48
Views
3
CrossRef citations to date
0
Altmetric
Review

Current status and perspectives in atomic force microscopy-based identification of cellular transformation

, &
Pages 2107-2118 | Published online: 17 May 2016

Abstract

Understanding the complex interplay between cells and their biomechanics and how the interplay is influenced by the extracellular microenvironment, as well as how the transforming potential of a tissue from a benign to a cancerous one is related to the dynamics of both the cell and its surroundings, holds promise for the development of targeted translational therapies. This review provides a comprehensive overview of atomic force microscopy-based technology and its applications for identification of cellular progression to a cancerous phenotype. The review also offers insights into the advancements that are required for the next user-controlled tool to allow for the identification of early cell transformation and thus potentially lead to improved therapeutic outcomes.

Malignant transformation: from a normal to a cancerous phenotype

As a cell or a community of cells, a biological system’s hierarchical organization allows for structure formation and system functionality. Alteration of biological processes, such as cell cycleCitation1 or cell division,Citation2 may lead to fundamental disorder at the molecular level and induce activation of genetic changes ().Citation3

Figure 1 Cell division and cell transformation under epigenetically induced factors.

Figure 1 Cell division and cell transformation under epigenetically induced factors.

Particular hallmarks of cell transformation, tumor initiation, and subsequently progression have been related to the intricacies of cellular regulation.Citation4 Similarly, epigenetic alterations inducing inflammation and microenvironment changes () have been shown to influence phenotype transformation.Citation5 For instance, previous reports on epigenetic transformation revealed that metastatic properties are related to the abnormal expression of key regulatory genes. Tumor-suppressor genes could, for instance, restrain cell division as well as induce cell death, while loss of gene function could result in abnormal cellular behavior, perturbation of the cell cycle, and lack of cell regulation.Citation6 Previous studies have also indicated that LIMD1 overexpression could retard cell-cycle progression and block S-phase entry, leading to cell accumulation in the G0/G1 phase and subsequent changes in cellular behavior;Citation7 LIMD1 is a tumor-suppressor gene located at chromosome 3p21.3, a region commonly deleted in many malignancies.Citation6 Another study found that deregulation of microcephalin and ASPM expression (involved in the regulation of neurogenesis)Citation8 leads to abnormal cell division and subsequently to cancer progression.Citation7 Microcephalin is involved in the DNA-damage response and has been linked to tumor formation and cancer invasion (). Other analysis revealed that aberrant hypomethylation-mediated AGR2 overexpression could cause an aggressive phenotype in ovarian cancer cells;Citation9 AGR2 is developmentally regulated initially discovered as an estrogen-responsive gene in breast cancer cell lines currently linked to tumors with poor outcome.Citation10

Figure 2 Epigenetic changes in tumor cells and tumor development.

Note: The later stages in tumor development are supported by angiogenesis.

Figure 2 Epigenetic changes in tumor cells and tumor development.Note: The later stages in tumor development are supported by angiogenesis.

Figure 3 Epigenetic transformation correlates with stages of cells and tumor transformation when microcephalin is considered.

Notes: Microcephalin is known to play a key role in carcinogenesis, by being a centrosome-associated protein and being involved in mitosis, with an important role in DNA-damage response. Microcephalin is encoded by the MCPH1 and MCPH5 genes in ovarian tissue. Normal ovarian epithelial tissue with high microcephalin expression (A). Strong microcephalin expression in low-grade tumor cells (B), moderate microcephalin in grade 2 (C), and low levels of nuclear microcephalin expression in a high-grade tumor (D). All images 40× magnification. (E) Nuclear microcephalin expression decreases with increasing tumor grade (P<0.0001 using analysis of variance). Reproduced from Alsiary R, Brüning-Richardson A, Bond J, Morrison EE, Wilkinson N, Bell SM. Deregulation of microcephalin and ASPM expression are correlated with epithelial ovarian cancer progression. Plos One. 2014;9(5):e97005 (http://creativecommons.org/licenses/by/4.0/).Citation7

Figure 3 Epigenetic transformation correlates with stages of cells and tumor transformation when microcephalin is considered.Notes: Microcephalin is known to play a key role in carcinogenesis, by being a centrosome-associated protein and being involved in mitosis, with an important role in DNA-damage response. Microcephalin is encoded by the MCPH1 and MCPH5 genes in ovarian tissue. Normal ovarian epithelial tissue with high microcephalin expression (A). Strong microcephalin expression in low-grade tumor cells (B), moderate microcephalin in grade 2 (C), and low levels of nuclear microcephalin expression in a high-grade tumor (D). All images 40× magnification. (E) Nuclear microcephalin expression decreases with increasing tumor grade (P<0.0001 using analysis of variance). Reproduced from Alsiary R, Brüning-Richardson A, Bond J, Morrison EE, Wilkinson N, Bell SM. Deregulation of microcephalin and ASPM expression are correlated with epithelial ovarian cancer progression. Plos One. 2014;9(5):e97005 (http://creativecommons.org/licenses/by/4.0/).Citation7

With regard to the influence of the microenvironment on cells and how changes in such microenvironmentsCitation11 could lead to phenotypic progression and cancer transformation,Citation12 analysis showed that alterations in external factors, such as the pH of a tissue, could trigger cellular changesCitation13 and regulate cell transformation.Citation14 An acidic environment (pH 6.5–6.9 compared with pH 7.2–7.4) could cause degradation of the extracellular matrix (ECM) and lead to the secretion of VEGF, as well as to angiogenesis, which can correlate with tumor transformation and cellular invasion ().Citation15 Analysis also showed that proteins in the ECM could interact directly with tumor cells, eg, via their integrin surface receptors, to influence and subsequently alter cellular behavior, proliferation, apoptosis, migration, or differentiation.Citation16 Remodeling of the ECM could occur in tumor-fibroblast cells and subsequently accelerate cancer progression.Citation17 During the remodeling process, the mechanical response of the ECM can change progressively,Citation18 leading to enhanced cell migrationCitation19 followed by invasion at specific interfaces.Citation20 Also, it was found that extracellular vesicles could regulate and modulate the microenvironment of the cancer cellCitation21 by regulating cellular communication and playing an important role in phenotype transformationCitation22 and cancer progression.Citation23

Figure 4 Glucose metabolism is increased in cancer cells and could produce an acidic environment that may promote degradation of the extracellular matrix and influence local cellular invasion.

Notes: Estrella et alCitation15 evaluated in vivo pH and its role on tumor development. Tumor pH measurements were obtained using a microelectrode and measuring under the skin of a sedated mouse near the tumor site. The authors performed two measurements at each position, and three positions were investigated at each time point and averaged. The average pH of the tumors for each cell line are shown, with a significantly (*P<0.01) lower pH in HCT116 compared to MDAMB231 tumors. Adapted from Cancer Research, Copyright 2013, Volume 73, Pages 1524–1535, Estrella V, Chen TA, Lloyd M, et al, Acidity generated by the tumor microenvironment drives local invasion, with permission from AACR.Citation15

Figure 4 Glucose metabolism is increased in cancer cells and could produce an acidic environment that may promote degradation of the extracellular matrix and influence local cellular invasion.Notes: Estrella et alCitation15 evaluated in vivo pH and its role on tumor development. Tumor pH measurements were obtained using a microelectrode and measuring under the skin of a sedated mouse near the tumor site. The authors performed two measurements at each position, and three positions were investigated at each time point and averaged. The average pH of the tumors for each cell line are shown, with a significantly (*P<0.01) lower pH in HCT116 compared to MDAMB231 tumors. Adapted from Cancer Research, Copyright 2013, Volume 73, Pages 1524–1535, Estrella V, Chen TA, Lloyd M, et al, Acidity generated by the tumor microenvironment drives local invasion, with permission from AACR.Citation15

The malignant transformation is accompanied by changes in cell structureCitation24 and morphology,Citation25 as well as changes in cellular responses to stimuli.Citation26 Studies revealed that alterations in cell biophysical properties, especially in their biomechanical characteristics, represent an early indicator of disease progression,Citation27Citation29 with phenotypic events being mostly a result of cytoskeleton remodeling.Citation30 It was observed that BRMS1 expression in 435 cells caused changes in their architecture, leading to alterations in their biomechanical properties, with such changes being due to the reconstruction of actin cytoskeletal networks.Citation31

However, even though the identification of the molecular link between chronic inflammation and cancer was elucidated through NFκB, the initial transformation and activation pathways remain to be clarified and chronic inflammation-induced tumorigenesis to be elucidated. Further, while significant advances have been made toward elucidating the molecular mechanisms that underlie cancer progression, fundamental questions associated with the biomechanical traits that lead to cancer development and subsequently to metastasis remain to be answered. For instance, with tumor progression being a result of the ability of transformed cells to communicate and influence one another, as well as their neighboring healthy cells,Citation32 and with clinical observations of distinct tumors from different organs seeming to be cell-specific,Citation33 it is unclear how and whether transformation of a cell or a community of cells in an organ could lead to different tumor phenotypes or if such transformation is propagated through the microenvironment.

Biomechanical cues help identify cell transformation

With the development of atomic force microscopy (AFM) technology, computer-assisted applications for biomedical fields for analyses of biological information in terms of biomechanical characteristics have spiked.Citation34Citation37 Obtaining the “bio-nano-mechanical signature” of cells or tissues has thus been proposed as the next method that could possibly lead to the development of a differentiation tool of pathological conditions from normal ones. As such, nanoindentation or the ability to detect and measure changes in the elasticity or the elastic responses of soft materials, such as cells,Citation38 manifested as a readout in displacement-versus-force curves (), indicated that cancer cells have different characteristics when compared to healthy cells.Citation26,Citation39 In particular, studies have shown that the stiffness,Citation40 migration ability,Citation41 and morphologyCitation42 of cancer cells differ from healthy cells, with single cancer cells showing lower stiffness and higher deformability,Citation43 and with such characteristics being dependent on the type of cell being analyzed.Citation44 Further, complex examination of individual cells and their cellular components showed that the different elastic properties of cancer cells are a result of local or global changes that contribute to or induce their transformation.Citation26

Figure 5 Schematic demonstration of the nanoindentation technique of an individual cell.

Note: The phases of the tip approaching the sample are illustrated, as well as local and temporal indentation in the surface of the cell.

Figure 5 Schematic demonstration of the nanoindentation technique of an individual cell.Note: The phases of the tip approaching the sample are illustrated, as well as local and temporal indentation in the surface of the cell.

Analysis of the membranes of both cancer and healthy cells showed that while the surface of the latter revealed brush-like structures of one specific length and were formed from glycocalyx layers with pericellular coatings,Citation45,Citation46 the surface of cancer cells displayed both long and short brushes with significantly different geometries and densities. Such brush-like geometry is known to provide support for cell–cell interactions, cell migration, and differentiation.Citation26

Paired with changes in the cellular membrane were changes in submembrane or cytoskeletal structures. For instance, analysis of malignant (MCF7) and nonmalignant (MCF10A) breast cells revealed that MCF10A cells had an apparent Young’s modulus significantly higher than that of their malignant counterparts,Citation47 with topographic images of such cells revealing well-structured microfilaments that were different than the ones observed for the MCF7 cells (). The differences in cytoskeletal organization were attributed to dissimilarities in the cell mechanical properties, and could possibly explain the increased migration and invasion ability of the malignant cells, especially during metastasis.

Figure 6 Contact-mode atomic force microscopy was used to investigate live MCF7 and MCF10A cells.

Notes: Analysis shows the central part of (A) MCF7 and (B) MCF10A cells with a scale bar of 2 μm. Specifically, MCF7 cells comprise less well-defined filamentous structures with disorganized ridges, which is in contrast with MCF10A cells, which have well-aligned filamentous structures below the membrane. Reprinted from Biochemical and Biophysical Research Communications, Volume 374, Li QS, Lee GY, Ong CN, Lim CT, AFM indentation study of breast cancer cells, Pages 609–613, Copyright © 2008, with permission from Elsevier.Citation47

Figure 6 Contact-mode atomic force microscopy was used to investigate live MCF7 and MCF10A cells.Notes: Analysis shows the central part of (A) MCF7 and (B) MCF10A cells with a scale bar of 2 μm. Specifically, MCF7 cells comprise less well-defined filamentous structures with disorganized ridges, which is in contrast with MCF10A cells, which have well-aligned filamentous structures below the membrane. Reprinted from Biochemical and Biophysical Research Communications, Volume 374, Li QS, Lee GY, Ong CN, Lim CT, AFM indentation study of breast cancer cells, Pages 609–613, Copyright © 2008, with permission from Elsevier.Citation47

Studies also showed variations in the stiffness of the cytoskeleton of nonmalignant MCF10A and malignant MDAMB231 breast cells resulting from the growth media,Citation48 with analysis revealing that the mean elastic modulus of MCF10A cells was higher than that of the MDAMB231 cells cultured in identical conditions. Furthermore, the characteristics of the malignant breast cells and their nonmalignant counterparts in the 27°C–34°C temperature rangeCitation49 indicated that the malignant cells were softer than their counterparts. The significant difference in the cellular Young’s modulus between malignant and healthy cells was due to the structural differences recorded, especially in their three-dimensional cytoskeleton, with the cytoskeletal structures of the cancer cells showing a more fluidlike state than the cytoskeleton of the healthy cells.Citation50

Membrane and cytoskeletal changes were largely accompanied by nuclear transformations, with AFM nanoindentation-based analysis of isolated nuclei of healthy (MCF10A) and malignant (MCF7) human breast epithelial cells showing mechanical changes in relation to individual cell type.Citation51 In particular, the apparent Young’s modulus of MCF7 cell nuclei was much lower than that of MCF10A cell nuclei, mainly because of the alternation of their underlying lamina (lamin A/C) structure. The study also indicated that the nucleus’ deformability was tightly related to the softening of cancer cells. Complementary AFM nanomechanical-based analysis of several areas of healthy and cancer oligodendro-cytesCitation52 showed that the elastic modulus of the nuclear regions of the cancer cells was lower than that of the cell periphery, with cancer cells being threefold softer than healthy cells. Similar results were obtained for malignant thyroid cells,Citation53 with the elastic analysis showing that such malignant cells were three- to fivefold softer relative to their primary and untransformed counterparts.

Nanoindentation allows cell-type and metastatic potential differentiation

Nanoindentation enabled detection of cancerous cells from a mixture with healthy cells.Citation44 In particular, the elasticity of human kidney cell lines from different types of tumors (ie, carcinoma [A498] and adenocarcinoma [ACHN]) was compared with that of a benign cell line (RC124). For this, dozens of individual cells were mapped, and more than 15,000 data points per cell were generated to calculate the sample’s viscoelastic properties and the elastic modulus. Systematic comparisons indicated that cancer cells had distinctive elastic properties, with the Young modulus of the A498 carcinoma cell line being larger than that of the adenocarcinoma. Similarly, a direct comparison of elastic properties of human breast cancer cells (MCF7) and human cervical carcinoma cells (HeLa), known to have similar phenotypes,Citation54 showed that HeLa cells were three times harder than MCF7 cells.

Different prostate cancer cells showed distinctive metastatic potential, which was subsequently correlated with variations in their elastic moduli.Citation55 For instance, the elastic modulus of a primary benign prostate hyperplasia cell line was greater than that of the LNCaP-clone-FGC and PC3 cell lines, with the Young modulus of the highly invasive PC3 cells being higher than that of the noninvasive LNCaP-clone-FGC cells. Such variations point to distinctive clinical behavior and potentially hint at different therapeutic outcomes for such cells.

When nanoindentation was used to detect cancer progression in tissue sections originating from patients,Citation56 analysis showed that the apparent Young modulus of the cancer region was half that of the nonneoplastic tissue. Moreover, elastic mapping conducted on healthy tissues showed homogeneous modulus profiles characterized by a single distinct peak. In contrast, malignant tissues had a distinctive broad distribution of their Young moduli mainly originating from the tissue heterogeneities, with cancer cells revealing prominent low-stiffness peaks.Citation57 Further, such differences in cell stiffness could also be related to identification of biomarkers with potential invasive and metastatic characteristics ().Citation58

Figure 7 Representative force-indentation curves revealing the stiffness distribution of different ovarian cell lines from IOSE and Hey cells.

Notes: Analyses revealed that ovarian cancer cells were generally softer and had lower intrinsic variability in stiffness when compared to their nonmalignant counterparts, thus pointing to cell-stiffness characteristics possibly being used as biomarkers for identifying metastatic potential. Adapted from Xu WW, Mezencev R, Kim B, Wang LJ, McDonald J, Sulchek T. Cell stiffness is a biomarker of the metastatic potential of ovarian cancer cells. Plos One. 2012;7:0046609 (http://creativecommons.org/licenses/by/4.0/).Citation58

Figure 7 Representative force-indentation curves revealing the stiffness distribution of different ovarian cell lines from IOSE and Hey cells.Notes: Analyses revealed that ovarian cancer cells were generally softer and had lower intrinsic variability in stiffness when compared to their nonmalignant counterparts, thus pointing to cell-stiffness characteristics possibly being used as biomarkers for identifying metastatic potential. Adapted from Xu WW, Mezencev R, Kim B, Wang LJ, McDonald J, Sulchek T. Cell stiffness is a biomarker of the metastatic potential of ovarian cancer cells. Plos One. 2012;7:0046609 (http://creativecommons.org/licenses/by/4.0/).Citation58

Bionanomechanical-based AFM functional analysis helped detect metastatic tumor cells in bodily fluids,Citation57 with Cross et al showing that lung, breast, and pancreas cancer cells displayed significantly decreased stiffness when compared to healthy control counterparts.Citation57 Rother et al investigated viscoelastic responses of nine cell lines (ie, NIH3T3 fibroblasts, MDCKII, NMuMG, A549, SW13, MCF7, MCF10A, MDAMB231, and Caki1 cells) from four different organs, all known to have variable metastatic potential,Citation56 () and showed that cells from the organ exhibiting malignancy were generally softer than their benign cell counterparts.Citation59

Figure 8 Young’s modulus distributions and Gaussian fits (as lines).

Notes: The Young’s modulus distributions with fitted Gaussian functions (lines) obtained for (A) prostate (PC-3, LNCaP, and Du145) cancer cell lines and breast (MCF7 and T47D) and (B) compared to normal breast (184A) and prostate (PZHPV-7) cell lines. Reprinted from Archives of Biochemistry and Biophysics, Volume 518, Lekka M, Gil D, Pogoda K, et al, Cancer cell detection in tissue sections using AFM, Pages 151–156, Copyright © 2012, with permission from Elsevier.Citation47

Figure 8 Young’s modulus distributions and Gaussian fits (as lines).Notes: The Young’s modulus distributions with fitted Gaussian functions (lines) obtained for (A) prostate (PC-3, LNCaP, and Du145) cancer cell lines and breast (MCF7 and T47D) and (B) compared to normal breast (184A) and prostate (PZHPV-7) cell lines. Reprinted from Archives of Biochemistry and Biophysics, Volume 518, Lekka M, Gil D, Pogoda K, et al, Cancer cell detection in tissue sections using AFM, Pages 151–156, Copyright © 2012, with permission from Elsevier.Citation47

When elastic moduli and cell-substrate adhesion analyses for lowly (LNCaP) and highly (CL1, CL2) metastatic human prostate cancer cells were performed,Citation60 results showed that the elastic moduli of CL1 and CL2 were greater than those of LNCaP, with CL1 and CL2 displaying a significantly larger area to cell-substrate adhesion relative to the LNCaP cells. The increased elastic moduli found in CL1 and CL2 were attributed to the enhanced tensile stress generated from the actin cytoskeleton anchored on more focal adhesion sites, hinting again at different metastatic potential of such cells. Complementary analysis of late-stage tumors in the lungs of mice indicated that migration and metastatic spreading characteristics were related to the low elasticity of hypoxia-associated cancer cells. In particular, bionanomechanical response experiments on benign mesothelial cells and metastatic cancer cells derived from human body fluidsCitation61 showed that the Young modulus of metastatic tumor cells was about 80% lower than that of the benign cells. Moreover, cell-adhesion analysis of these malignant cells revealed an overall 33% reduced adhesion when compared to the adhesion of benign cell counterparts.

A summary of the changes in elasticity of different types of tissues and cells, all relative to controls (noncancerous cells) is in . The results suggest that the morphology factor alone was sufficient to explain differences in stiffness for the cell types being considered and hint at mechanisms of cell invasion being based on reduced adhesion of cells to their three-dimensional environment.

Table 1 Changes in elasticity of different types of tissues and cells, all relative to controls (noncancerous cells)

Is there a next generation of AFM-based tools to be used for clinical identification of pathological conditions?

To advance AFM-based technologies for clinical detection of cell transformation, a trained workforce and consecutive probing of cell characteristics are required. However, with reports showing that such consecutive measurements can influence the mechanics of individual cells,Citation62 and with rigorous statistical analyses revealing that sample-preparation conditions need to be accounted for when reliability in cancer diagnosis is envisioned, the heterogeneity and complexity of a transformed cell is more than a mechanical cue indicator that permits evaluation and readability of changes in terms of cellular elasticity. As such, if AFM is to be used for advanced diagnosis and for clinical detection, the heterogeneity of the cellular sample needs to be accounted for. In particular, with analysis showing that molecular composition plays a critical role in the mechanics of the tissue,Citation63 for AFM to be a viable and useful tool for clinicians, analysis of the plasticity of a cell populationCitation64,Citation65 needs to be supported by biochemical assays,Citation66 gene expression,Citation67 and immunofluorescent labeling.Citation68

Research combining AFM and biochemical assays has already shown that the interaction and bond formation of single P-selectin–ligand complexes can be quantified. P-selectin is located on the endothelial cell wall, and helps support the leukocyte when under hydrodynamic flow; the complex formed upon association with glycoproteins was shown to exhibit chain-like elasticity with a 5.3 pN·nm−1 molecular spring constant and a 0.35 nm persistence length.Citation69 Other studies indicated that DNA-dependent protein kinase can bind at the DNA termini, with such binding being characterized by AFM.Citation70 Also, high-speed AFM was used to reveal ATP-driven motor F1-rotary behavior and quantify its conformational changes, as well as confirm the dependence of the initial rates of ATP hydrolysis, as determined through biochemical assay ().Citation71 Lastly, stretching single polysaccharides and proteins in the presence of AFM has been demonstrated, thus contributing to the body of work illustrating the biophysics and chemical biology of a protein–protein bond or enzymatic reactions related to force applications.Citation72

Figure 9 F1-ATPase is a rotary motor protein present in the cellular membrane as the water-soluble portion of ATP synthase.

Notes: Changes in the rotary motor evaluated using atomic force microscopy. Analysis showed that the crystal structure of the motor protein was a function of the complex binding of ATP (A, B), with dependence of initial rates of ATP hydrolysis being determined by biochemical assay and being correlated with rates of conformational change of the motor protein (C). From Uchihashi T, Iino R, Ando T, Noji H. High-speed atomic force micros copy reveals rotary catalysis of rotorless F1-ATPase. Science. 2011;333:755–758. Available from: http://science.sciencemag.org/content/333/6043/755. Reprinted with permission from AAAS.Citation71

Figure 9 F1-ATPase is a rotary motor protein present in the cellular membrane as the water-soluble portion of ATP synthase.Notes: Changes in the rotary motor evaluated using atomic force microscopy. Analysis showed that the crystal structure of the motor protein was a function of the complex binding of ATP (A, B), with dependence of initial rates of ATP hydrolysis being determined by biochemical assay and being correlated with rates of conformational change of the motor protein (C). From Uchihashi T, Iino R, Ando T, Noji H. High-speed atomic force micros copy reveals rotary catalysis of rotorless F1-ATPase. Science. 2011;333:755–758. Available from: http://science.sciencemag.org/content/333/6043/755. Reprinted with permission from AAAS.Citation71

Research combining AFM with gene-expression analysis has reported on DNA condensation and how this could be used for gene therapy.Citation73 Polylysine covalently attached to the glycoprotein asialoorosomucoid was shown to enhance gene expression in the liver of a mouse model, with the enhancement being up to 50-fold higher when compared to polylysine alone. By combing AFM with confocal microscopy, scholars evaluated bioeffects of therapeutic ultrasound-mediated DNA on cells and nuclei,Citation74 with the therapeutic ultrasound proving to be useful for increasing transfection efficiency. Zhu et al mapped the nucleotide-binding site of uncoupling protein 1 with the help of AFM and using both topographic and recognition modes of the mitochondrial membrane to elucidate the mechanism of the nucleotide binding, and thus helped explain how reactive oxygen-species generation can be used for obesity-, inflammation-, neurodegeneration-, or ischemia-related therapies.Citation75 Kim et al investigated fundamental structural units in the Escherichia coli nucleoid using AFM,Citation76 and showed that fibrous structures of 40 nm and 80 nm were found in the different growth phases of the bacteria.

Lastly, preliminary research combining AFM and immunolabeling has already demonstrated that cytoskeletal structures can be probed by combining AFM with stimulated emission depletion (STED)Citation77,Citation78 () and/or stochastic optical reconstruction microscopy,Citation79 all to ensure high resolution.Citation80 Research has also shown that by combining AFM with immunofluorescence, both HDAC6 and its role in mediating vimentin’s reorganization can be elucidated. Vimentin is known to modify the cytoskeleton structure and thus influence and regulate cellular mechanics.Citation81 Specifically, the authors showed that the organization of vimentin fibers in the oncogene-expressing cells was different than in the controls, with STED revealing a more entangled structure and an increase in cellular stiffness relative to controls. Such ultrahigh-resolution analysis also showed increased cellular disorganization, consistent with the possibility that oncogenes induce an HDAC6-mediated collapse of the cytoskeletal network to contribute to changes in cellular stiffness and ultimately in cell invasion that could accompany epithelial-to-mesenchymal transition. Further, Harke et al demonstrated that by combining AFM and STED, a novel advanced nanoscopic tool can be created to allow for fluorescence, topographical, and elastic-modulus analysis of biosamples.Citation82 Other studies also revealed cytoskeleton structures (such as actin filaments and microtubules) of living astrocytes in neural cells by coupling AFM with confocal microscopy. The results showed that the cytoskeleton fibers had high elastic moduli and demonstrated AFM’s great potential to differentiate the roles of the cytoskeleton in neurodegeneration.Citation83 Cell-wall elongation and cytoskeleton changes have also been observed for Gram-negative bacteria using superresolution-fluorescence microscopy combining AFM and stochastic optical reconstruction microscopy.Citation84

Figure 10 Live monitoring with stimulated emission-depletion atomic force microscopy of a microtubule-related surgery at nanoscale level.

Notes: The arrow indicates that a single microtubule could be cut in a precise and controlled place, thus emphasizing the benefits of combining the two techniques. Reprinted from Chacko JV, Harke B, Canale C, Diaspro A. Cellular level nanomanipulation using atomic force microscope aided with superresolution imaging. J Biomed Opt. 2014;19:105003. Copyright 2014.Citation78

Figure 10 Live monitoring with stimulated emission-depletion atomic force microscopy of a microtubule-related surgery at nanoscale level.Notes: The arrow indicates that a single microtubule could be cut in a precise and controlled place, thus emphasizing the benefits of combining the two techniques. Reprinted from Chacko JV, Harke B, Canale C, Diaspro A. Cellular level nanomanipulation using atomic force microscope aided with superresolution imaging. J Biomed Opt. 2014;19:105003. Copyright 2014.Citation78

While this information provides evidence that AFM integration with current diagnosis assays might become a useful tool for clinicians, concerns regarding this technique’s limited ability to allow for automation and real-time cell-transformation analysis and evaluation, all in the natural viscoelastic conditions of both the cell and its ECM, need to be addressed. In particular, with stiffness being a function of the time scale of the measurement,Citation85 and knowing that well-controlled and tightly regulated balances of forces are required to help maintain homeostasis in a cell or a community of cells,Citation86 and with the ECM controlling survival,Citation87 cellular development,Citation88 migration,Citation89 proliferation,Citation90 shape,Citation91 and ultimately cellular fate and function, quantifying heterogeneity at the level of a single cell or a community of cells makes analysis even more challenging.

As such, the next generation of AFM-based technologies to be considered for the identification of cell transformation should allow for possible differentiation of the different tumor elements, as well as the capability to assess how such elements can work together synergistically to contribute to or define tumor heterogeneity. For instance, such a “clinician-friendly” tool should allow for parallel tests to be performed in individual labs on chip “rooms”, in which immobilized cell samples could permit evaluation of levels of protein expressions, changes in morphology, or changes in cellular elasticity (). Moreover, an ideal instrument should also allow for real-time monitoring of cellular physiological changes in a nondestructive manner and in a fast and reliable way by combining such physiological changes with the morphological and biochemical cues in a manner that could be easily interpreted by the user, ie, the clinician. Without the complexity of the biology, the analysis of mechanical cues, while perhaps providing behavioral insights, cannot provide a time-dependent cell-transformation analysis nor can it allow for evaluation of heterogeneity based on the individual changes in the cell cycle.

Figure 11 Schematic representation of a real-time user-friendly, nondestructive atomic force microscopy-based monitoring technique to be used in clinical settings.

Notes: The different reservoirs on the chip design allow for sample analysis and characteristics associated with morphological, biophysical, and biochemical changes.

Figure 11 Schematic representation of a real-time user-friendly, nondestructive atomic force microscopy-based monitoring technique to be used in clinical settings.Notes: The different reservoirs on the chip design allow for sample analysis and characteristics associated with morphological, biophysical, and biochemical changes.

Acknowledgments

This work was supported by National Science Foundation (NSF) grants 1300757 and 1434503.

Disclosure

The authors report no conflicts of interest in this work.

References

  • MoWChenJPatelACXCR4/CXCL12 mediate autocrine cell-cycle progression in NF1-associated malignant peripheral nerve sheath tumorsCell20131521077109023434321
  • ChafferCLWeinbergRAA perspective on cancer cell metastasisScience20113311559156421436443
  • GordonDJResioBPellmanDCauses and consequences of aneuploidy in cancerNat Rev Genet20121318920322269907
  • VermeulenKVan BockstaeleDRBernemanZNThe cell cycle: a review of regulation, deregulation and therapeutic targets in cancerCell Prolif20033613114912814430
  • KwonOJZhangLIttmannMMXinLProstatic inflammation enhances basal-to-luminal differentiation and accelerates initiation of prostate cancer with a basal cell originProc Natl Acad Sci U S A2014111E592E60024367088
  • MayankAKSharmaSDeshwalRKLalSKLIMD1 antagonizes E2F1 activity and cell cycle progression by enhancing Rb function in cancer cellsCell Biol Int20143880981724523249
  • AlsiaryRBrüning-RichardsonABondJMorrisonEEWilkinsonNBellSMDeregulation of microcephalin and ASPM expression are correlated with epithelial ovarian cancer progressionPlos One20149e9700524819048
  • KouprinaNPavlicekACollinsNKThe microcephaly ASPM gene is expressed in proliferating tissues and encodes for a mitotic spindle proteinHum Mol Genet2005142155216515972725
  • SungHYChoiENLyuDParkAKJuWAhnJHAberrant hypomethylation-mediated AGR2 overexpression induces an aggressive phenotype in ovarian cancer cellsOncol Rep20143281582024920423
  • SalmansMLZhaoFAndersenBThe estrogen-regulated anterior gradient 2 (AGR2) protein in breast cancer: a potential drug target and biomarkerBreast Cancer Res20131520423635006
  • CairnsRAHarrisISMakTWRegulation of cancer cell metabolismNat Rev Cancer201111859521258394
  • GoldsteinASWitteONDoes the microenvironment influence the cell types of origin for prostate cancer?Genes Dev2013271539154423873937
  • AllenMJonesJLJekyll and Hyde: the role of the microenvironment on the progression of cancerJ Pathol201122316217621125673
  • BloomABZamanMHInfluence of the microenvironment on cell fate determination and migrationPhysiol Genomics20144630931424619520
  • EstrellaVChenTALloydMAcidity generated by the tumor microenvironment drives local invasionCancer Res2013731524153523288510
  • NelsonCMBissellMJModeling dynamic reciprocity: engineering three-dimensional culture models of breast architecture, function, and neoplastic transformationSemin Cancer Biol20051534235215963732
  • FullarADudasJOlahLRemodeling of extracellular matrix by normal and tumor-associated fibroblasts promotes cervical cancer progressionBMC Cancer20151525625885552
  • MalikRLelkesPICukiermanEBiomechanical and biochemical remodeling of stromal extracellular matrix in cancerTrends Biotechnol20153323023625708906
  • BiHXShiHBZhangTCuiGPRDM14 promotes the migration of human non-small cell lung cancer through extracellular matrix degradation in vitroChin Med J (Engl)201512837337725635434
  • ZhuJRLiangLJiaoYLiuLYEnhanced invasion of metastatic cancer cells via extracellular matrix interfacePlos One2015100118058
  • WebberJYeungVClaytonAExtracellular vesicles as modulators of the cancer microenvironmentSemin Cell Dev Biol201540273425662446
  • RakJGuhaAExtracellular vesicles – vehicles that spread cancer genesBioessays20123448949722442051
  • CandelarioKMSteindlerDAThe role of extracellular vesicles in the progression of neurodegenerative disease and cancerTrends Mol Med20142036837424835084
  • FearonERMolecular genetics of colorectal cancerAnnu Rev Pathol2011647950721090969
  • YeeDSTangYXLiXSThe Wnt inhibitory factor 1 restoration in prostate cancer cells was associated with reduced tumor growth, decreased capacity of cell migration and invasion and a reversal of epithelial to mesenchymal transitionMol Cancer2010916220573255
  • IyerSGaikwadRMSubba-RaoVWoodworthCDSokolovIAtomic force microscopy detects differences in the surface brush of normal and cancerous cellsNat Nanotechnol2009438939319498402
  • DischerDEJanmeyPWangYLTissue cells feel and respond to the stiffness of their substrateScience20053101139114316293750
  • BercoffJChaffaiSTanterMIn vivo breast tumor detection using transient elastographyUltrasound Med Biol2003291387139614597335
  • McKnightALKugelJLRossmanPJManducaAHartmannLCEhmanRLMR elastography of breast cancer: preliminary resultsAJR Am J Roentgenol20021781411141712034608
  • ShapiroIMChengAWFlytzanisNCAn EMT-driven alternative splicing program occurs in human breast cancer and modulates cellular phenotypePLoS Genet20117e100221821876675
  • WuYZMcEwenGDHariharSBakerSMDeWaldDBZhouAHBRMS1 expression alters the ultrastructural, biomechanical and biochemical properties of MDA-MB-435 human breast carcinoma cells: an AFM and Raman microspectroscopy studyCancer Lett2010293829120083343
  • AntonyakMALiBBoroughsLKCancer cell-derived microvesicles induce transformation by transferring tissue transglutaminase and fibronectin to recipient cellsProc Natl Acad Sci U S A20111084852485721368175
  • MinnAJKangYBSerganovaIDistinct organ-specific metastatic potential of individual breast cancer cells and primary tumorsJ Clin Invest2005115445515630443
  • ZhongJMaMJZhouJWeiDXYanZGHeDNTip-induced micropatterning of silk fibroin protein using in situ solution atomic force microscopyACS Appl Mater Interfaces2013573774623276203
  • MaMJZhongJLiWYComparison of four synthetic model peptides to understand the role of modular motifs in the self-assembly of silk fibroinSoft Matter201391132511333
  • MartinesEZhongJMuzardJSingle-molecule force spectroscopy of the aplysia cell adhesion molecule reveals two homophilic bondsBiophys J201210364965722947926
  • ZhongJFrom simple to complex: investigating the effects of lipid composition and phase on the membrane interactions of biomolecules using in situ atomic force microscopyIntegr Biol (Camb)2011363264421541434
  • RettlerEHoeppenerSSiguschBWSchubertUSMapping the mechanical properties of biomaterials on different length scales: depth-sensing indentation and AFM based nanoindentationJ Mater Chem B Mater Biol Med2013127892806
  • PlodinecMLoparicMMonnierCAThe nanomechanical signature of breast cancerNat Nanotechnol2012775776523085644
  • BakerELLuJYuDHBonnecazeRTZamanMHCancer cell stiffness: integrated roles of three-dimensional matrix stiffness and transforming potentialBiophys J2010992048205720923638
  • FriedlPWolfKTumour-cell invasion and migration: diversity and escape mechanismsNat Rev Cancer2003336237412724734
  • DeakinNOTurnerCEDistinct roles for paxillin and Hic-5 in regulating breast cancer cell morphology, invasion, and metastasisMol Biol Cell20112232734121148292
  • LekkaMPogodaKGostekJCancer cell recognition – mechanical phenotypeMicron2012431259126622436422
  • RebeloLMde SousaJSMendesJRadmacherMComparison of the viscoelastic properties of cells from different kidney cancer phenotypes measured with atomic force microscopyNanotechnology201324055122
  • CohenMKleinEGeigerBAddadiLOrganization and adhesive properties of the hyaluronan pericellular coat of chondrocytes and epithelial cellsBiophys J2003851996200512944312
  • JonesLMGardnerMJCatterallJBTurnerGAHyaluronic acid secreted by mesothelial cells: a natural barrier to ovarian cancer cell adhesionClin Exp Metastasis1995133733807641421
  • LiQSLeeGYOngCNLimCTAFM indentation study of breast cancer cellsBiochem Biophys Res Commun200837460961318656442
  • NikkhahMStroblJSSchmelzEMAgahMEvaluation of the influence of growth medium composition on cell elasticityJ Biomech20114476276621109247
  • LiQSLeeGYOngCNLimCTProbing the elasticity of breast cancer cells using AFMIFMBE Proc20092321222125
  • CoughlinMFBielenbergDRLenormandGCytoskeletal stiffness, friction, and fluidity of cancer cell lines with different metastatic potentialClin Exp Metastasis20133023725022961212
  • LiQSLimCTStructure-mechanical property changes in nucleus arising from breast cancerGefenACellular and Biomolecular Mechanics and MechanobiologyHeidelbergSpringer2011465475
  • WangJHYuMLiuLZhaoJLiangYCDongSAtomic force microscopy-based investigation on the elastic properties of oligodendrocytesMater Sci Forum2011694886890
  • PrabhuneMBelgeGDotzauerABullerdiekJRadmacherMComparison of mechanical properties of normal and malignant thyroid cellsMicron2012431267127222522060
  • LeporattiSVergaraDZacheoACytomechanical and topological investigation of MCF-7 cells by scanning force microscopyNanotechnology20092005510319417334
  • FariaECMaNGaziEMeasurement of elastic properties of prostate cancer cells using AFMAnalyst20081331498150018936825
  • LekkaMGilDPogodaKCancer cell detection in tissue sections using AFMArch Biochem Biophys201251815115622209753
  • CrossSEJinYSRaoJGimzewskiJKNanomechanical analysis of cells from cancer patientsNat Nanotechnol2007278078318654431
  • XuWWMezencevRKimBWangLJMcDonaldJSulchekTCell stiffness is a biomarker of the metastatic potential of ovarian cancer cellsPlos One201270046609
  • RotherJNodingHMeyIJanshoffAAtomic force microscopy-based microrheology reveals significant differences in the viscoelastic response between malign and benign cell linesOpen Biol2014414004624850913
  • BastatasLMartinez-MarinDMatthewsJAFM nano-mechanics and calcium dynamics of prostate cancer cells with distinct metastatic potentialBiochim Biophys Acta201218201111112022366469
  • CrossSEJinYSTondreJWongRRaoJGimzewskiJKAFM-based analysis of human metastatic cancer cellsNanotechnology20081938400321832563
  • PfreundschuhMMartinez-MartinDMulvihillEWegmannSMullerDJMultiparametric high-resolution imaging of native proteins by force-distance curve-based AFMNat Protoc201491113113024743419
  • Sanchez-AdamsJWiluszREGuilakFAtomic force microscopy reveals regional variations in the micromechanical properties of the pericellular and extracellular matrices of the meniscusJ Orthop Res2013311218122523568545
  • CerchiariAEGarbeJCJeeNYA strategy for tissue self-organization that is robust to cellular heterogeneity and plasticityProc Natl Acad Sci U S A20151122287229225633040
  • SchepelerTPageMEJensenKBHeterogeneity and plasticity of epidermal stem cellsDevelopment20141412559256724961797
  • HanBGChoJWChoYDExpression, purification and biochemical characterization of the N-terminal regions of human TIG3 and HRASLS3 proteinsProtein Expr Purif20107110310720100577
  • SotiriouCPusztaiLGene-expression signatures in breast cancerN Engl J Med200936079080019228622
  • ChenCPengJXiaHSQuantum-dot-based immunofluorescent imaging of HER2 and ER provides new insights into breast cancer heterogeneityNanotechnology20102109510120110584
  • FritzJKatopodisAGKolbingerFAnselmettiDForce-mediated kinetics of single P-selectin ligand complexes observed by atomic force microscopyProc Natl Acad Sci U S A19989512283122889770478
  • YanevaMKowalewskiTLieberMRInteraction of DNA-dependent protein kinase with DNA and with Ku: biochemical and atomic-force microscopy studiesEMBO J199716509851129305651
  • UchihashiTIinoRAndoTNojiHHigh-speed atomic force microscopy reveals rotary catalysis of rotorless F1-ATPaseScience201133375575821817054
  • BillingsleyDJBonassWACramptonNKirkhamJThomsonNHSingle-molecule studies of DNA transcription using atomic force microscopyPhys Biol2012902100122473059
  • HansmaHGGolanRHsiehWLolloCPMullen-LeyPKwohDDNA condensation for gene therapy as monitored by atomic force microscopyNucleic Acids Res199826248124879580703
  • Duvshani-EshetMBaruchLKesselmanEShimoniEMachlufMTherapeutic ultrasound-mediated DNA to cell and nucleus: bioeffects revealed by confocal and atomic force microscopyGene Ther20061316317216177822
  • ZhuRRupprechtAEbnerAMapping the nucleotide binding site of uncoupling protein 1 using atomic force microscopyJ Am Chem Soc20131353640364623414455
  • KimJYoshimuraSHHizumeKOhniwaRLIshihamaATakeyasuKFundamental structural units of the Escherichia coli nucleoid revealed by atomic force microscopyNucleic Acids Res2004321982199215060178
  • VicidominiGMoneronGHanKYSharper low-power STED nanoscopy by time gatingNat Methods2011857157321642963
  • ChackoJVHarkeBCanaleCDiasproACellular level nanomanipulation using atomic force microscope aided with superresolution imagingJ Biomed Opt20141910500325291208
  • ChackoJVZanacchiFCHarkeBLanzanoLCanaleCDiasproAInsight into hybrid nanoscopy techniques: STED AFM & STORM AFMBiophys J2014106396a
  • ChackoJVZanacchiFCDiasproAProbing cytoskeletal structures by coupling optical superresolution and AFM techniques for a correlative approachCytoskeleton20137072974024027190
  • RathjeLSNordgrenNPetterssonTOncogenes induce a vimentin filament collapse mediated by HDAC6 that is linked to cell stiffnessProc Natl Acad Sci U S A20141111515152024474778
  • HarkeBChakoJVHaschkeHCanaleCDiasproAA novel nanoscopic tool by combining AFM with STED microscopyOpt Nanoscopy201213
  • EfremovYMDzyubenkoEVBagrovDVMaksimovGVShramSIShaitanKVAtomic force microscopy study of the arrangement and mechanical properties of astrocytic cytoskeleton in growth mediumActa Naturae20113939922649699
  • TurnerRDHurdAFCadbyAHobbsJKFosterSJCell wall elongation mode in Gram-negative bacteria is determined by peptidoglycan architectureNat Commun20134149623422664
  • NaSTracheATrzeciakowskiJSunZMeiningerGAHumphreyJDTime-dependent changes in smooth muscle cell stiffness and focal adhesion area in response to cyclic equibiaxial stretchAnn Biomed Eng20083636938018214679
  • HumphreyJDVascular adaptation and mechanical homeostasis at tissue, cellular, and sub-cellular levelsCell Biochem Biophys200850537818209957
  • SmitsPLefebvreVSox5 and Sox6 are required for notochord extracellular matrix sheath formation, notochord cell survival and development of the nucleus pulposus of intervertebral discsDevelopment20031301135114812571105
  • LuPFTakaiKWeaverVMWerbZExtracellular matrix degradation and remodeling in development and diseaseCold Spring Harb Perspect Biol20113a00505821917992
  • WolfKte LindertMKrauseMPhysical limits of cell migration: control by ECM space and nuclear deformation and tuning by proteolysis and traction forceJ Cell Biol20132011069108423798731
  • Schlie-WolterSNgezahayoAChichkovBNThe selective role of ECM components on cell adhesion, morphology, proliferation and communication in vitroExp Cell Res20133191553156123588204
  • MaruthamuthuVSabassBSchwarzUSGardelMLCell-ECM traction force modulates endogenous tension at cell-cell contactsProc Natl Acad Sci U S A20111084708471321383129