1
|
Li Y, Zhang H, Li Q, Deng Y, Ye Z, Gui L. Texture-structure-based liquid metal filling for blind-end microchannels and its application on multi-layer chips. RSC Adv 2023; 13:24228-24236. [PMID: 37583671 PMCID: PMC10424060 DOI: 10.1039/d3ra04497a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/04/2023] [Indexed: 08/17/2023] Open
Abstract
This research work reports a novel method to achieve fast liquid metal (LM) injection in blind-end microchannels which is especially suitable for multi-layer microfluidic chips. This method is based on a texture-like surface bonding technology. The texture-like surface is fabricated on a polydimethylsiloxane (PDMS) slab with standard soft-lithography technology and bonded with another PDMS slab with microelectrode patterns on it. When injected with LM, the texture-like structure can prevent the LM from entering but allows the air inside to be released during the injection to achieve perfect blind-end complex LM electrodes. The experimental results show that it can achieve fast and perfect LM injection in the blind-end pattern and can also prevent the large area of the flat chamber from collapsing during bonding. We also parametrically studied the texture structure's size for bonding strength between the texture structure and the blank PDMS surface. In addition, we integrate three layers of blind-end complex liquid metal patterns into one multi-layer chip using this technology and later use this structure to realize series connection of two LM-based electroosmotic micropumps (EOP). Compared with the conventional LM-based EOP, the structure of the EOP chip was greatly simplified and resulted in a higher level of integration.
Collapse
Affiliation(s)
- Yuqing Li
- Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences 29 Zhongguancun East Road, Haidian District Beijing 100190 China
- School of Future Technology, University of Chinese Academy of Sciences Beijing 10039 China
| | - Huimin Zhang
- Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences 29 Zhongguancun East Road, Haidian District Beijing 100190 China
- School of Engineering Science, University of Chinese Academy of Sciences Beijing 10039 China
| | - Qian Li
- Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences 29 Zhongguancun East Road, Haidian District Beijing 100190 China
- School of Engineering Science, University of Chinese Academy of Sciences Beijing 10039 China
| | - Yuqin Deng
- Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences 29 Zhongguancun East Road, Haidian District Beijing 100190 China
- School of Future Technology, University of Chinese Academy of Sciences Beijing 10039 China
| | - Zi Ye
- Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences 29 Zhongguancun East Road, Haidian District Beijing 100190 China
| | - Lin Gui
- Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences 29 Zhongguancun East Road, Haidian District Beijing 100190 China
- School of Future Technology, University of Chinese Academy of Sciences Beijing 10039 China
| |
Collapse
|
2
|
Elias KM, Ng NW, Dam KU, Milne A, Disler ER, Gockley A, Holub N, Seshan ML, Church GM, Ginsburg ES, Anchan RM. Fertility restoration in mice with chemotherapy induced ovarian failure using differentiated iPSCs. EBioMedicine 2023; 94:104715. [PMID: 37482511 PMCID: PMC10435842 DOI: 10.1016/j.ebiom.2023.104715] [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: 11/02/2022] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Treatment options for premature ovarian insufficiency (POI) are limited to hormone replacement and donor oocytes. A novel induced pluripotent stem cell (iPSC) transplant paradigm in a mouse model has potential translational applications for management of POI. METHODS Mouse ovarian granulosa cell derived-iPSCS were labelled with green fluorescent protein (GFP) reporter and differentiated in vitro into oocytes. Differentiated cells were assayed for estradiol and progesterone secretion by enzyme-linked immunosorbent assays. After Fluorescence-Activated Cell Sorting (FACS) for the cell surface marker anti-Mullerian hormone receptor (AMHR2), enriched populations of differentiated cells were surgically transplanted into ovaries of mice that had POI secondary to gonadotoxic pre-treatment with alkylating agents. A total of 100 mice were used in these studies in five separate experiments with 56 animals receiving orthotopic ovarian injections of either FACS sorted or unsorted differentiated iPSCSs and the remaining animals receiving sham injections of PBS diluent. Following transplantation surgery, mice were stimulated with gonadotropins inducing oocyte development and underwent oocyte retrieval. Nine transplanted mice were cross bred with wild-type mice to assess fertility. Lineage tracing of resultant oocytes, F1 (30 pups), and F2 (42 pups) litters was interrogated by GFP expression and validation by short tandem repeat (STR) lineage tracing. FINDINGS [1] iPSCs differentiate into functional oocytes and steroidogenic ovarian cells which [2] express an ovarian (GJA1) and germ cell (ZP1) markers. [3] Endocrine function and fertility were restored in mice pretreated with gonadotoxic alkylating agents via orthotopic transplantation of differentiated iPSCS, thus generating viable, fertile mouse pups. INTERPRETATION iPSC-derived ovarian tissue can reverse endocrine and reproductive sequelae of POI. FUNDING Center for Infertility and Reproductive Surgery Research Award, Siezen Foundation award (RMA). Reproductive Scientist Development Program, Marriott Foundation, Saltonstall Foundation, Brigham Ovarian Cancer Research Fund (K.E).
Collapse
Affiliation(s)
- Kevin M Elias
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Nicholas W Ng
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Kh U Dam
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Ankrish Milne
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Emily R Disler
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Alison Gockley
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Nicole Holub
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Maya L Seshan
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - George M Church
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Elizabeth S Ginsburg
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Raymond M Anchan
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA.
| |
Collapse
|
3
|
Yang S, Chen Z, Cheng Y, Liu T, Pu Y, Liang G. Environmental toxicology wars: Organ-on-a-chip for assessing the toxicity of environmental pollutants. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 268:115861. [PMID: 33120150 DOI: 10.1016/j.envpol.2020.115861] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 05/07/2023]
Abstract
Environmental pollution is a widespread problem, which has seriously threatened human health and led to an increase of human diseases. Therefore, it is critical to evaluate environmental pollutants quickly and efficiently. Because of obvious inter-species differences between animals and humans, and lack of physiologically-relevant microenvironment, animal models and in vitro two-dimensional (2D) models can not accurately describe toxicological effects and predicting actual in vivo responses. To make up the limitations of conventional environmental toxicology screening, organ-on-a-chip (OOC) systems are increasingly developing. OOC systems can provide a well-organized architecture with comparable to the complex microenvironment in vivo and generate realistic responses to environmental pollutants. The feasibility, adjustability and reliability of OCC systems make it possible to offer new opportunities for environmental pollutants screening, which can study their metabolism, collective response, and fate in vivo. Further progress can address the challenges to make OCC systems better investigate and evaluate environmental pollutants with high predictive power.
Collapse
Affiliation(s)
- Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, PR China, 210096.
| | - Yanping Cheng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| | - Tong Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| |
Collapse
|
4
|
Fogg KC, Miller AE, Li Y, Flanigan W, Walker A, O'Shea A, Kendziorski C, Kreeger PK. Ovarian cancer cells direct monocyte differentiation through a non-canonical pathway. BMC Cancer 2020; 20:1008. [PMID: 33069212 PMCID: PMC7568422 DOI: 10.1186/s12885-020-07513-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 10/08/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Alternatively-activated macrophages (AAMs), an anti-inflammatory macrophage subpopulation, have been implicated in the progression of high grade serous ovarian carcinoma (HGSOC). Increased levels of AAMs are correlated with poor HGSOC survival rates, and AAMs increase the attachment and spread of HGSOC cells in vitro. However, the mechanism by which monocytes in the HGSOC tumor microenvironment are differentiated and polarized to AAMs remains unknown. METHODS Using an in vitro co-culture device, we cultured naïve, primary human monocytes with a panel of five HGSOC cell lines over the course of 7 days. An empirical Bayesian statistical method, EBSeq, was used to couple RNA-seq with observed monocyte-derived cell phenotype to explore which HGSOC-derived soluble factors supported differentiation to CD68+ macrophages and subsequent polarization towards CD163+ AAMs. Pathways of interest were interrogated using small molecule inhibitors, neutralizing antibodies, and CRISPR knockout cell lines. RESULTS HGSOC cell lines displayed a wide range of abilities to generate AAMs from naïve monocytes. Much of this variation appeared to result from differential ability to generate CD68+ macrophages, as most CD68+ cells were also CD163+. Differences in tumor cell potential to generate macrophages was not due to a MCSF-dependent mechanism, nor variance in established pro-AAM factors. TGFα was implicated as a potential signaling molecule produced by tumor cells that could induce macrophage differentiation, which was validated using a CRISPR knockout of TGFA in the OVCAR5 cell line. CONCLUSIONS HGSOC production of TGFα drives monocytes to differentiate into macrophages, representing a central arm of the mechanism by which AAMs are generated in the tumor microenvironment.
Collapse
Affiliation(s)
- Kaitlin C Fogg
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Ave, WIMR 4553, Madison, WI, 53705, USA
| | - Andrew E Miller
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Ave, WIMR 4553, Madison, WI, 53705, USA
| | - Ying Li
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Will Flanigan
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Ave, WIMR 4553, Madison, WI, 53705, USA
| | - Alyssa Walker
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Ave, WIMR 4553, Madison, WI, 53705, USA
| | - Andrea O'Shea
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Pamela K Kreeger
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Ave, WIMR 4553, Madison, WI, 53705, USA.
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
5
|
Zeevaert K, Elsafi Mabrouk MH, Wagner W, Goetzke R. Cell Mechanics in Embryoid Bodies. Cells 2020; 9:E2270. [PMID: 33050550 PMCID: PMC7599659 DOI: 10.3390/cells9102270] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022] Open
Abstract
Embryoid bodies (EBs) resemble self-organizing aggregates of pluripotent stem cells that recapitulate some aspects of early embryogenesis. Within few days, the cells undergo a transition from rather homogeneous epithelial-like pluripotent stem cell colonies into a three-dimensional organization of various cell types with multifaceted cell-cell interactions and lumen formation-a process associated with repetitive epithelial-mesenchymal transitions. In the last few years, culture methods have further evolved to better control EB size, growth, cellular composition, and organization-e.g., by the addition of morphogens or different extracellular matrix molecules. There is a growing perception that the mechanical properties, cell mechanics, and cell signaling during EB development are also influenced by physical cues to better guide lineage specification; substrate elasticity and topography are relevant, as well as shear stress and mechanical strain. Epithelial structures outside and inside EBs support the integrity of the cell aggregates and counteract mechanical stress. Furthermore, hydrogels can be used to better control the organization and lineage-specific differentiation of EBs. In this review, we summarize how EB formation is accompanied by a variety of biomechanical parameters that need to be considered for the directed and reproducible self-organization of early cell fate decisions.
Collapse
Affiliation(s)
- Kira Zeevaert
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany; (K.Z.); (M.H.E.M.)
- Institute for Biomedical Engineering–Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Mohamed H. Elsafi Mabrouk
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany; (K.Z.); (M.H.E.M.)
- Institute for Biomedical Engineering–Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany; (K.Z.); (M.H.E.M.)
- Institute for Biomedical Engineering–Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Roman Goetzke
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany; (K.Z.); (M.H.E.M.)
- Institute for Biomedical Engineering–Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany
| |
Collapse
|
6
|
Kennedy R, Kuvshinov D, Sdrolia A, Kuvshinova E, Hilton K, Crank S, Beavis AW, Green V, Greenman J. A patient tumour-on-a-chip system for personalised investigation of radiotherapy based treatment regimens. Sci Rep 2019; 9:6327. [PMID: 31004114 PMCID: PMC6474873 DOI: 10.1038/s41598-019-42745-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/01/2019] [Indexed: 01/22/2023] Open
Abstract
Development of personalised cancer models to predict response to radiation would benefit patient care; particularly in malignancies where treatment resistance is prevalent. Herein, a robust, easy to use, tumour-on-a-chip platform which maintains precision cut head and neck cancer for the purpose of ex vivo irradiation is described. The device utilises sintered discs to separate the biopsy and medium, mimicking in vivo microvascular flow and diffusion, maintaining tissue viability for 68 h. Integrity of tissues is demonstrated by the low levels of lactate dehydrogenase release and retained histology, accompanied by assessment of cell viability by trypan blue exclusion and flow cytometry; fluid dynamic modelling validates culture conditions. An irradiation jig is described for reproducible delivery of clinically-relevant doses (5 × 2 Gy) to newly-presenting primary tumours (n = 12); the addition of concurrent cisplatin is also investigated (n = 8) with response analysed by immunohistochemistry. Fractionated irradiation reduced proliferation (BrdU, p = 0.0064), increased DNA damage (ƴH2AX, p = 0.0043) and caspase-dependent apoptosis (caspase-cleaved cytokeratin-18) compared to control; caspase-dependent apoptosis was further increased by concurrent cisplatin compared to control (p = 0.0063). This is a proof of principle study showing the response of cancer tissue to irradiation ex vivo in a bespoke system. The novel platform described has the potential to personalise treatment for patients in a cost-effective manner with applicability to any solid tumour.
Collapse
Affiliation(s)
- R Kennedy
- Department of Biomedical Sciences, The University of Hull, Cottingham Road, Hull, UK
| | - D Kuvshinov
- School of Engineering & Computer Science, The University of Hull, Cottingham Road, Hull, UK
| | - A Sdrolia
- Department of Medical Physics, Hull and East Yorkshire Hospitals NHS Trust, Cottingham, UK
| | - E Kuvshinova
- Department of Chemical & Biological Engineering, The University of Sheffield, Sheffield, UK
| | - K Hilton
- Department of Medical Physics, Hull and East Yorkshire Hospitals NHS Trust, Cottingham, UK
| | - S Crank
- Department of Maxillofacial Surgery, Hull and East Yorkshire Hospitals NHS Trust, Hull, UK
| | - A W Beavis
- Department of Biomedical Sciences, The University of Hull, Cottingham Road, Hull, UK
- Department of Medical Physics, Hull and East Yorkshire Hospitals NHS Trust, Cottingham, UK
- Faculty of Health and Well Being, Sheffield-Hallam University, Sheffield, UK
| | - V Green
- Department of Biomedical Sciences, The University of Hull, Cottingham Road, Hull, UK
| | - J Greenman
- Department of Biomedical Sciences, The University of Hull, Cottingham Road, Hull, UK.
| |
Collapse
|
7
|
Lam KH, Fernandez-Perez A, Schmidtke DW, Munshi NV. Functional cargo delivery into mouse and human fibroblasts using a versatile microfluidic device. Biomed Microdevices 2018; 20:52. [PMID: 29938310 DOI: 10.1007/s10544-018-0292-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Efficient intracellular cargo delivery is a key hurdle for the translation of many emerging stem cell and cellular reprogramming therapies. Recently, a microfluidic-based device constructed from silicon was shown to transduce macromolecules into cells via shear-induced formation of plasma membrane pores. However, the scalability and widespread application of the current platform is limited since physical deformation-mediated delivery must be optimized for each therapeutic application. Therefore, we sought to create a low-cost, versatile device that could facilitate rapid prototyping and application-specific optimization in most academic research labs. Here we describe the design and implementation of a microfluidic device constructed from Polydimethylsiloxane (PDMS) that we call Cyto-PDMS (Cytoplasmic PDMS-based Delivery and Modification System). Using a systematic Cyto-PDMS workflow, we demonstrate intracellular cargo delivery with minimal effects on cellular viability. We identify specific flow rates at which a wide range of cargo sizes (1-70 kDa) can be delivered to the cell interior. As a proof-of-principle for the biological utility of Cyto-PDMS, we show (i) F-actin labeling in live human fibroblasts and (ii) intracellular delivery of recombinant Cre protein with appropriate genomic recombination in recipient fibroblasts. Taken together, our results demonstrate that Cyto-PDMS can deliver small-molecules to the cytoplasm and biologically active cargo to the nucleus without major effects on viability. We anticipate that the cost and versatility of PDMS can be leveraged to optimize delivery to a broad array of possible cell types and thus expand the potential impact of cellular therapies.
Collapse
Affiliation(s)
- Kevin H Lam
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | | | - David W Schmidtke
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Nikhil V Munshi
- Department of Internal Medicine-Cardiology, UT Southwestern Medical Center, Dallas, TX, USA. .,Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA. .,McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA. .,Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
8
|
Lan D, Chen X, Li P, Zou W, Wu L, Chen W. Using a Novel Supramolecular Gel Cryopreservation System in Microchannel to Minimize the Cell Injury. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:5088-5096. [PMID: 29629777 DOI: 10.1021/acs.langmuir.8b00265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The storage of living cells is the major challenge for cell research and cell treatment. Here, we introduced a novel supramolecular gel cryopreservation system which was prepared in the microchannel, and the supramolecular gel (BDTC) was self-assembled by gelator Boc- O-dodecyl-l-tyrosine (BDT). This cryopreservation system could obviously minimize the cell injury because the BDTC supramolecular gel had a more compact three-dimensional network structure when the BDT gelator self-assembled in the confined space of microchannel. This compact structure could confine the growth of the ice crystal, reduce the change rate of cell volumes and osmotic shock, decrease the freezing point of the cryopreservation system, and possess better protection capability. Furthermore, the results of functionality assessments showed that the thawed cells could grow and proliferate well and remain the same growth trend of the fresh cells after the RSC96 cells flowed out from the microchannel. This novel method has potential to be used for the cryopreservation of cells, cell therapy, and tissue engineering.
Collapse
Affiliation(s)
- Dongxu Lan
- School of Materials Science and Engineering , Wuhan University of Technology , Wuhan , Hubei 430070 , China
| | - Xi Chen
- School of Materials Science and Engineering , Wuhan University of Technology , Wuhan , Hubei 430070 , China
| | - Pengcheng Li
- School of Materials Science and Engineering , Wuhan University of Technology , Wuhan , Hubei 430070 , China
| | - Wei Zou
- School of Materials Science and Engineering , Wuhan University of Technology , Wuhan , Hubei 430070 , China
| | - Lili Wu
- School of Materials Science and Engineering , Wuhan University of Technology , Wuhan , Hubei 430070 , China
| | - Wanyu Chen
- School of Materials Science and Engineering , Wuhan University of Technology , Wuhan , Hubei 430070 , China
| |
Collapse
|
9
|
Nagashima JB, El Assal R, Songsasen N, Demirci U. Evaluation of an ovary-on-a-chip in large mammalian models: Species specificity and influence of follicle isolation status. J Tissue Eng Regen Med 2018; 12:e1926-e1935. [PMID: 29222841 DOI: 10.1002/term.2623] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/09/2017] [Accepted: 11/27/2017] [Indexed: 12/20/2022]
Abstract
The ability to grow oocytes from immature ovarian follicles in vitro has significant potential for fertility preservation; yet, it has proved challenging in large mammalian species due to the complex metabolic needs and long-term culture requirements. Currently, follicular incubations are based on a "static" system with manual exchange of medium. Despite the numerous advantages of conventional culturing approaches, recapitulating the native microenvironment and supporting the survival of ovarian follicles from large mammalian species still represent challenges. In this study, we utilized an innovative, dynamic microfluidic system to support the in vitro survival of domestic cat and dog follicles enclosed within the ovarian cortex or isolated from ovarian cortex. Results indicate both species-specific and tissue type-specific differences in response to microfluidic culture. Domestic cat but not dog ovarian cortical tissues maintained viability under flow similar to conventional agarose gel controls. Preantral stage isolated follicles from both species that grew most favourably in conventional alginate bead culture, but overall, there was no influence of culture system on expression of follicle development or oocyte health markers. This system represents an important exploration toward the development of an improved ovarian in vitro culture system of large mammalian species (e.g., cats and dogs), which has potential applications for fertility preservation, reproductive toxicology, and endangered mammal conservation efforts.
Collapse
Affiliation(s)
- Jennifer B Nagashima
- Smithsonian Conservation Biology Institute, National Zoological Park, Front Royal, VA, USA.,Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Rami El Assal
- Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Nucharin Songsasen
- Smithsonian Conservation Biology Institute, National Zoological Park, Front Royal, VA, USA
| | - Utkan Demirci
- Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA.,Department of Electrical Engineering (by courtesy), Stanford University School of Engineering, Palo Alto, CA, USA
| |
Collapse
|
10
|
Lipskind S, Lindsey JS, Gerami-Naini B, Eaton JL, O'Connell D, Kiezun A, Ho JWK, Ng N, Parasar P, Ng M, Nickerson M, Demirci U, Maas R, Anchan RM. An Embryonic and Induced Pluripotent Stem Cell Model for Ovarian Granulosa Cell Development and Steroidogenesis. Reprod Sci 2017; 25:712-726. [PMID: 28854867 DOI: 10.1177/1933719117725814] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Embryoid bodies (EBs) can serve as a system for evaluating pluripotency, cellular differentiation, and tissue morphogenesis. In this study, we use EBs derived from mouse embryonic stem cells (mESCs) and human amniocyte-derived induced pluripotent stem cells (hAdiPSCs) as a model for ovarian granulosa cell (GC) development and steroidogenic cell commitment. We demonstrated that spontaneously differentiated murine EBs (mEBs) and human EBs (hEBs) displayed ovarian GC markers, such as aromatase (CYP19A1), FOXL2, AMHR2, FSHR, and GJA1. Comparative microarray analysis identified both shared and unique gene expression between mEBs and the maturing mouse ovary. Gene sets related to gonadogenesis, lipid metabolism, and ovarian development were significantly overrepresented in EBs. Of the 29 genes, 15 that were differentially regulated in steroidogenic mEBs displayed temporal expression changes between embryonic, postnatal, and mature ovarian tissues by polymerase chain reaction. Importantly, both mEBs and hEBs were capable of gonadotropin-responsive estradiol (E2) synthesis in vitro (217-759 pg/mL). Live fluorescence-activated cell sorting-sorted AMHR2+ granulosa-like cells from mEBs continued to produce E2 after purification (15.3 pg/mL) and secreted significantly more E2 than AMHR2- cells (8.6 pg/mL, P < .05). We conclude that spontaneously differentiated EBs of both mESC and hAdiPSC origin can serve as a biologically relevant model for ovarian GC differentiation and steroidogenic cell commitment. These cells should be further investigated for therapeutic uses, such as stem cell-based hormone replacement therapy and in vitro maturation of oocytes.
Collapse
Affiliation(s)
- Shane Lipskind
- 1 Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jennifer S Lindsey
- 1 Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Behzad Gerami-Naini
- 2 Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jennifer L Eaton
- 2 Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Daniel O'Connell
- 2 Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Adam Kiezun
- 3 Computational Methods Development, Cancer Genome Analysis, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joshua W K Ho
- 4 Victor Chang Cardiac Research Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Nicholas Ng
- 1 Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Parveen Parasar
- 1 Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michelle Ng
- 1 Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael Nickerson
- 1 Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Utkan Demirci
- 5 Canary Center at Stanford for Early Cancer Detection, Stanford School of Medicine, Palo Alto, CA, USA
| | - Richard Maas
- 2 Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,6 Affiliated Faculty, Harvard Stem Cell Institute, Cambridge, MA, USA. Gerami-Naini is now with the Department of Diagnostic Sciences, School of Dental Medicine, Tufts University, Boston MA, USA. Eaton is now with the Division of Reproductive Endocrinology and Fertility, Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA. O'Connell is now with the Intellia Therapeutics, Inc, Cambridge, MA, USA. Kiezun is now with the Amazon.com , Boston, MA, USA
| | - Raymond M Anchan
- 1 Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,6 Affiliated Faculty, Harvard Stem Cell Institute, Cambridge, MA, USA. Gerami-Naini is now with the Department of Diagnostic Sciences, School of Dental Medicine, Tufts University, Boston MA, USA. Eaton is now with the Division of Reproductive Endocrinology and Fertility, Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA. O'Connell is now with the Intellia Therapeutics, Inc, Cambridge, MA, USA. Kiezun is now with the Amazon.com , Boston, MA, USA
| |
Collapse
|
11
|
Arslan-Yildiz A, Assal RE, Chen P, Guven S, Inci F, Demirci U. Towards artificial tissue models: past, present, and future of 3D bioprinting. Biofabrication 2016; 8:014103. [DOI: 10.1088/1758-5090/8/1/014103] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
12
|
Asghar W, El Assal R, Shafiee H, Pitteri S, Paulmurugan R, Demirci U. Engineering cancer microenvironments for in vitro 3-D tumor models. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2015; 18:539-553. [PMID: 28458612 PMCID: PMC5407188 DOI: 10.1016/j.mattod.2015.05.002] [Citation(s) in RCA: 229] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
The natural microenvironment of tumors is composed of extracellular matrix (ECM), blood vasculature, and supporting stromal cells. The physical characteristics of ECM as well as the cellular components play a vital role in controlling cancer cell proliferation, apoptosis, metabolism, and differentiation. To mimic the tumor microenvironment outside the human body for drug testing, two-dimensional (2-D) and murine tumor models are routinely used. Although these conventional approaches are employed in preclinical studies, they still present challenges. For example, murine tumor models are expensive and difficult to adopt for routine drug screening. On the other hand, 2-D in vitro models are simple to perform, but they do not recapitulate natural tumor microenvironment, because they do not capture important three-dimensional (3-D) cell-cell, cell-matrix signaling pathways, and multi-cellular heterogeneous components of the tumor microenvironment such as stromal and immune cells. The three-dimensional (3-D) in vitro tumor models aim to closely mimic cancer microenvironments and have emerged as an alternative to routinely used methods for drug screening. Herein, we review recent advances in 3-D tumor model generation and highlight directions for future applications in drug testing.
Collapse
Affiliation(s)
- Waseem Asghar
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Department of Computer Engineering & Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Rami El Assal
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Hadi Shafiee
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Division of Biomedical Engineering, Division of Infectious Diseases, Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Sharon Pitteri
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Ramasamy Paulmurugan
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Utkan Demirci
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Division of Biomedical Engineering, Division of Infectious Diseases, Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| |
Collapse
|
13
|
Guven S, Chen P, Inci F, Tasoglu S, Erkmen B, Demirci U. Multiscale assembly for tissue engineering and regenerative medicine. Trends Biotechnol 2015; 33:269-279. [PMID: 25796488 DOI: 10.1016/j.tibtech.2015.02.003] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/30/2015] [Accepted: 02/03/2015] [Indexed: 01/11/2023]
Abstract
Our understanding of cell biology and its integration with materials science has led to technological innovations in the bioengineering of tissue-mimicking grafts that can be utilized in clinical and pharmaceutical applications. Bioengineering of native-like multiscale building blocks provides refined control over the cellular microenvironment, thus enabling functional tissues. In this review, we focus on assembling building blocks from the biomolecular level to the millimeter scale. We also provide an overview of techniques for assembling molecules, cells, spheroids, and microgels and achieving bottom-up tissue engineering. Additionally, we discuss driving mechanisms for self- and guided assembly to create micro-to-macro scale tissue structures.
Collapse
Affiliation(s)
- Sinan Guven
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, USA
| | - Pu Chen
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, USA
| | - Fatih Inci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, USA
| | - Savas Tasoglu
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, USA
| | - Burcu Erkmen
- BAMM Laboratory, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, USA
- BAMM Laboratory, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|