1
|
Briones-Orta MA, Delgado-Coello B, Gutiérrez-Vidal R, Sosa-Garrocho M, Macías-Silva M, Mas-Oliva J. Quantitative Expression of Key Cancer Markers in the AS-30D Hepatocarcinoma Model. Front Oncol 2021; 11:670292. [PMID: 34737944 PMCID: PMC8561839 DOI: 10.3389/fonc.2021.670292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
Hepatocellular carcinoma is one of the cancers with the highest mortality rate worldwide. HCC is often diagnosed when the disease is already in an advanced stage, making the discovery and implementation of biomarkers for the disease a critical aim in cancer research. In this study, we aim to quantify the transcript levels of key signaling molecules relevant to different pathways known to participate in tumorigenesis, with special emphasis on those related to cancer hallmarks and epithelial-mesenchymal transition, using as a model the murine transplantable hepatocarcinoma AS-30D. Using qPCR to quantify the mRNA levels of genes involved in tumorigenesis, we found elevated levels for Tgfb1 and Spp1, two master regulators of EMT. A mesenchymal signature profile for AS-30D cells is also supported by the overexpression of genes encoding for molecules known to be associated to aggressiveness and metastatic phenotypes such as Foxm1, C-met, and Inppl1. This study supports the use of the AS-30D cells as an efficient and cost-effective model to study gene expression changes in HCC, especially those associated with the EMT process.
Collapse
Affiliation(s)
- Marco A Briones-Orta
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Blanca Delgado-Coello
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Roxana Gutiérrez-Vidal
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marcela Sosa-Garrocho
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marina Macías-Silva
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jaime Mas-Oliva
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
2
|
SKI activates the Hippo pathway via LIMD1 to inhibit cardiac fibroblast activation. Basic Res Cardiol 2021; 116:25. [PMID: 33847835 PMCID: PMC8043893 DOI: 10.1007/s00395-021-00865-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 03/24/2021] [Indexed: 01/14/2023]
Abstract
We have previously shown that overexpression of SKI, an endogenous TGF-β1 repressor, deactivates the pro-fibrotic myofibroblast phenotype in the heart. We now show that SKI also functions independently of SMAD/TGF-β signaling, by activating the Hippo tumor-suppressor pathway and inhibiting the Transcriptional co-Activator with PDZ-binding motif (TAZ or WWTR1). The mechanism(s) by which SKI targets TAZ to inhibit cardiac fibroblast activation and fibrogenesis remain undefined. A rat model of post-myocardial infarction was used to examine the expression of TAZ during acute fibrogenesis and chronic heart failure. Results were then corroborated with primary rat cardiac fibroblast cell culture performed both on plastic and on inert elastic substrates, along with the use of siRNA and adenoviral expression vectors for active forms of SKI, YAP, and TAZ. Gene expression was examined by qPCR and luciferase assays, while protein expression was examined by immunoblotting and fluorescence microscopy. Cell phenotype was further assessed by functional assays. Finally, to elucidate SKI’s effects on Hippo signaling, the SKI and TAZ interactomes were captured in human cardiac fibroblasts using BioID2 and mass spectrometry. Potential interactors were investigated in vitro to reveal novel mechanisms of action for SKI. In vitro assays on elastic substrates revealed the ability of TAZ to overcome environmental stimuli and induce the activation of hypersynthetic cardiac myofibroblasts. Further cell-based assays demonstrated that SKI causes specific proteasomal degradation of TAZ, but not YAP, and shifts actin cytoskeleton dynamics to inhibit myofibroblast activation. These findings were supported by identifying the bi-phasic expression of TAZ in vivo during post-MI remodeling and fibrosis. BioID2-based interactomics in human cardiac fibroblasts suggest that SKI interacts with actin-modifying proteins and with LIM Domain-containing protein 1 (LIMD1), a negative regulator of Hippo signaling. Furthermore, we found that LATS2 interacts with TAZ, whereas LATS1 does not, and that LATS2 knockdown prevented TAZ downregulation with SKI overexpression. Our findings indicate that SKI’s capacity to regulate cardiac fibroblast activation is mediated, in part, by Hippo signaling. We postulate that the interaction between SKI and TAZ in cardiac fibroblasts is arbitrated by LIMD1, an important intermediary in focal adhesion-associated signaling pathways. This study contributes to the understanding of the unique physiology of cardiac fibroblasts, and of the relationship between SKI expression and cell phenotype.
Collapse
|
3
|
Melchionna R, Trono P, Tocci A, Nisticò P. Actin Cytoskeleton and Regulation of TGFβ Signaling: Exploring Their Links. Biomolecules 2021; 11:biom11020336. [PMID: 33672325 PMCID: PMC7926735 DOI: 10.3390/biom11020336] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 12/14/2022] Open
Abstract
Human tissues, to maintain their architecture and function, respond to injuries by activating intricate biochemical and physical mechanisms that regulates intercellular communication crucial in maintaining tissue homeostasis. Coordination of the communication occurs through the activity of different actin cytoskeletal regulators, physically connected to extracellular matrix through integrins, generating a platform of biochemical and biomechanical signaling that is deregulated in cancer. Among the major pathways, a controller of cellular functions is the cytokine transforming growth factor β (TGFβ), which remains a complex and central signaling network still to be interpreted and explained in cancer progression. Here, we discuss the link between actin dynamics and TGFβ signaling with the aim of exploring their aberrant interaction in cancer.
Collapse
Affiliation(s)
- Roberta Melchionna
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, via Chianesi 53, 00144 Rome, Italy; (R.M.); (P.T.); (A.T.)
| | - Paola Trono
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, via Chianesi 53, 00144 Rome, Italy; (R.M.); (P.T.); (A.T.)
- Institute of Biochemistry and Cell Biology, National Research Council, via Ramarini 32, 00015 Monterotondo Scalo, Rome, Italy
| | - Annalisa Tocci
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, via Chianesi 53, 00144 Rome, Italy; (R.M.); (P.T.); (A.T.)
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, via Chianesi 53, 00144 Rome, Italy; (R.M.); (P.T.); (A.T.)
- Correspondence: ; Tel.: +39-0652662539
| |
Collapse
|
4
|
Landry NM, Dixon IMC. Fibroblast mechanosensing, SKI and Hippo signaling and the cardiac fibroblast phenotype: Looking beyond TGF-β. Cell Signal 2020; 76:109802. [PMID: 33017619 DOI: 10.1016/j.cellsig.2020.109802] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022]
Abstract
Cardiac fibroblast activation to hyper-synthetic myofibroblasts following a pathological stimulus or in response to a substrate with increased stiffness may be a key tipping point for the evolution of cardiac fibrosis. Cardiac fibrosis per se is associated with progressive loss of heart pump function and is a primary contributor to heart failure. While TGF-β is a common cytokine stimulus associated with fibroblast activation, a druggable target to quell this driver of fibrosis has remained an elusive therapeutic goal due to its ubiquitous use by different cell types and also in the signaling complexity associated with SMADs and other effector pathways. More recently, mechanical stimulus of fibroblastic cells has been revealed as a major point of activation; this includes cardiac fibroblasts. Further, the complexity of TGF-β signaling has been offset by the discovery of members of the SKI family of proteins and their inherent anti-fibrotic properties. In this respect, SKI is a protein that may bind a number of TGF-β associated proteins including SMADs, as well as signaling proteins from other pathways, including Hippo. As SKI is also known to directly deactivate cardiac myofibroblasts to fibroblasts, this mode of action is a putative candidate for further study into the amelioration of cardiac fibrosis. Herein we provide a synthesis of this topic and highlight novel candidate pathways to explore in the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Natalie M Landry
- Department of Physiology and Pathophysiology, Institute of Cardiovascular Sciences, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Ian M C Dixon
- Department of Physiology and Pathophysiology, Institute of Cardiovascular Sciences, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
5
|
Serna-Márquez N, Rodríguez-Hernández A, Ayala-Reyes M, Martínez-Hernández LO, Peña-Rico MÁ, Carretero-Ortega J, Hautefeuille M, Vázquez-Victorio G. Fibrillar Collagen Type I Participates in the Survival and Aggregation of Primary Hepatocytes Cultured on Soft Hydrogels. Biomimetics (Basel) 2020; 5:E30. [PMID: 32630500 PMCID: PMC7345357 DOI: 10.3390/biomimetics5020030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Liver is an essential organ that carries out multiple functions such as glycogen storage, the synthesis of plasma proteins, and the detoxification of xenobiotics. Hepatocytes are the parenchyma that sustain almost all the functions supported by this organ. Hepatocytes and non-parenchymal cells respond to the mechanical alterations that occur in the extracellular matrix (ECM) caused by organogenesis and regenerating processes. Rearrangements of the ECM modify the composition and mechanical properties that result in specific dedifferentiation programs inside the hepatic cells. Quiescent hepatocytes are embedded in the soft ECM, which contains an important concentration of fibrillar collagens in combination with a basement membrane-associated matrix (BM). This work aims to evaluate the role of fibrillar collagens and BM on actin cytoskeleton organization and the function of rat primary hepatocytes cultured on soft elastic polyacrylamide hydrogels (PAA HGs). We used rat tail collagen type I and Matrigel® as references of fibrillar collagens and BM respectively and mixed different percentages of collagen type I in combination with BM. We also used peptides obtained from decellularized liver matrices (dECM). Remarkably, hepatocytes showed a poor adhesion in the absence of collagen on soft PAA HGs. We demonstrated that collagen type I inhibited apoptosis and activated extracellular signal-regulated kinases 1/2 (ERK1/2) in primary hepatocytes cultured on soft hydrogels. Epidermal growth factor (EGF) was not able to rescue cell viability in conjugated BM but affected cell aggregation in soft PAA HGs conjugated with combinations of different proportions of collagen and BM. Interestingly, actin cytoskeleton was localized and preserved close to plasma membrane (cortical actin) and proximal to intercellular ducts (canaliculi-like structures) in soft conditions; however, albumin protein expression was not preserved, even though primary hepatocytes did not remodel their actin cytoskeleton significantly in soft conditions. This investigation highlights the important role of fibrillar collagens on soft hydrogels for the maintenance of survival and aggregation of the hepatocytes. Data suggest evaluating the conditions that allow the establishment of optimal biomimetic environments for physiology and cell biology studies, where the phenotype of primary cells may be preserved for longer periods of time.
Collapse
Affiliation(s)
- Nathalia Serna-Márquez
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Adriana Rodríguez-Hernández
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Marisol Ayala-Reyes
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Lorena Omega Martínez-Hernández
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Instituto de Biotecnología, Universidad del Papaloapan, Tuxtepec CP 68301, Oaxaca, Mexico;
| | - Miguel Ángel Peña-Rico
- Instituto de Biotecnología, Universidad del Papaloapan, Tuxtepec CP 68301, Oaxaca, Mexico;
| | - Jorge Carretero-Ortega
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Mathieu Hautefeuille
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico
| | - Genaro Vázquez-Victorio
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico
| |
Collapse
|
6
|
Li X, Wang J. Mechanical tumor microenvironment and transduction: cytoskeleton mediates cancer cell invasion and metastasis. Int J Biol Sci 2020; 16:2014-2028. [PMID: 32549750 PMCID: PMC7294938 DOI: 10.7150/ijbs.44943] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/15/2020] [Indexed: 12/13/2022] Open
Abstract
Metastasis is a complicated, multistep process that is responsible for over 90% of cancer-related death. Metastatic disease or the movement of cancer cells from one site to another requires dramatic remodeling of the cytoskeleton. The regulation of cancer cell migration is determined not only by biochemical factors in the microenvironment but also by the biomechanical contextual information provided by the extracellular matrix (ECM). The responses of the cytoskeleton to chemical signals are well characterized and understood. However, the mechanisms of response to mechanical signals in the form of externally applied force and forces generated by the ECM are still poorly understood. Furthermore, understanding the way cellular mechanosensors interact with the physical properties of the microenvironment and transmit the signals to activate the cytoskeletal movements may help identify an effective strategy for the treatment of cancer. Here, we will discuss the role of tumor microenvironment during cancer metastasis and how physical forces remodel the cytoskeleton through mechanosensing and transduction.
Collapse
Affiliation(s)
- Xingchen Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - Jianliu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
- Beijing Key Laboratory of Female Pelvic Floor Disorders Diseases, Beijing, 100044, China
| |
Collapse
|
7
|
Abstract
Being concerned by the understanding of the mechanism underlying chronic degenerative diseases , we presented in the previous chapter the medical systems biology conceptual framework that we present for that purpose in this volume. More specifically, we argued there the clear advantages offered by a state-space perspective when applied to the systems-level description of the biomolecular machinery that regulates complex degenerative diseases. We also discussed the importance of the dynamical interplay between the risk factors and the network of interdependencies that characterizes the biochemical, cellular, and tissue-level biomolecular reactions that underlie the physiological processes in health and disease. As we pointed out in the previous chapter, the understanding of this interplay (articulated around cellular phenotypic plasticity properties, regulated by specific kinds of gene regulatory networks) is necessary if prevention is chosen as the human-health improvement strategy (potentially involving the modulation of the patient's lifestyle). In this chapter we provide the medical systems biology mathematical and computational modeling tools required for this task.
Collapse
|
8
|
Tecalco-Cruz AC, Ríos-López DG, Vázquez-Victorio G, Rosales-Alvarez RE, Macías-Silva M. Transcriptional cofactors Ski and SnoN are major regulators of the TGF-β/Smad signaling pathway in health and disease. Signal Transduct Target Ther 2018; 3:15. [PMID: 29892481 PMCID: PMC5992185 DOI: 10.1038/s41392-018-0015-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 02/16/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022] Open
Abstract
The transforming growth factor-β (TGF-β) family plays major pleiotropic roles by regulating many physiological processes in development and tissue homeostasis. The TGF-β signaling pathway outcome relies on the control of the spatial and temporal expression of >500 genes, which depend on the functions of the Smad protein along with those of diverse modulators of this signaling pathway, such as transcriptional factors and cofactors. Ski (Sloan-Kettering Institute) and SnoN (Ski novel) are Smad-interacting proteins that negatively regulate the TGF-β signaling pathway by disrupting the formation of R-Smad/Smad4 complexes, as well as by inhibiting Smad association with the p300/CBP coactivators. The Ski and SnoN transcriptional cofactors recruit diverse corepressors and histone deacetylases to repress gene transcription. The TGF-β/Smad pathway and coregulators Ski and SnoN clearly regulate each other through several positive and negative feedback mechanisms. Thus, these cross-regulatory processes finely modify the TGF-β signaling outcome as they control the magnitude and duration of the TGF-β signals. As a result, any alteration in these regulatory mechanisms may lead to disease development. Therefore, the design of targeted therapies to exert tight control of the levels of negative modulators of the TGF-β pathway, such as Ski and SnoN, is critical to restore cell homeostasis under the specific pathological conditions in which these cofactors are deregulated, such as fibrosis and cancer. Proteins that repress molecular signaling through the transforming growth factor-beta (TGF-β) pathway offer promising targets for treating cancer and fibrosis. Marina Macías-Silva and colleagues from the National Autonomous University of Mexico in Mexico City review the ways in which a pair of proteins, called Ski and SnoN, interact with downstream mediators of TGF-β to inhibit the effects of this master growth factor. Aberrant levels of Ski and SnoN have been linked to diverse range of diseases involving cell proliferation run amok, and therapies that regulate the expression of these proteins could help normalize TGF-β signaling to healthier physiological levels. For decades, drug companies have tried to target the TGF-β pathway, with limited success. Altering the activity of these repressors instead could provide a roundabout way of remedying pathogenic TGF-β activity in fibrosis and oncology.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- 1Instituto de Investigaciones Biomédicas at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Diana G Ríos-López
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | | | - Reyna E Rosales-Alvarez
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Marina Macías-Silva
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| |
Collapse
|
9
|
Walldén K, Nyman T, Hällberg BM. SnoN Stabilizes the SMAD3/SMAD4 Protein Complex. Sci Rep 2017; 7:46370. [PMID: 28397834 PMCID: PMC5387736 DOI: 10.1038/srep46370] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/15/2017] [Indexed: 12/30/2022] Open
Abstract
TGF-β signaling regulates cellular processes such as proliferation, differentiation and apoptosis through activation of SMAD transcription factors that are in turn modulated by members of the Ski-SnoN family. In this process, Ski has been shown to negatively modulate TGF-β signaling by disrupting active R-SMAD/Co-SMAD heteromers. Here, we show that the related regulator SnoN forms a stable complex with the R-SMAD (SMAD3) and the Co-SMAD (SMAD4). To rationalize this stabilization at the molecular level, we determined the crystal structure of a complex between the SAND domain of SnoN and the MH2-domain of SMAD4. This structure shows a binding mode that is compatible with simultaneous coordination of R-SMADs. Our results show that SnoN, and SMAD heteromers can form a joint structural core for the binding of other transcription modulators. The results are of fundamental importance for our understanding of the molecular mechanisms behind the modulation of TGF-β signaling.
Collapse
Affiliation(s)
- Karin Walldén
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Tomas Nyman
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - B Martin Hällberg
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden.,Röntgen-Ångström-Cluster, Karolinska Institutet Outstation, Centre for Structural Systems Biology, DESY-Campus, 22603 Hamburg, Germany.,European Molecular Biology Laboratory, Hamburg Unit, 22603 Hamburg, Germany
| |
Collapse
|
10
|
Escutia-Guadarrama L, Vázquez-Victorio G, Martínez-Pastor D, Nieto-Rivera B, Sosa-Garrocho M, Macías-Silva M, Hautefeuille M. Fabrication of low-cost micropatterned polydimethyl-siloxane scaffolds to organise cells in a variety of two-dimensioanl biomimetic arrangements for lab-on-chip culture platforms. J Tissue Eng 2017; 8:2041731417741505. [PMID: 29225769 PMCID: PMC5714070 DOI: 10.1177/2041731417741505] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/18/2017] [Indexed: 12/13/2022] Open
Abstract
We present the rapid-prototyping of type I collagen micropatterns on poly-dimethylsiloxane substrates for the biomimetic confinement of cells using the combination of a surface oxidation treatment and 3-aminopropyl triethoxysilane silanisation followed by glutaraldehyde crosslinking. The aim of surface treatment is to stabilise microcontact printing transfer of this natural extracellular matrix protein that usually wears out easily from poly-dimethylsiloxane, which is not suitable for biomimetic cell culture platforms and lab-on-chip applications. A low-cost CD-DVD laser was used to etch biomimetic micropatterns into acrylic sheets that were in turn replicated to poly-dimethylsiloxane slabs with the desired features. These stamps were finally inked with type I collagen for microcontact printing transfer on the culture substrates in a simple manner. Human hepatoma cells (HepG2) and rat primary hepatocytes, which do not adhere to bare poly-dimethylsiloxane, were successfully seeded and showed optimal adhesion and survival on simple protein micropatterns with a hepatic cord geometry in order to validate our technique. HepG2 cells also proliferated on the stamps. Soft and stiff poly-dimethylsiloxane layers were also tested to demonstrate that our cost-effective process is compatible with biomimetic organ-on-chip technology integrating tunable stiffness with a potential application to drug testing probes development where such cells are commonly used.
Collapse
Affiliation(s)
- Lidia Escutia-Guadarrama
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Genaro Vázquez-Victorio
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - David Martínez-Pastor
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Brenda Nieto-Rivera
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Marcela Sosa-Garrocho
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Marina Macías-Silva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mathieu Hautefeuille
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
11
|
Abstract
A multitude of physiological processes regulated by G protein-coupled receptors (GPCRs) signaling are accomplished by the participation of active rearrangements of the cytoskeleton. In general, it is common that a cross talk occurs among networks of microfilaments, microtubules, and intermediate filaments in order to reach specific cell responses. In particular, actin-cytoskeleton dynamics regulate processes such as cell shape, cell division, cell motility, and cell polarization, among others. This chapter describes the current knowledge about the regulation of actin-cytoskeleton dynamic by diverse GPCR signaling pathways, and also includes some protocols combining immunofluorescence and confocal microscopy for the visualization of the different rearrangements of the actin-cytoskeleton. We report how both the S1P-GPCR/G12/13/Rho/ROCK and glucagon-GPCR/Gs/cAMP axes induce differential actin-cytoskeleton rearrangements in epithelial cells. We also show that specific actin-binding molecules, like phalloidin and LifeAct, are very useful to analyze F-actin reorganization by confocal microscopy, and also that both molecules show similar results in fixed cells, whereas the anti-actin antibody is useful to detect both the G- and F-actin, as well as their compartmentalization. Thus, it is highly recommended to utilize different approaches to investigate the regulation of actin dynamics by GPCR signaling, with the aim to get a better picture of the phenomenon under study.
Collapse
|