Elsevier

Acta Biomaterialia

Volume 118, December 2020, Pages 141-152
Acta Biomaterialia

Cytoskeleton systems contribute differently to the functional intrinsic properties of chondrospheres

https://doi.org/10.1016/j.actbio.2020.10.007Get rights and content

Abstract

Cytoskeleton systems, actin microfilaments, microtubules (MTs) and intermediate filaments (IFs) provide the biomechanical stability and spatial organization in cells. To understand the specific contributions of each cytoskeleton systems to intrinsic properties of spheroids, we've scrutinized the effects of the cytoskeleton perturbants, cytochalasin D (Cyto D), nocodazole (Noc) and withaferin A (WFA) on fusion, spreading on adhesive surface, morphology and biomechanics of chondrospheres (CSs). We confirmed that treatment with Cyto D but not with Noc or WFA severely affected CSs fusion and spreading dynamics and significantly reduced biomechanical properties of cell aggregates. Noc treatment affected spheroids spreading but not the fusion and surprisingly enhanced their stiffness. Vimentin intermediate filaments (VIFs) reorganization affected CSs spreading only. The analysis of all three cytoskeleton systems contribution to spheroids intrinsic properties was performed for the first time.

Introduction

CSs represent three-dimensional structures organized by densely packed chondrocytes in physiological environment, including in particular naturally synthesized specific ECM proteins, collagen type II and aggrecan. These self-organized structures retain the architectural and functional characteristics typical for the tissue of origin, the cartilage. CSs are the promising tool for cartilage repair, upon transplantation they are able to adhere and to integrate into native hyaline cartilage thus meeting the essential requirement for tissue reconstitution [1]. This approach is known as autologous chondrocyte implantation (ACI), in which articular cartilage biopsy is taken to obtain primary chondrocytes, to culture and multiply them in vitro and to produce CSs to be implanted then into the chondral defect [2]. Additionally, this method overcome the possibility of contamination, inflammation, and immune response since autologous сells without any additional synthetic scaffold are used for implantation. Despite the encouraging potential of CSs in clinical practice, the mechanisms guiding their fundamental functional characteristics such as intrinsic capacity to fuse, attach and spread on the adhesive surface are still poorly understood. Furthermore, CSs biomechanical properties which strongly influence the tissue regeneration and repair, need further elucidation since to form a healthy mature tissue cells must be organized both by spatial and temporal manners.

The development of a three-dimensional configuration involves cell migration, polarization, shape change, the establishment of specific intercellular adhesions and redistribution of cell-cell and cell-surface adhesion forces, aggregation and compaction. These processes are intimately guided by cytoskeleton systems. In self-assembled systems each cell has to organize its cytoskeletal structure in accordance with the cytoskeletal arrangement of the neighboring cells. There is a wealth of evidence to suggest that three cytoskeletal systems, namely the actin cytoskeleton, MTs and IFs network regulate all or part of fore mentioned processes. Several studies have demonstrated the engagement of cytoskeleton systems in course of spheroids formation [3,4], though their precise role in regulating the intrinsic functional capacity of cell aggregates to fuse, to disassemble on adhesive surface and maintain specific morphology has not been thoroughly explored.

Tissue fusion is a fundamental principle in developmental biology [5,6] and is highly relevant to tissue engineering methodology. When spheroids are placed in close contact with each other, they spontaneously coalesce to form one larger structure following the self-assembly principle. Fusion of spheroids is one of the approaches to study how tissues and organs are formed in tissue engineering [7], [8], [9], [10], and CSs are commonly used to study spheroid fusion for cartilage engineering [11], [12], [13], [14]. The intrinsic property of spheroids to fuse offers the exciting possibility of fabricating tissue constructs with a complex cytoarchitecture by directed assembly of numerous cell aggregates. Previous studies analyzing the fusion of spheroids revealed that their fusion could be described by a model of the coalescence of highly viscous liquid drops [5,6,[15], [16], [17], [18]]. According to this concept, the viscosity and surface tension determine the fusion [19]. In agreement with the differential adhesion hypothesis (DAH) [6,16,17], spheroids fuse over time to a more energetically favorable configuration of one larger spheroid. Cell aggregates, however, are much more complicated than liquid drops and the mechanisms involved in spheroids fusion needs further elucidation. Another possible approach in tissue engineering is biofabrication of tissue sheets by attachment and spreading of patterned spheroids on the biocompatible adhesive matrix or scaffold followed by gradual fusion of disassembling cell aggregates [20,21]. Spreading of spheroids could be also used for biocompatibility tests of various biomaterials and scaffolds.

The biomechanical properties of cells guide their ultimate three-dimensional organization in tissue. In gastrulating zebrafish embryos, actin- and myosin-mediated tension and differential adhesion among cells drive the sorting of germ-layer progenitor cells [22]. Cell proliferation within spheroids is affected not only by spacial gradients of soluble factors such as oxygen, nutrients, and regulatory macromolecules, but by external forces also. Spheroids from murine mammary carcinoma cells generated within an agarose gel, were exposed to compressive loads during growth, the cells proliferated more slowly at zones of high stress and induced apoptotic cell death in regions of significantly higher mechanical stress [23]. As more examples of cells within three-dimensional aggregates responding to mechanical cues through the cytoskeleton systems are found, the questions of exact role of each component, namely actin microfilaments, microtubules, intermediate filaments, are becoming central.

Applying the classic perturbants of cytoskeleton, Cyto D, Noc and WFA, we assessed the potential role of actin microfilaments, MTs and IFs respectively in CSs fusion and spreading dynamics, in course of maintaining their biomechanical properties. Cyto D was used to demonstrate that an intact F-actin network is required for CSs fusion, spreading and supports biomechanical properties. According to the classic study by Cooper [24], low concentration of 0.2μM Cyto D inhibits membrane ruffling, higher concentrations of 2-20μM Cyto D are necessary to remove stress fibers. To evaluate the effect of stress fibers on cardiovascular spheroid composition, the concentration of 5μM Cyto D for 5 days incubation was used [25]. Cyto D and Noc concentrations used in this study were reported to disrupt the specific cytoskeletal elements without compromising the cell viability [26,27]. Noc is commonly used to study MT-dependent processes because of its ability to depolymerize rapidly MTs when administered at micromolar concentrations [28], while low concentrations of nocodazole inhibit microtubule dynamic instability in vitro (4 nM-12μM) and in cells (4-400 nM) [29]. Noc didn't affect СS fusion, but influenced essentially CSs spreading and biomechanical properties. WFA which is known to influence a variety of IFs, has shown promise as a potent vimentin inhibitor. At concentrations between 250 nM and 1 μM, increased serine-38 phosphorylation of vimentin causes the retraction of VIFs from the cell periphery and its juxtanuclear localization [30]. Concentrations at and above 1 μM WFA show dose-dependent effects on VIFs causing perinuclear condensation [31,32]. Exceeding 2 μM, WFA causes apoptosis and secondary vimentin cleavage within 2–4 h [30,31,33]. According to our data, WFA has affected spheroids spreading only. In our study we deliberate the mechanisms underlying observed effects of cytoskeleton perturbants on CSs intrinsic properties.

Section snippets

Chondrocyte culture

Primary sheep chondrocytes were a generous gift from Dr. N.P. Omelyanenko (National Medical Research Centre of Traumatology and Orthopedics of N.N. Priorov, Moscow, Russian Federation). Cells were cultivated in DMEM (Gibco, Cat # 12491-015) containing 10% FBS (Gibco, Cat # 16000-044), supplemented with antibiotic/antimycotic (Gibco, Cat # 15240-062) and 2mM L-glutamine (Paneco, Cat # F032). Primary sheep chondrocytes were cultured in cell culture flasks (Greiner Bio-One, Cat # 658175) at

Cytoskeleton-disorganizing drugs alter morphology of individual cells and spheroids

To determine the role of actin microfilaments, MTs and IFs on cell morphology, the cells and spheroids were cultured in the presence of 10μM Cyto D, 1μM Noc or 1μM WFA. Prior to analysing CSs morphology, the cytoskeleton perturbants effect was determined on chondrocytes in 2D monolayer.

Actin visualization in single cells showed a well-organized microfilament network including leading edge meshwork and clearly distinguishable stress fibers running the length of the cells and appeared prominent

Discussion

Three types of filaments, actin filaments, MTs and IFs, collectively termed cytoskeleton, are differently distributed within the cytoplasm. The migrating cell leading edge is formed exclusively by actin meshwork but little or no MTs or IFs enter this region. Mitotic apparatus is rich in MTs but no microfilaments or IFs are found wherein. The specific spacial distribution of filaments with different structural properties could be indicative to the divergent function of each cytoskeleton system.

Conclusion

Understanding the impact of each cytoskeleton system on intrinsic functional properties of three-dimensional chondrocytes aggregates is a step towards tuning these interactions and opens the door to CSs biology and cartilage research. Applying specific perturbants for each cytoskeleton system we showed that intact actin network but not MTs or VIFs has a quintessential role in all intrinsic functional CSs properties as a cardinal biomechanical and contractile system in spheroids. Wherein

Authors' contribution

E.A.B. and V.A.M. designed the research; A.A.G, E.V.K., N.Yu.M., S.A.R., F.D.A.S.P., V.A.P., V.A.K. performed the research; A.A.G., E.V.K., S.A.R., A.V.K. analysed the data; E.A.B, A.A.G., E.V.K., Y.D.K. and V.A.M. wrote the paper.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

The reported study was funded by RFBR, project number 19-315-90017.

References (84)

  • C. Rotsch et al.

    Drug-induced changes of cytoskeletal structure and mechanics in fibroblasts: an atomic force microscopy study

    Biophys. J.

    (2000)
  • R. Foty et al.

    The differential adhesion hypothesis: a direct evaluation

    Dev. Biol.

    (2005)
  • H.I. Jung et al.

    Colchicine activates actin polymerization by microtubule depolymerization

    Mol. Cells

    (1997)
  • S.-H. Kee et al.

    Cell-cell adhesion and RhoA-mediated actin polymerization are independent phenomena in microtubule disrupted keratinocytes

    J. Invest. Dermatol.

    (2002)
  • D. Borin et al.

    An engineering insight into the relationship of selective cytoskeletal impairment and biomechanics of HeLa cells

    Micron

    (2017)
  • J. Li et al.

    Vimentin dephosphorylation at ser-56 is regulated by type 1 protein phosphatase in smooth muscle

    Respir. Res.

    (2016)
  • J. Ivaska et al.

    Novel functions of vimentin in cell adhesion, migration, and signaling

    Exp. Cell Res.

    (2007)
  • C. Leduc et al.

    Intermediate filaments in cell migration and invasion: the unusual suspects

    Curr. Opin. Cell Biol.

    (2015)
  • T. Schubert et al.

    Long-term effects of chondrospheres on cartilage lesions in an autologous chondrocyte implantation model as investigated in the SCID mouse model

    Int. J. Mol. Med.

    (2009)
  • R. Siebold et al.

    Good clinical and MRI outcome after arthroscopic autologous chondrocyte implantation for cartilage repair in the knee

    Knee Surg. Sports Traumatol. Arthrosc.

    (2018)
  • E.S. Tzanakakis et al.

    The role of actin filaments and microtubules in hepatocyte spheroid self-assembly

    Cell Motil. Cytoskelet.

    (2001)
  • A. Tsai et al.

    Compaction, fusion, and functional activation of three-dimensional human mesenchymal stem cell aggregate

    Tissue Eng. Part A

    (2015)
  • J.M. Perez-Pomares et al.

    Tissue fusion and cell sorting in embryonic development and disease: biomedical implications

    Bioessays

    (2006)
  • K. Jakab et al.

    Engineering biological structures of prescribed shape using self-assembling multicellular systems

    Proc. Natl. Acad. Sci. USA

    (2004)
  • P.A. Fleming et al.

    Fusion of uniluminal vascular spheroids: a model for assembly of blood vessels

    Dev. Dyn.

    (2010)
  • M. Lehmann et al.

    Three-dimensional scaffold- free fusion culture: the way to enhance chondrogenesis of in vitro propagated human articular chondrocytes

    Eur. J. Histochem.

    (2013)
  • E. Vrij et al.

    Directed assembly and development of material-free tissues with complex architectures

    Adv. Mater.

    (2016)
  • J.M. Kelm et al.

    Design of custom- shaped vascularized tissues using microtissue spheroids as minimal building units

    Tissue Eng.

    (2006)
  • S. Bhumiratana et al.

    Large, stratified, and mechanically functional human cartilage grown in vitro by mesenchymal condensation

    Proc. Natl Acad. Sci.

    (2014)
  • M.S. Steinberg et al.

    Experimental specification of cell sorting, tissue spreading, and specific spatial patterning by quantitative differences in cadherin expression

    Proc. Natl. Acad. Sci. USA

    (1994)
  • M.S. Steinberg

    Reconstruction of tissues by dissociated cells. Some morphogenetic tissue movements and the sorting out of embryonic cells may have a common explanation

    Science

    (1963)
  • K. Jakab et al.

    Relating cell and tissue mechanics: implications and applications

    Dev. Dyn.

    (2008)
  • T.V. Stirbat et al.

    Fine tuning of tissues’ viscosity and surface tension through contractility suggests a new role for α-catenin

    PLoS ONE

    (2013)
  • E.V. Koudan et al.

    Patterning of tissue spheroids biofabricated from human fibroblasts on the surface of electrospun polyurethane matrix using 3D bioprinter

    Int. J. Bioprinting

    (2016)
  • V. Beachley et al.

    The fusion of tissue spheroids attached to pre-stretched electrospun polyurethane scaffolds

    J. Tissue Eng.

    (2014)
  • M. Krieg M et al.

    Tensile forces govern germ-layer organization in zebrafish

    Nat. Cell Biol.

    (2008)
  • G. Cheng et al.

    Micro-environmental mechanical stress controls tumor spheroid size and morphology by suppressing proliferation and inducing apoptosis in cancer cells

    PLoS ONE

    (2009)
  • J.A. Cooper

    Effects of cytochalasin and phalloidin on actin

    J. Cell Biol.

    (1987)
  • Y. Yan et al.

    Cell population balance of cardiovascular spheroids derived from human induced pluripotent stem cells

    Sci. Rep.

    (2019)
  • G.W. Beresford et al.

    Cytochalasin D exerts stimulatory and inhibitory effects on insulin-induced glucokinase mRNA expression in hepatocytes

    Mol. Cell Biochem.

    (1994)
  • D.J. Mooney et al.

    Cytoskeletal filament assembly and the control of cell spreading and function by extracellular matrix

    J. Cell Sci.

    (1995)
  • M.J. De Brabander et al.

    The effects of methyl [5-(2-thienylcarbonyl)-1H- benzimadazol-2-yl]carbanate(R 17934; NSC 238159), a new synthetic antitumoral drug interfering with microtubules, on mammalian cells cultured in vitro

    Cancer Res.

    (1976)
  • Cited by (9)

    • Engineered biomaterials to guide spheroid formation, function, and fabrication into 3D tissue constructs

      2023, Acta Biomaterialia
      Citation Excerpt :

      Spheroid fusion is a biophysical process that occurs over a 24–48 h period and is thought to be driven by the difference in interfacial surface tension between the surrounding culture media and cells, which can be modulated by ECM secretion, cell type, actin dynamics, and the formation of a condensation boundary (ECM deposition and peripheral localization) with time [2,15,16]. Spheroid fusion and the corresponding increase in inter-spheroid contact area results in reduced surface area and tension for individual spheroids [17]. Additionally, as thoroughly described by Bhumiratana et al., it is important that initial spheroid contact occurs before the setting of a condensation boundary (e.g., <7days for human MSCs) [2]; otherwise, spheroid fusion is limited and rather aggregates of spheroids loosely connected by peripherally deposited ECM are observed [18].

    • Nose to Spine: spheroids generated by human nasal chondrocytes for scaffold-free nucleus pulposus augmentation

      2021, Acta Biomaterialia
      Citation Excerpt :

      For NCS-NPS fusion, NPS pre-formed in DDD mimicking conditions (Section 2.3) were placed together with 1-, 3- or 7-day-old NCS. The pairs of NCS-NCS and NCS-NPS were incubated at 37 °C with 5% CO2, 2% O2 (hypoxia) or 20% O2 (standard) for 96 h [37]. Images of spheroid doublets were used for quantitative analysis of the fusion process.

    View all citing articles on Scopus

    Both authors made an equal contribution to this article.

    View full text