Hydrogel-Integrated Millifluidic Systems: Advancing the Fabrication of Mucus-Producing Human Intestinal Models

Publikation: Beitrag in FachzeitschriftArtikelForschungPeer-Review

Autoren

  • Ahed Almalla
  • Nadra Alzain
  • Laura Elomaa
  • Fiona Richter
  • Johanna Scholz
  • Marcus Lindner
  • Britta Siegmund
  • Marie Weinhart

Externe Organisationen

  • Freie Universität Berlin (FU Berlin)
  • Charité - Universitätsmedizin Berlin
Forschungs-netzwerk anzeigen

Details

OriginalspracheEnglisch
Aufsatznummer1080
FachzeitschriftCells
Jahrgang13
Ausgabenummer13
PublikationsstatusVeröffentlicht - 21 Juli 2024

Abstract

The luminal surface of the intestinal epithelium is protected by a vital mucus layer, which is essential for lubrication, hydration, and fostering symbiotic bacterial relationships. Replicating and studying this complex mucus structure in vitro presents considerable challenges. To address this, we developed a hydrogel-integrated millifluidic tissue chamber capable of applying precise apical shear stress to intestinal models cultured on flat or 3D structured hydrogel scaffolds with adjustable stiffness. The chamber is designed to accommodate nine hydrogel scaffolds, 3D-printed as flat disks with a storage modulus matching the physiological range of intestinal tissue stiffness (~3.7 kPa) from bioactive decellularized and methacrylated small intestinal submucosa (dSIS-MA). Computational fluid dynamics simulations were conducted to confirm a laminar flow profile for both flat and 3D villi-comprising scaffolds in the physiologically relevant regime. The system was initially validated with HT29-MTX seeded hydrogel scaffolds, demonstrating accelerated differentiation, increased mucus production, and enhanced 3D organization under shear stress. These characteristic intestinal tissue features are essential for advanced in vitro models as they critically contribute to a functional barrier. Subsequently, the chamber was challenged with human intestinal stem cells (ISCs) from the terminal ileum. Our findings indicate that biomimicking hydrogel scaffolds, in combination with physiological shear stress, promote multi-lineage differentiation, as evidenced by a gene and protein expression analysis of basic markers and the 3D structural organization of ISCs in the absence of chemical differentiation triggers. The quantitative analysis of the alkaline phosphatase (ALP) activity and secreted mucus demonstrates the functional differentiation of the cells into enterocyte and goblet cell lineages. The millifluidic system, which has been developed and optimized for performance and cost efficiency, enables the creation and modulation of advanced intestinal models under biomimicking conditions, including tunable matrix stiffness and varying fluid shear stresses. Moreover, the readily accessible and scalable mucus-producing cellular tissue models permit comprehensive mucus analysis and the investigation of pathogen interactions and penetration, thereby offering the potential to advance our understanding of intestinal mucus in health and disease.

Zitieren

Hydrogel-Integrated Millifluidic Systems: Advancing the Fabrication of Mucus-Producing Human Intestinal Models. / Almalla, Ahed; Alzain, Nadra; Elomaa, Laura et al.
in: Cells, Jahrgang 13, Nr. 13, 1080, 21.07.2024.

Publikation: Beitrag in FachzeitschriftArtikelForschungPeer-Review

Almalla, A, Alzain, N, Elomaa, L, Richter, F, Scholz, J, Lindner, M, Siegmund, B & Weinhart, M 2024, 'Hydrogel-Integrated Millifluidic Systems: Advancing the Fabrication of Mucus-Producing Human Intestinal Models', Cells, Jg. 13, Nr. 13, 1080. https://doi.org/10.3390/cells13131080
Almalla, A., Alzain, N., Elomaa, L., Richter, F., Scholz, J., Lindner, M., Siegmund, B., & Weinhart, M. (2024). Hydrogel-Integrated Millifluidic Systems: Advancing the Fabrication of Mucus-Producing Human Intestinal Models. Cells, 13(13), Artikel 1080. https://doi.org/10.3390/cells13131080
Almalla A, Alzain N, Elomaa L, Richter F, Scholz J, Lindner M et al. Hydrogel-Integrated Millifluidic Systems: Advancing the Fabrication of Mucus-Producing Human Intestinal Models. Cells. 2024 Jul 21;13(13):1080. doi: 10.3390/cells13131080
Almalla, Ahed ; Alzain, Nadra ; Elomaa, Laura et al. / Hydrogel-Integrated Millifluidic Systems : Advancing the Fabrication of Mucus-Producing Human Intestinal Models. in: Cells. 2024 ; Jahrgang 13, Nr. 13.
Download
@article{cbee17c8f0e942c9a1b2610c7914b5c3,
title = "Hydrogel-Integrated Millifluidic Systems: Advancing the Fabrication of Mucus-Producing Human Intestinal Models",
abstract = "The luminal surface of the intestinal epithelium is protected by a vital mucus layer, which is essential for lubrication, hydration, and fostering symbiotic bacterial relationships. Replicating and studying this complex mucus structure in vitro presents considerable challenges. To address this, we developed a hydrogel-integrated millifluidic tissue chamber capable of applying precise apical shear stress to intestinal models cultured on flat or 3D structured hydrogel scaffolds with adjustable stiffness. The chamber is designed to accommodate nine hydrogel scaffolds, 3D-printed as flat disks with a storage modulus matching the physiological range of intestinal tissue stiffness (~3.7 kPa) from bioactive decellularized and methacrylated small intestinal submucosa (dSIS-MA). Computational fluid dynamics simulations were conducted to confirm a laminar flow profile for both flat and 3D villi-comprising scaffolds in the physiologically relevant regime. The system was initially validated with HT29-MTX seeded hydrogel scaffolds, demonstrating accelerated differentiation, increased mucus production, and enhanced 3D organization under shear stress. These characteristic intestinal tissue features are essential for advanced in vitro models as they critically contribute to a functional barrier. Subsequently, the chamber was challenged with human intestinal stem cells (ISCs) from the terminal ileum. Our findings indicate that biomimicking hydrogel scaffolds, in combination with physiological shear stress, promote multi-lineage differentiation, as evidenced by a gene and protein expression analysis of basic markers and the 3D structural organization of ISCs in the absence of chemical differentiation triggers. The quantitative analysis of the alkaline phosphatase (ALP) activity and secreted mucus demonstrates the functional differentiation of the cells into enterocyte and goblet cell lineages. The millifluidic system, which has been developed and optimized for performance and cost efficiency, enables the creation and modulation of advanced intestinal models under biomimicking conditions, including tunable matrix stiffness and varying fluid shear stresses. Moreover, the readily accessible and scalable mucus-producing cellular tissue models permit comprehensive mucus analysis and the investigation of pathogen interactions and penetration, thereby offering the potential to advance our understanding of intestinal mucus in health and disease.",
keywords = "3D printing, cell-based mucus model, CFD, dynamic cell culture, extracellular matrix, human terminal ileum, intestinal organoid, physiological shear stress, vat photopolymerization",
author = "Ahed Almalla and Nadra Alzain and Laura Elomaa and Fiona Richter and Johanna Scholz and Marcus Lindner and Britta Siegmund and Marie Weinhart",
note = "Publisher Copyright: {\textcopyright} 2024 by the authors.",
year = "2024",
month = jul,
day = "21",
doi = "10.3390/cells13131080",
language = "English",
volume = "13",
number = "13",

}

Download

TY - JOUR

T1 - Hydrogel-Integrated Millifluidic Systems

T2 - Advancing the Fabrication of Mucus-Producing Human Intestinal Models

AU - Almalla, Ahed

AU - Alzain, Nadra

AU - Elomaa, Laura

AU - Richter, Fiona

AU - Scholz, Johanna

AU - Lindner, Marcus

AU - Siegmund, Britta

AU - Weinhart, Marie

N1 - Publisher Copyright: © 2024 by the authors.

PY - 2024/7/21

Y1 - 2024/7/21

N2 - The luminal surface of the intestinal epithelium is protected by a vital mucus layer, which is essential for lubrication, hydration, and fostering symbiotic bacterial relationships. Replicating and studying this complex mucus structure in vitro presents considerable challenges. To address this, we developed a hydrogel-integrated millifluidic tissue chamber capable of applying precise apical shear stress to intestinal models cultured on flat or 3D structured hydrogel scaffolds with adjustable stiffness. The chamber is designed to accommodate nine hydrogel scaffolds, 3D-printed as flat disks with a storage modulus matching the physiological range of intestinal tissue stiffness (~3.7 kPa) from bioactive decellularized and methacrylated small intestinal submucosa (dSIS-MA). Computational fluid dynamics simulations were conducted to confirm a laminar flow profile for both flat and 3D villi-comprising scaffolds in the physiologically relevant regime. The system was initially validated with HT29-MTX seeded hydrogel scaffolds, demonstrating accelerated differentiation, increased mucus production, and enhanced 3D organization under shear stress. These characteristic intestinal tissue features are essential for advanced in vitro models as they critically contribute to a functional barrier. Subsequently, the chamber was challenged with human intestinal stem cells (ISCs) from the terminal ileum. Our findings indicate that biomimicking hydrogel scaffolds, in combination with physiological shear stress, promote multi-lineage differentiation, as evidenced by a gene and protein expression analysis of basic markers and the 3D structural organization of ISCs in the absence of chemical differentiation triggers. The quantitative analysis of the alkaline phosphatase (ALP) activity and secreted mucus demonstrates the functional differentiation of the cells into enterocyte and goblet cell lineages. The millifluidic system, which has been developed and optimized for performance and cost efficiency, enables the creation and modulation of advanced intestinal models under biomimicking conditions, including tunable matrix stiffness and varying fluid shear stresses. Moreover, the readily accessible and scalable mucus-producing cellular tissue models permit comprehensive mucus analysis and the investigation of pathogen interactions and penetration, thereby offering the potential to advance our understanding of intestinal mucus in health and disease.

AB - The luminal surface of the intestinal epithelium is protected by a vital mucus layer, which is essential for lubrication, hydration, and fostering symbiotic bacterial relationships. Replicating and studying this complex mucus structure in vitro presents considerable challenges. To address this, we developed a hydrogel-integrated millifluidic tissue chamber capable of applying precise apical shear stress to intestinal models cultured on flat or 3D structured hydrogel scaffolds with adjustable stiffness. The chamber is designed to accommodate nine hydrogel scaffolds, 3D-printed as flat disks with a storage modulus matching the physiological range of intestinal tissue stiffness (~3.7 kPa) from bioactive decellularized and methacrylated small intestinal submucosa (dSIS-MA). Computational fluid dynamics simulations were conducted to confirm a laminar flow profile for both flat and 3D villi-comprising scaffolds in the physiologically relevant regime. The system was initially validated with HT29-MTX seeded hydrogel scaffolds, demonstrating accelerated differentiation, increased mucus production, and enhanced 3D organization under shear stress. These characteristic intestinal tissue features are essential for advanced in vitro models as they critically contribute to a functional barrier. Subsequently, the chamber was challenged with human intestinal stem cells (ISCs) from the terminal ileum. Our findings indicate that biomimicking hydrogel scaffolds, in combination with physiological shear stress, promote multi-lineage differentiation, as evidenced by a gene and protein expression analysis of basic markers and the 3D structural organization of ISCs in the absence of chemical differentiation triggers. The quantitative analysis of the alkaline phosphatase (ALP) activity and secreted mucus demonstrates the functional differentiation of the cells into enterocyte and goblet cell lineages. The millifluidic system, which has been developed and optimized for performance and cost efficiency, enables the creation and modulation of advanced intestinal models under biomimicking conditions, including tunable matrix stiffness and varying fluid shear stresses. Moreover, the readily accessible and scalable mucus-producing cellular tissue models permit comprehensive mucus analysis and the investigation of pathogen interactions and penetration, thereby offering the potential to advance our understanding of intestinal mucus in health and disease.

KW - 3D printing

KW - cell-based mucus model

KW - CFD

KW - dynamic cell culture

KW - extracellular matrix

KW - human terminal ileum

KW - intestinal organoid

KW - physiological shear stress

KW - vat photopolymerization

UR - http://www.scopus.com/inward/record.url?scp=85198357752&partnerID=8YFLogxK

U2 - 10.3390/cells13131080

DO - 10.3390/cells13131080

M3 - Article

C2 - 38994934

AN - SCOPUS:85198357752

VL - 13

JO - Cells

JF - Cells

SN - 2073-4409

IS - 13

M1 - 1080

ER -

Von denselben Autoren