode sEVs proteins, among which, FLNA, TUBB, JUP, and FLNC were differentially expressed in secretory cells, the precursor cells for HGSOC. Our study provides insights into the establishment of the baseline proteomic profile of sEVs derived from hFTE tissue, and its correlation with hFTE lineage-specific transcripts, which can be used to evaluate whether the fallopian tube shifts its sEV cargo during ovarian cancer carcinogenesis and the role of sEV proteins in fallopian tube reproductive functions. Full article
(This article belongs to the Special Issue Bioengineering Technologies to Advance Reproductive Health)
Show Figures

Graphical abstract

12 pages, 2445 KiB  
Article
Placenta-Targeted Nanoparticles Loaded with PFKFB3 Overexpression Plasmids Enhance Angiogenesis and Placental Function
by Qi Li, Xiaoxia Liu, Weifang Liu, Yang Zhang, Wen Liu, Mengying Wu, Zhirui Chen, Yin Zhao and Li Zou
Bioengineering 2022, 9(11), 652; https://doi.org/10.3390/bioengineering9110652 - 4 Nov 2022
Cited by 3 | Viewed by 1515
Abstract
Placental angiogenesis disorder and placental dysplasia are important causes of many pregnancy complications. Due to safety and economic benefits, effective treatment strategies are currently limited. PFKFB3 is a key regulator of glycolysis that controls angiogenesis through a metabolic pathway independent of genetic signals. [...] Read more.
Placental angiogenesis disorder and placental dysplasia are important causes of many pregnancy complications. Due to safety and economic benefits, effective treatment strategies are currently limited. PFKFB3 is a key regulator of glycolysis that controls angiogenesis through a metabolic pathway independent of genetic signals. In this study, we constructed the nanodrug T-NPPFKFB3 and explored its feasibility to promote angiogenesis and enhance placental function. First, liposomes containing PFKFB3 overexpression plasmids modified by the placental homing peptide CGKRK were synthesized by the thin film method. In vivo experiments revealed that T-NPPFKFB3 injected intravenously specifically accumulated in the mouse placenta and therein upregulated the expression of PFKFB3 without affecting its expression in other important organs. In addition, T-NPPFKFB3 promoted placental angiogenesis and increased the fetal and placental weights of the mice. Finally, we evaluated the safety of T-NPPFKFB3. The expression levels of ALS/AST/BUN in the sera of pregnant mice were not significantly different from those in the sera of control group mice. However, T-NPPFKFB3 did not cause obvious fetal abnormalities or alter the average litter size. In conclusion, T-NPPFKFB3 can specifically target the placenta, promote angiogenesis, and enhance placental function without obvious side effects. Therefore, it has potential as a new strategy for the treatment of pregnancy complications. Full article
(This article belongs to the Special Issue Bioengineering Technologies to Advance Reproductive Health)
Show Figures

Graphical abstract

Review

Jump to: Research

15 pages, 1647 KiB  
Review
Bioengineered 3D Ovarian Models as Paramount Technology for Female Health Management and Reproduction
by Julieta S. Del Valle and Susana M. Chuva de Sousa Lopes
Bioengineering 2023, 10(7), 832; https://doi.org/10.3390/bioengineering10070832 - 13 Jul 2023
Cited by 2 | Viewed by 2115
Abstract
Ovarian dysfunction poses significant threats to the health of female individuals. Ovarian failure can lead to infertility due to the lack or inefficient production of fertilizable eggs. In addition, the ovary produces hormones, such as estrogen and progesterone, that play crucial roles not [...] Read more.
Ovarian dysfunction poses significant threats to the health of female individuals. Ovarian failure can lead to infertility due to the lack or inefficient production of fertilizable eggs. In addition, the ovary produces hormones, such as estrogen and progesterone, that play crucial roles not only during pregnancy, but also in maintaining cardiovascular, bone, and cognitive health. Decline in estrogen and progesterone production due to ovarian dysfunction can result in menopausal-associated syndromes and lead to conditions, such as osteoporosis, cardiovascular disease, and Alzheimer’s disease. Recent advances in the design of bioengineered three-dimensional (3D) ovarian models, such as ovarian organoids or artificial ovaries, have made it possible to mimic aspects of the cellular heterogeneity and functional characteristics of the ovary in vitro. These novel technologies are emerging as valuable tools for studying ovarian physiology and pathology and may provide alternatives for fertility preservation. Moreover, they may have the potential to restore aspects of ovarian function, improving the quality of life of the (aging) female population. This review focuses on the state of the art of 3D ovarian platforms, including the latest advances modeling female reproduction, female physiology, ovarian cancer, and drug screening. Full article
(This article belongs to the Special Issue Bioengineering Technologies to Advance Reproductive Health)
Show Figures

Figure 1

Back to TopTop