Home Physical Sciences Special contribution of atomic force microscopy in cell death research
Article Open Access

Special contribution of atomic force microscopy in cell death research

  • Ning Li , Li Zhang , Ou Qiao , Xinyue Wang , Linyan Xu and Yanhua Gong EMAIL logo
Published/Copyright: February 15, 2024
Become an author with De Gruyter Brill

Abstract

Cell death is an important life activity in individual development. Changes in morphological and mechanical properties during cell death are crucial to identify the modes of cell death. However, due to technical limitations, little is known about these characteristics. The emergence of atomic force microscopy (AFM), a nanoscale research tool that integrates imaging and mechanical measurement functions, provides new insights into our understanding of cell death. Based on a brief introduction to the structure, principle, and working modes of AFM, this article elaborates on the contribution of AFM in cell death to detect morphological and mechanical properties, especially in apoptotic cells. Meanwhile, the potential of AFM in distinguishing different cell death modes and visualizing membrane pores (medicated by apoptosis and pyroptosis) is illustrated. In addition, this article states that using single-molecule force spectroscopy by AFM to study the mechanical and adhesive properties of cell death-related molecules. Finally, we discuss the challenges facing and further perspective of AFM.

Graphical abstract

Abbreviations

AFM

atomic force microscopy

AFM-SMFS

single-molecule force spectroscopy by AFM

PCD

programmed cell death

MP-AFM

multiparametric imaging AFM

HS-AFM

high-speed AFM

STM

scanning tunneling microscope

OM

optical microscopy

TEM

transmission electron microscopy

SEM

scanning electron microscopy

STS

staurosporine

IV

chrysin-organogermanium

Chry-Ge

chrysin-organogermanium complex

SNP

sodium nitroprusside

MOM

mitochondrial outer membrane

PFR

perforin

MAC

membrane attack complex

MOMP

mitochondrial outer membrane permeability

SLBs

supported lipid bilayers

SLMs

supported lipid membranes

MR-AFM

molecular recognition AFM

MF-AFM

multifrequency AFM

FluidFM

fluidic force microscopy

QI™

Quantitative Imaging™

SMLM

single-molecule localization microscopy

1 Introduction

Cell death is a ubiquitous biological phenomenon in all living organisms and is inseparable from organ development, aging, and the removal of damaged cells [1,2]. Research on the process of cell death has become a research hotspot in the fields of biology, medicine, and pharmacy. Based on functional differences, cell death can be classified into two types: programmed cell death (PCD) and non-PCD (necrosis) [3]. Currently known PCDs mainly include apoptosis, necroptosis, pyroptosis, autophagy, ferroptosis, and so on. The molecular mechanisms/signaling pathways of different death modes have been extensively explored, and meanwhile, morphological and mechanical properties are also particularly important for the characterization of different death modes. Especially in the field of oncology research, alterations in the elasticity and adhesion of individual cancer cells have been recognized as a property that promotes the spread of cancer cells [2,46]. The morphological characteristics and mechanical properties of classical cell death modes such as apoptosis have been initially explored, but the morphological and mechanical properties of new cell death modes have not been fully analyzed.

Atomic force microscopy (AFM) is a nanoscale research tool that integrates imaging and mechanical measurement functions [7]. Compared with other traditional microscopy techniques, AFM has many advantages. For example, in terms of sample imaging, the imaging environments are diverse, and samples can be imaged under various conditions, such as air, vacuum, or liquid, including dynamic observation of living cells [8,9]. In addition, the sample is not limited by its conductive properties. Moreover, AFM does not require staining, labeling, or fixing. The possibility to operate in liquid environments and at ambient temperature moved the application of AFM extends from the materials science to the biological science [10], which needs to overcome many difficulties, including the difference between material samples and biological samples. In general, the preparation of biological samples is more complex, such as the preparation of suspension cell samples, which needs to use diverse methods (such as electrostatic adsorption and microporous membrane filtration) to fix the cells to the substrate (such as Petri dishes, cover slides) [11]. During imaging of certain biological samples, such as membrane proteins protruding ∼1 nm from the membrane, the AFM contact mode can provide sub-nanometer resolution (≤1 nm) for imaging individual membrane proteins [810,12,13]. Meanwhile, various physical, chemical, and biological parameters can be characterized during imaging. In addition, as a multifunctional tool, AFM is one of the important tools to study the mechanical properties of cells, including the measurements of cell Young’s modulus, adhesion, and viscoelasticity [1416]. And AFM can measure the rupture forces between molecules or particles in the piconewton (pN) range, such as analyzing the interaction force of receptor–ligand [1720]. Over the past few decades, AFM has been successfully applied to the imaging analysis of various biological systems, including nucleic acids [21], proteins [22], cells [23,24], and tissues [23,25].

At the same time, recent research advances in AFM technology have strengthened its capabilities, such as multiparametric imaging AFM (MP-AFM) [26], and high-speed AFM (HS-AFM) [27], which enables simultaneous multiparametric imaging in a short time. The extraordinary ability of AFM makes it show a very broad contribution prospect in the field of cell death. It can clearly observe the morphological changes of different cell death methods, including changes in cell size, cell height, cell surface roughness, and the formation of cell membrane pores. In addition, AFM can measure dynamic changes in mechanical properties during cell death. Based on the previous research, this article briefly introduces the basic structure, imaging principle, and working modes of AFM and reviews its contribution in the field of cell death in detail, including the study of morphological and mechanical properties of different cell death modes (Graphical abstract, the curve in the figure is a schematic and does not represent real experimental data). At the end of this article, we also expound on the research status of AFM and discuss its current challenges and future development directions for the shortcomings of AFM.

2 The morphology and mechanisms of different cell death modes

There are multiple cell death modes, such as apoptosis, pyroptosis, necroptosis, and ferroptosis. Apoptosis was first described by Kerr et al. from a morphological point of view to describe the physiological death of cells and named it apoptosis [28]. The main morphological features of apoptotic cells are chromatin condensation, cell shrinkage, cell membrane blebbing, cell budding to form apoptotic bodies, etc. [2,28,29] (Table 1). After induction of apoptosis, BAX and BAK are activated and aggregated and inserted on the mitochondrial outer membrane (MOM), undergo conformational rearrangement, oligomerize to form pores, and release pro-apoptotic factors such as cytochrome C and SMAC/DIABLO, etc., thereby initiating the apoptotic program [3336]. Then in 2001, Cookson et al. first proposed the definition of pyroptosis, which is described as pro-inflammatory PCD [37]. The main morphological manifestations were DNA breakage, cell swelling, cell membrane pore formation, and release of cell contents (Table 1). During pyroptosis, gasdermin D (GSDMD) or GSDME is activated by caspase-1/4/5/11 or caspase-3 after cleavage, oligomerization and inserts into the cell membrane to form pyroptotic pores, leading to cell swelling and rupture [3840]. Afterward, in 2005, the necroptosis term was first proposed by Degterev et al. [41]. Its main features include cell membrane rupture, autophagosome formation, and significant organelle swelling. RIPK1, RIPK3, and MLKL are key molecules in the process of necroptosis. Researchers found that p-MLKL has the potential to create pores in the plasma membrane resulting in membrane rupture [42,43]. Ferroptosis as a novel cell death mode was first proposed in 2012 by Dixon et al. [44]. The morphological characteristics were cell membrane rupture, increased density, and decreased volume of the mitochondrial membrane [45] (Table 1).

Table 1

The morphological features of different cell death modes

Cell death modes Apoptosis Pyroptosis Necroptosis Ferroptosis
Morphological features Cell shrinkage increased; numerous smaller apoptotic bodies [30] Very swollen morphology and burst; pores form in the cell membrane [31] Cells do not shrink but detach and swell; pores form in the cell membrane [30] Circular protrusions formed on the membrane, about 1–5 μm [30,32]

3 Introduction to AFM structure, principle, and working modes

3.1 AFM structure and principle

AFM was invented by physicist Binnig et al. in 1986 [7]. The invention of AFM overcomes the shortcomings of STM in measuring nonconducting samples and expands the detection from conductors and semiconductors to insulators [7]. In addition, AFM has a wide range of applications compared to other microscopes, including working in liquid environments (PBS) as well as soft samples (living cells), combining topographic imaging with force spectroscopy and nanomechanics (Table 2). AFM mainly consists of the following systems: a laser system for laser generation, a head detection system consisting of a cantilever and a very sharp tip at its end, a laser detection system that receives laser reflection, and a feedback system that processes and transmits feedback signals to the piezoelectric scanner. The working principle of AFM is shown in Figure 1a. When the tip of the probe begin to scan the sample, the weak interaction force (mechanical contact force, van der Waals force, electrostatic force, etc.) between the probe tip and the sample causes the cantilever to swing. The deformation signal is converted into a photoelectric signal and amplified, and the signal of the interaction force between atoms can be obtained. Among them, the interaction force between the tip and the sample is related to the distance between them, which can be simply divided into attractive force and repulsive force, as shown in Figure 1b.

Table 2

Comparison of AFM and other high-resolution techniques

Characteristics AFM [10,46,47] Super-resolution microscopy (SLML) [4851] Transmission electron microscopy [46,52] Scanning electron microscopy [10,46]
Resolution ≤1 nm <10 nm 0.2 nm 1 nm
Sample preparation Sample on supported. Organic dyes or fluorescence labeling. Sample onto a grid. Freezing or critical point drying and metal spraying.
Advantages Imaging conditions are native; high signal-to-noise ratio; without staining, labeling, or fixing. Monitoring of dynamic cellular or biomolecular movements; high spatial and temporal resolution; visualization of subcellular structures. Imaging individual nanoscale objects in real space and reciprocal space. Imaging features of the sample surfaces.
Limitation Limited to surfaces. Imaging produces photobleaching and phototoxicity. Imaging in a vacuum environment; the specimen must be thin. Imaging in a vacuum environment.
Figure 1 
                  AFM. (a) Schematic diagram of AFM operation. (b) Force–distance curve by AFM. (c) Three working modes are commonly used in AFM.
Figure 1

AFM. (a) Schematic diagram of AFM operation. (b) Force–distance curve by AFM. (c) Three working modes are commonly used in AFM.

3.2 AFM working modes

AFM provides three commonly used working modes, namely contact mode, non-contact mode, and dynamic mode (originally called tapping or oscillation mode), as shown in Figure 1c. In the contact mode, the sample and the tip are always in contact (corresponding to segments 1–2 in Figure 1b). In this mode, high-resolution images can be obtained due to the close proximity of the sample to the tip [53]. But also because of this, too close distance may cause contamination and damage to the tip, and the friction force of the tip to the sample will also cause damage to the sample and affect the quality of imaging [54]. Therefore, when applying it to softer biological samples, it is necessary to adjust the force applied to the tip, generally avoiding more than 100 pN, as excessive force may lead to reversible or even irreversible deformation [5557]. In the non-contact mode, the microcantilever vibrates near the sample surface (corresponding to segments 3–4 in Figure 1b) [58]. In this mode, the tip is not in direct contact with the sample, which makes up for the problems of sample damage and tip contamination in the contact mode. However, compared with the contact mode, the non-contact mode has a lower resolution and is unsuitable for imaging in liquid environments. In the dynamic mode, the cantilever oscillates with a larger amplitude (greater than 20 nm) near the resonance frequency during the scanning process (corresponding to segments 1–4 in Figure 1b). This mode minimizes friction and shear forces between the tip and the sample while maintaining a resolution that is essentially the same as the contact mode. In summary, the dynamic mode is suitable for objects that are more flexible and more brittle and only weakly adsorb to supports, such as DNA [59,60], proteins [61], and fibrils of Tau [62].

4 Use AFM to detect the dynamic changes of morphological and mechanical properties of different cell death modes

Some caution should be taken into account to reduce the invasiveness degree toward the delicate biological cell samples during AFM technique. The first is sample preparation. In living cells, the position change of the sample caused by the lateral force applied by the AFM probe during scanning imaging should be avoided [63]. For adherent cells, because they can be naturally attached to the substrate (such as Petri dishes, cover slides), they can be directly imaged after adherent [64]. The substrate surface can also be covered with a layer of polylysine to increase the adhesion effect [65]. For suspension cells, appropriate fixation methods (such as electrostatic adsorption and microporous membrane filtration) are needed to adsorb them to the substrate and then image them [11]. The second is the choice of probe. Probe quality and probe parameters will directly affect the final result. Because of its small contact area with the cell, the pyramidal probe can capture the details of the geometric properties of the cell surface and is often used for cell surface topography scanning [66]. The micrometer-sized spherical probe is often used to measure the more holistic mechanical properties of cells because of their stable contact [66].

4.1 AFM exploring the morphological characteristics of apoptotic cells

Human understanding of cell death has gone through a long process. Prior to the advent of cell staining and microscopy, early studies of cell death largely focused on morphological observations with the naked eye [67,68]. So far, morphological observation is still one of the important basis for judging different cell death modes. And different death modes show different morphological characteristics. Currently, cell morphology is mainly observed by optical microscopy (OM), transmission electron microscopy (TEM), and scanning electron microscopy (SEM), but these methods all have certain limitations [6971] (Table 2). For example, conventional OM suffers from a resolution limit of 200 nm, making it impossible to reveal the nanostructure of cells [72,73]. Second, the essential advantage of AFM over TEM/SEM is the ability to measure the biophysical properties of biological samples under physiological conditions [74]. In recent years, with the development of microscopy technology, AFM has been gradually applied to the morphological observation of different cell death modes. According to the obtained 2D and 3D topological maps of cells, relevant information such as cell volume, cell height, cell membrane surface roughness, and cell membrane pore formation can be clearly collected, which provides a strong support for further understanding the morphological characteristics of specific cell death modes.

AFM has been used to observe the morphological characteristics of different cell death modes, but existing studies have mainly focused on apoptosis. Previous studies have shown that reduction in cell volume is a prerequisite for the early stages of apoptosis [7577]. In 2005, Hessler et al. used AFM to observe early morphological changes in human oral epidermoid carcinoma cells (KB cells) apoptosis induced by staurosporine (STS) [77]. The images were obtained using the contact mode by silicon nitride cantilevers MSCT-AUNM (cantilever C) at the spring constant of about 0.01 N m−1. AFM data showed a 50% reduction in total cell volume and a 32% reduction in total cell height, and the reduction in apoptotic cell volume preceded other key hallmarks of apoptosis, such as loss of mitochondrial membrane potential [77]. Researches also indicated that there was a significant change in cell volume during the initial stage of apoptosis [77]. Cells are generally in the range of an AFM scan, but even the same cell type can vary by up to a few micrometers. Therefore, there are many difficulties that need to be overcome in using changes in cell volume as a marker of early apoptosis. However, it is much more convenient to observe the surface characteristics of the membrane. Moreover, the variation of cell membrane surface roughness is an indication of early apoptosis. AFM morphological imaging allows the extraction of 3D topology-related data of cells, which enables analysis of cell membrane surface roughness. Therefore, to identify apoptosis at an early stage, in 2011, Wang et al. employed AFM to observe changes in the membrane surface roughness of hydrogen peroxide (H2O2)-induced apoptosis in the mouse macrophage cell line RAW264.7 [78]. Results showed that the mean roughness increased between 24 and 60 nm with increasing H2O2 concentration [78]. It is worth noting that, although under normal circumstances, with the occurrence of apoptosis, the surface roughness of the cell membrane shows an increasing trend. However, there are some exceptions. For example, when Cai et al. used AFM to observe the apoptotic K562 cells induced by peripheral lymphocytes. The UL20B cantilever whose length, width, and thickness are 115, 30, and 3.5 μm, respectively, was chosen in the contact mode. The oscillation frequency is 255 kHz and the force constant is 0.01 N m−1. And they found that after co-culturing these two type cells, K562 cells showed typical characteristics of apoptosis, but the cell surface roughness was significantly reduced [79]. Furthermore, the formation of cell membrane pores is one of the sources of cell surface roughness, and as the pores become deeper, the cell surface becomes rougher [80]. Therefore, to better describe the morphological characteristics of apoptosis, cell membrane pores are usually observed and analyzed. These studies further reveal morphological changes during apoptosis and deepen our understanding of cell death.

4.2 Advances in AFM measurement of mechanical properties during cell death

Likewise, cell mechanics can be used to describe the state of a cell, where certain changes in mechanical properties occur during cell death. AFM has been shown to be a powerful non-destructive nanotechnology that can be used to obtain dynamic processes related to cellular mechanics, such as elasticity and adhesion. Studies have shown that apoptosis may be associated with changes in cell elasticity [81]. In addition, the adhesion of cell membranes plays a very important role in cell physiology and pathological processes. Hu et al. used AFM measurement and calculated by Hertz model to obtain the Young’s moduli of lymphocytes in three states of resting, activated, and apoptotic, which were 11.2 ± 5.9, 19.7 ± 4.0, and 7.1 ± 4.1 kPa, respectively, indicating that the Young’s modulus of apoptotic cells was significantly reduced [82]. The high values of the Young’s modulus of elasticity reported in this article may be due to the limitations of experimental conditions, such as force profiles measured in air, where humidity, temperature, cell dehydration, or cell drying can have a dramatic effect on the results. For example, in 2014, Jin et al. showed the damaged effect of sodium nitroprusside (SNP) on cell surface adhesion (30% reduction) and elasticity (90% reduction) at the nano level in apoptotic chondrocytes [83]. In this study, the force spectrum experiment was performed at the force loading rate is 1.2 × 105 pN s−1. The mechanical properties of cells have gradually become an important characteristic to distinguish healthy cells from dead cells. Meanwhile, due to the extremely high mechanical resolution of AFM, AFM single-molecule force spectroscopy technology (AFM-SMFS) developed in AFM technology has gradually become an effective tool to measure the intra- and inter-cellular interactions of biological macromolecules at the single-cell level. Future AFM holds great potential for the mechanistic measurement of different cell death modes.

4.3 Use AFM to study the relationship between cell death and cytoskeleton

In the process of studying cell mechanics, the study of the cytoskeleton is an essential part. The cytoskeleton is important for maintaining cell mechanical properties [84], and cytoskeleton remodeling plays an important role in the process of apoptosis [30,85]. Importantly, it has been shown that changes in cellular elasticity are inextricably linked to the components of the cytoskeleton in addition to the intrinsic properties of the cell membrane [86]. Actin filaments and microtubules, two major components of the cytoskeleton, are severely damaged in structure, organization, and function, leading to cell death [74]. For example, Jin et al. studied the cytoskeleton of SNP-induced chondrocyte apoptosis, and after SNP treatment, the F-actin and α-tubulin cytoskeleton of chondrocytes reorganized and polymerized [83]. In addition, other researchers have continued to explore how the composition of the cytoskeleton affects cell elasticity. Rotsch and Radmacher found drugs that depolymerized actin filaments could significantly reduce the elastic modulus of cells, while drugs that stabilized microtubules had no significant effect on cell elasticity, suggesting that the actin network primarily determines the elastic properties of living cells [87]. To learn more about the effects of how microtubules and actin filaments affect cytoskeletal mechanical properties during apoptosis. Pelling et al. indicated that cell structure was highly dynamic during the early stages of apoptosis, with F-actin in actin filaments controlling the cell’s initial elastic response, while microtubules appear to control the cell’s viscous relaxation for extended periods [88]. Due to the limitations of AFM imaging, the most common way to monitor cytoskeletal changes is to combine AFM with confocal microscopy to study the dynamic changes of actin filaments and microtubules. However, the study by Henderson et al. showed that AFM could directly image actin filaments in living cells and observe the dynamic changes of actin filaments [89]. Although researchers have a preliminary understanding of the role of the cytoskeleton in maintaining the mechanical properties of cells, the relationship between the cytoskeleton and cell death remains to be further explored.

4.4 Potential of AFM to distinguish different cell death modes

With the development of AFM technology year by year, studies using AFM to observe various cell death modes have emerged in recent years [79,90,91]. Meeren et al. tested the differences in morphological and mechanical properties between three different PCD modes: intrinsic and extrinsic apoptosis, necroptosis, and ferroptosis in mouse tumor cell lines (L929sAhFas cell line) [30]. In morphological measurements, the resulting topographic image was obtained using the ATEC-CONT cantilever in a JPK QI® mode using the selected AFM nanowizard 4™ (JPK GmbH Instruments/Bruker). In cell elasticity measurements, all force curves were obtained in a contact mode using a colloidal probe containing a spherical tip of 5 μm diameter (CP-qp-SCONT-BSG, force constant 0.1 N m−1) and using a setpoint of 2 nN at 2 µm s−1. During intrinsic apoptosis, cells shrink and irregular structures are formed (Figure 2a, middle left). During extrinsic apoptosis, cell shrinkage was increased with concomitant apoptotic body generation (Figure 3a, middle right). During necroptosis, cells did not shrink but detached and swelled, and pores of different sizes were formed in the cell membrane (Figure 2b, middle). During ferroptosis, circular protrusions of 1–5 μm were observed to form on the cell membrane surface (Figure 2c, middle). In addition, the data showed that as the cell death process progressed, the cell surface roughness of these three cell death modes increased significantly. And the smallest increase is necroptosis. Overall, the AFM data clearly showed morphological characteristics in different cell death modes. Second, Meeren et al. also used AFM to measure the elasticity of multiple cells in these three PCDs. In the process of intrinsic apoptosis, Young’s modulus decreased rapidly 15 min after induction (Figure 2a, bottom left) and remained basically stable after the decrease. In contrast, for extrinsic apoptosis, Young’s modulus showed a trend of first increasing and then rapidly decreasing 60 min after induction (Figure 2a, bottom right). For both necroptosis and ferroptosis, Young’s modulus showed a continuous decreasing trend (Figure 2b and c, bottom). When comparing the Young’s modulus of these three cell death modes, it was found that the Young’s modulus of apoptosis (intrinsic and extrinsic apoptosis) decreased more rapidly compared with necroptosis and ferroptosis. Therefore, using AFM to study different cell death modes will help us to more clearly understand the dynamic changes of morphological and mechanical properties in the process of cell death.

Figure 2 
                  Schematic diagram of different cell death modes morphology (top), AFM imaging 3D topology (middle), and Young’s modulus (bottom). (a) Apoptosis, including intrinsic apoptosis and extrinsic apoptosis; (b) necroptosis; and (c) ferroptosis. The AFM imaging 3D topology (middle) and Young’s modulus (bottom) are reprinted, with permission, from [30].
Figure 2

Schematic diagram of different cell death modes morphology (top), AFM imaging 3D topology (middle), and Young’s modulus (bottom). (a) Apoptosis, including intrinsic apoptosis and extrinsic apoptosis; (b) necroptosis; and (c) ferroptosis. The AFM imaging 3D topology (middle) and Young’s modulus (bottom) are reprinted, with permission, from [30].

Figure 3 
                  MAC, BAX, BAK, and perforin pore morphological characteristics detected by AFM scanning. AFM imaging 3D topography of the MAC, BAK, BAX, and perforin pore (top of each image). The MAC, BAX, BAK, and perforin molecules around the pore protrude around the membrane plane, as confirmed by the height cross-sections below each image (corresponding to the gray dashed line in the AFM image) (bottom of each image). (a) Open access source, under Creative Commons license, from [92]. (b) Reprinted, with permission, from [93]. (c) Open access source, under Creative Commons license, from [94]. (d) Reprinted, with permission, from [95].
Figure 3

MAC, BAX, BAK, and perforin pore morphological characteristics detected by AFM scanning. AFM imaging 3D topography of the MAC, BAK, BAX, and perforin pore (top of each image). The MAC, BAX, BAK, and perforin molecules around the pore protrude around the membrane plane, as confirmed by the height cross-sections below each image (corresponding to the gray dashed line in the AFM image) (bottom of each image). (a) Open access source, under Creative Commons license, from [92]. (b) Reprinted, with permission, from [93]. (c) Open access source, under Creative Commons license, from [94]. (d) Reprinted, with permission, from [95].

5 AFM visualizes membrane pores in different cell death

A membrane pore can be defined as any local membrane perturbation that allows passive flow of molecules [96]. Different types of pores formation in the membrane may lead to different types of cell death [31]. For example, after induction of apoptosis, BAX and BAK will aggregate into pores in the mitochondrial membrane, release pro-apoptotic factors, and start the apoptosis process [34]. In addition, during apoptosis, perforin (PFR) released by T/NK cells can also form pores in the cell membrane [97]. Similarly, the gasdermin family (such as GSDMD or GSDME), the ultimate executor of pyroptosis, induces the occurrence of pyroptosis by punching holes in the cell membrane [38]. MLKL is the only known effector molecule associated with necroptosis [98]. Available data suggest that p-MLKL has the potential to create pores in the plasma membrane resulting in membrane rupture [42,43]. Unfortunately, we are not yet able to understand the structure and topology of MLKL in the cell membrane [99]. In addition, the membrane attack complex (MAC), a terminal pathway common to the three complement activation pathways, can also be assembled by soluble monomeric proteins and form killer transmembrane pores that mediate cell death [100]. AFM has successfully imaged the morphology of membrane attack complex structures on the cell membrane (Figure 3a) [92,101]. Similarly, in the past experiments, SEM, TEM, and other microscopes were mostly used to observe the formed membrane pores. However, because of the harsh imaging environment, it is usually difficult to observe clear membrane pore images and the shape of the formed oligomers [102104]. In contrast, AFM, as a microscope that allows the acquisition of native membrane images at sub-molecular resolution, enables a clearer observation of the dynamic assembly process of transmembrane pores [105]. Therefore, we broadly review relevant studies on the dynamic visualization of membrane pore formation mediated by various molecules by AFM. These studies reveal real-time morphology during pore formation and greatly advance our understanding of pore formation-mediated cell death [106108].

5.1 AFM visualization of BAX/BAK and PFR-mediated apoptosis pore formation

BAX and BAK are pro-apoptotic members of the BCL-2 family required for mitochondrial outer membrane permeability (MOMP) and play a key role in apoptosis [33]. Under normal conditions, BAX and BAK are present in healthy cells in inactive forms. After receiving an apoptotic signal, they are activated and oligomerically inserted into the MOM, which is accompanied by an increase in MOMP, and then BAX and BAK form giant pores [33,34,90]. However, the mechanism of how BAX and BAK aggregate and form apoptotic pores is still not fully understood. A recent study showed that AFM analysis of supported lipid bilayers (SLBs) prepared from proteoliposomes containing activated BAX monomers revealed that BAX formed linear, arc-shaped, and ring-shaped in SLBs of varying sizes and shapes. In other words, AFM can observe the formation of membrane pores associated with this protein (Figure 3b), which are not uniform in size and shape but are generally circular with diameters between 24 and 176 nm [93]. In the same way, Cosentino et al. observed the structure formed by BAK in SLBs and compared it with the pores formed by BAX. BAK also formed straight lines, arcs, and ring structures in SLBs (Figure 3c), but the pores formed by BAK were smaller and more uniform, and the rings had an average pore radius of about 8.12 nm [94].

As a pore-forming protein, PFR can form oligomeric pores in target cell membranes, which allow the entry of pro-apoptotic granzymes, thereby causing apoptosis of target cells [109,110]. Leung et al. visualized the PFR nanopore assembly in real time, where PFR first forms loosely but irreversibly bound, short prepore oligomers on target cell membranes. These short oligomers, after insertion into the membrane, recruit additional prepore oligomers, facilitating further assembly to form larger arc- and ring-shaped transmembrane pores (Figure 3d) [95].

5.2 AFM visualization of gasdermin family protein-mediated pyroptosis pore formation

Gasdermin family proteins, as the final executors of pyroptosis, consist of six members in humans, namely GSDMA, GSDMB, GSDMC, GSDMD, GSDME (DFNA5), and PJVK (DFNB59) [111,112]. Among them, GSDMD, as one of the earliest discovered, most widely studied, and most in-depth members of the gasdermin family, can be activated and cleaved by caspase 1/4/5/11 to form N-GSDMD and C-GSDMD. N-GSDMD acts directly on the cell membrane, forming pores in the membrane and triggering pyroptosis [111113]. However, how the N-GSDMD domain assembles the pores and the mechanism of penetrating the plasma membrane remains poorly understood. To address this issue, Mulvihil et al. applied high-resolution and time-lapse AFM to directly image the pore-forming process of N-GSDMD on supported lipid membranes (SLMs) [91]. Results showed that N-GSDMD released from GSDMD cleavage by caspase-1 can assemble into slit-, ring- and few arc-shaped oligomers (Figure 4a). And each oligomer structure occasionally forms transmembrane pores penetrating the SLMs. The structure height of slit- and ring-shaped oligomers protruding from the SLM is about 3.6 nm, indicating that they are almost completely inserted into the membrane. The analytical data also showed that the diameters of the ring-shaped oligomers were widely distributed between 13.5 and 33.5 nm, with an average value of approximately 22.6 nm [91]. These findings are consistent with the study by Sborgi et al. that N-GSDMD binds to lipid membranes and forms arc-, slit-, and ring-shaped oligomers, and the formed arcs and slits may fuse into ring-shaped N-GSDMD oligomers of variable diameter with an average value of 21 nm [103]. On the basis of AFM imaging, it can be speculated that the pathway for N-GSDMD to form pores is roughly as shown in Figure 4b. Furthermore, it is necessary to explore whether the pore-forming activity of GSDMD-N is also present in other gasdermin family members. Mari et al. characterized the pore-forming activity of mouse GSDMA3 by high-resolution time-lapse AFM [102]. It was found that GSDMA3 oligomers assemble on the SLMs and remain in a mobile or attached state. And once inserted into the membrane, it oligomerizes to form arc-, slit-, and ring-shaped oligomers, each of which also forms transmembrane pores, the pathway of N-GSDMA3 to form pores is shown in Figure 4b [102]. In addition, Liu et al. visualized the pore-forming process of GSDME on the cell membrane by AFM [31]. Overall, these studies collectively suggest that the gasdermin family proteins are the direct and ultimate executor of pyroptosis. The high-resolution information of the pyroptotic pores shows the unique value of AFM in the study of pyroptosis.

Figure 4 
                  GSDMD/A3 pore formation diagram. (a) AFM images of arc-, slit-, and ring-formed by N-GSDMD/A3 and (b) model of N-GSDMD/A3 oligomerization and pore formation. (a) (Top) Reprinted, with permission, from [91] and (a) (bottom) open access source, under Creative Commons license, from [102].
Figure 4

GSDMD/A3 pore formation diagram. (a) AFM images of arc-, slit-, and ring-formed by N-GSDMD/A3 and (b) model of N-GSDMD/A3 oligomerization and pore formation. (a) (Top) Reprinted, with permission, from [91] and (a) (bottom) open access source, under Creative Commons license, from [102].

5.3 Problems with AFM visualization of membrane pores

To date, AFM has made great progress in visualizing membrane pore formation, but most studies have imaged pores on artificial liposomes or SLMs [91,93,103], and only a few studies have imaged membrane pores on real living cells [31]. This could be due to the strict requirements of high-resolution imaging (nanoscale) and sample preparation problems, imaging in real cell membrane pores is difficult and complicated [31]. Because cell membranes are mainly composed of membrane proteins and phospholipids. Artificial lipid bilayers model (artificial membranes) can simulate cell membranes and keep their function intact [114]. Therefore, the artificial membranes are good alternative to cell membranes. It provides a lot of information about the morphology and properties of a phospholipid bilayer. However, there are still some limitations in the use of artificial liposomes and SLMs for pore visualization, including the lack of a membrane protein-intrinsic cytoskeleton, leading to possible discrepancies among pores in liposomes and SLMs and the actual cell membrane, generating possible errors in observing the dynamics of pore formation [31,115]. For example, related studies have shown that in real cells, PFR-induced pore size is about 200 nm, much larger than that detected from SLMs (about 10–20 nm) [95,115]. In addition, the size of pores formed by GSDMD proteins in real cell membranes and SLMs is also inconsistent [31,91,103]. Therefore, the results obtained using artificial liposomes and SLMs as research subjects may not reflect the relationship between cell membrane pore formation and cell death. To make a better connection between the formation of membrane pores and cell death, the study of real cell membrane pore formation using AFM should be accelerated. However, there is another problem with the visualization of membrane pores by AFM. Although studies are comparing the membrane pores depth with the cytoplasmic membrane thickness to confirm that the membrane pores imaged by AFM are real cell membrane pores and not from membrane depression or folds [31]. We still do not know how to exclude that only hollows were formed as the AFM tip cannot probe the inner part of the cell nor the underneath part of a supported lipid layer.

6 Study of cell death-related molecules using AFM-SMFS

The reasons for different cell death modes are not the same, it has been described above that the changes in morphological and mechanical properties of different cell death modes induced by different substances can be directly detected by AFM. However, as with other imaging techniques, we cannot obtain quantitative information on intermolecular or intramolecular interactions from AFM images, which also leads us to be unable to fully determine that cell death is induced by a specific substance. And the single-molecule force spectroscopy technique of atomic force microscopy (AFM-SMFS) is a good solution to this problem [116]. Since this technique requires tip-sample interaction, we can achieve this by functionalizing the AFM tip with particular chemical groups/ligands/antibodies. The main methods for attaching specific molecules to the AFM tip include physical adsorption, specific interaction, and chemical coupling (silanization, covalent coupling). In the experimental process, according to the nature of the sample, different connection methods can be used [117119]. After functionalization, AFM-SMFS allows the measurement of the adhesion and mechanical strength of specific bonds formed between the tip and individual sample molecules. Therefore, AFM can not only be used for imaging but has also become an indispensable measurement tool for studying biological macromolecular interactions.

The rapid development of AFM-SMFS has made it successfully applied to the analysis of single-molecule mechanical properties and intermolecular interactions, including DNA melting and dynamic structural changes [120], the dissociation mechanisms of DNA duplex [121], protein folding and unfolding [122], and interactions between biological macromolecules (protein–protein interaction, protein–nucleic acid interaction, protein–ligand interaction, etc.) [123125]. However, past studies have typically used only one biomolecule to functionalize the AFM tip when applying single-molecule force spectroscopy, but Pfreundschuh et al. further developed this technique [126]. Pfreundschuh et al. functionalized the AFM tip with two different ligands to map the two binding sites of human G protein-coupled receptors [126]. It can be seen that AFM-SMFS has great prospects in the field of biological research. Therefore, in recent years, AFM-SMFS has also been gradually applied in the field of cell death. pf-80 is an anti-PFR monoclonal antibody that recognizes PFR epitopes without interfering with plasma membrane binding of PFR [109,127]. In studying PFR-induced apoptosis in bulk tumor cells [128] and tumor-repopulating cells (TRC, low flexibility prevents membrane pore formation caused by PFR released by cytotoxic T cells), they attached pf-80 to the AFM tip and examined the adhesion between pf-80 and PFR (Figure 5a, left) [129]. The results showed that in bulk tumor cells, the adhesion between pf-80 and PFR was very high. But in TRC cells, the adhesion between pf-80 and PFR was very low, which may be due to the decreased expression of MYH9 (a motor protein that interacts with PFR) in TRC, which has the function of cross-linking and hardening F-actin. Its reduced expression may lead to a decrease in the interaction force between stable pf-80 and PFR, which ultimately leads to less adhesion between pf-80 and PFR in TRC cells [129]. Epidermal growth factor receptor (EGFR) is a cell transmembrane protein whose overexpression is common in many cancers, which can be activated by binding to epidermal growth factor (EGF). In other words, low expression of EGFR can induce tumor cell death. Resveratrol, as an anti-tumor drug, can inhibit the activity of EGFR, thereby inducing tumor cell death [86,130,131]. Zhang et al. used EGF-functionalized AFM probes to study the cell surface EGFR expression changes after treatment with different resveratrol concentrations (Figure 5a, right) [86]. The results showed that there was no significant change in the unbinding force of the low-dose resveratrol-treated group (up to 20 μg ml−1) compared with the untreated group. However, when cells were treated with 40 μg ml−1 resveratrol, the unbinding force was significantly reduced. Furthermore, they also demonstrated that when EGF (100 mg ml−1) is added gently to untreated MCF-7 cells, the interaction forces between EGF and EGFR were specifically blocked (Figure 5b) [86]. Thus, can we apply a similar idea to other cell death processes (Gasdermin family-mediated pyroptosis, MLKL protein-mediated necroptosis) using single-molecule force spectroscopy? This will help us to understand the dynamic changes and mechanisms of single molecules in the process of inducing cell death.

Figure 5 
               The specific recognition and detection of the molecule related to cell death by single-molecule force spectroscopy technique of atomic force microscopy (AFM-SMFS). (a) Schematic diagram of measuring the interaction between perforin and its monoclonal antibody pf-80, epidermal growth factor (EGF) and its receptor EGFR on the cell membrane by AFM-SMFS technology. (b) Typical force curve of EGF and EGFR interaction with specific unbinding peaks on cancer cells.
Figure 5

The specific recognition and detection of the molecule related to cell death by single-molecule force spectroscopy technique of atomic force microscopy (AFM-SMFS). (a) Schematic diagram of measuring the interaction between perforin and its monoclonal antibody pf-80, epidermal growth factor (EGF) and its receptor EGFR on the cell membrane by AFM-SMFS technology. (b) Typical force curve of EGF and EGFR interaction with specific unbinding peaks on cancer cells.

7 Discussion and further perspective

Clearly, AFM has emerged as a powerful imaging tool capable of imaging biological systems at high resolution (nanoscale). AFM topography can be used to characterize the morphological characteristics of different cell death modes. Analysis of cell biomechanical properties can detect the early onset of cell death and the dynamic changes in mechanical properties during death, thereby distinguishing different cell death modes [30]. Notably, AFM is currently the only force measurement technique that can map the nanoscale lateral distribution of individual molecular recognition sites on biological surfaces, which is beyond the reach of traditional biochemical methods [118]. And in recent years, great progress has been made in enhancing the function of AFM, including the development of different AFMs such as multiparametric AFM (MP-AFM) [26], molecular recognition AFM (MR-AFM) [132], multifrequency AFM (MF-AFM) [61], high-speed AFM (HS-AFM) [27], and fluidic force microscopy (FluidFM) [133]. At the same time, to better image and identify the complex structure of some biological systems, AFM can also be combined with other complementary techniques, including OM, fluorescence microscopy, confocal microscopy, super-resolution microscopy, etc. The most commonly used combination of AFM is fluorescence microscopy or confocal microscopy [10]. For example, in addressing the limitations of AFM imaging on the cell surface, a study combined AFM with confocal microscopy to directly penetrate the cell membrane and localize to the nucleus through the AFM tip to measure the elasticity of the nucleus [134]. Obviously, this has become a hot spot of current AFM research and a trend of future AFM development. The combination and development of these technologies and functions have greatly enhanced the roles of traditional AFM, but the application of AFM still faces some challenges.

First, how to further improve the spatiotemporal resolution of AFM. At present, conventional AFM is only used to take static snapshots or very slow dynamic processes, because it takes up several minutes to acquire an image of each sample, which is far more than the time required for normal cellular physiological activities. Although high-speed atomic force microscopes with high temporal resolution in the subsecond range have been developed in recent years [135]. However, due to the limitations of its high-speed imaging, this technique is currently only used for imaging small and relatively flat biological samples, such as single proteins and DNA molecules [136,137]. But, for some large biological systems, the spatial resolution of AFM imaging becomes another challenge. For example, the resolution of current AFM imaging of mammalian cells is limited to 50–100 nm, which makes it impossible to observe the individual components of cell-surface machinery [132]. The imaging resolution of microbial cells is slightly higher, but only about 10 nm [10]. So, next, we should consider how AFM can maintain its high temporal resolution and high spatial resolution at the same time when imaging different biological samples.

Second, AFM is low throughput and labor intensive. Current AFM experiments are manual rather than automated. Meanwhile, the traditional AFM takes several minutes or even longer to process one sample, and the next one can only be processed after one sample is processed, which makes the entire operation very time-consuming and labor-intensive. In addition, when measuring the mechanical property changes during cell death based on the force-distance curve, it is usually necessary to measure and analyze thousands of force curves, which will undoubtedly further increase the time and labor consumption. In response to this problem, Chopinet et al. provide a novel modality called quantitative imaging™ (QI™), which overcomes the data processing problems associated with acquiring thousands of force curves [138]. However, since this model has not yet been popularized, further researches are needed on related issues. Furthermore, the technical requirements for operators during AFM operation are very strict, especially for dynamic monitoring of living cells, which requires operators to have rich experience and good patience. Therefore, it is imminent to study the AFM of automated operation and simplified operation of data processing systems.

Finally, AFM does not need to be marked to be a double-edged sword. AFM does not need to mark the sample when observing the sample, which is one of the advantages of AFM. However, as the research system becomes more complex, this will turn into a weakness of AFM. For example, many proteins do not exhibit distinct morphologies, which are difficult to distinguish using AFM [8]. Although the existing technology has been able to functionalize the AFM probe, using the tip with specific molecular modification to specifically recognize the sample molecule. However, this technique is complicated and time-consuming in the molecular modification process, the experimental process is difficult to standardize, and the damage to the tip is unavoidable [139]. Therefore, further development of techniques that allow molecular-specific recognition by AFM is critical for imaging complex biological systems.

Generally speaking, the powerful functions exhibited by AFM make its research prospects in cell death immeasurable. However, as mentioned above, AFM still has many deficiencies in studying the physiological process of cells. Although some novel AFMs have been developed under the joint efforts of experts in multidisciplinary fields, the underlying problems remain unsolved. We expect a new and more powerful AFM that integrates multi-mode, multi-parameter, multi-frequency, and high-speed modes in the future. Coupled with the advancement of various complementary technologies, this will allow us to more clearly explore the unsolved problems of the cell death process and the entire field of biology.


# These authors contributed equally to this work and should be considered first co-authors.

tel: +8622 27893596

  1. Funding information: This work was supported by the National Natural Science Foundation of China (No. 82273998), the Ministry of Science and Technology of China (National Key R&D Program of China, 2021YFC3002203), and the Funding of Thinkingbiomed Biotechnology (Beijing) Co., Ltd (2022GKF-0667) to Prof. Yanhua Gong.

  2. Author contributions: Ning Li: conception and design of study, investigation, visualization, writing – review and editing, supervision. Li Zhang: investigation, visualization, writing – original draft. Qiao Ou: visualization. Xinyue Wang: visualization. Linyan Xu: visualization. Yanhua Gong: conception and design of study, writing – review and editing, supervision, funding acquisition. All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  3. Conflict of interest: The authors state no conflict of interest.

References

[1] Bertheloot D, Latz E, Franklin BS. Necroptosis, pyroptosis and apoptosis: An intricate game of cell death. Cell Mol Immunol. 2021;18(5):1106–21.10.1038/s41423-020-00630-3Search in Google Scholar PubMed PubMed Central

[2] Ketelut-Carneiro N, Fitzgerald KA. Apoptosis, pyroptosis, and necroptosis-oh my! The many ways a cell can die. J Mol Biol. 2022;434(4):167378.10.1016/j.jmb.2021.167378Search in Google Scholar PubMed

[3] Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res. 2019;29(5):347–64.10.1038/s41422-019-0164-5Search in Google Scholar PubMed PubMed Central

[4] Snyder AG, Oberst A. The antisocial network: Cross talk between cell death programs in host defense. Annu Rev Immunol. 2021;39:77–101.10.1146/annurev-immunol-112019-072301Search in Google Scholar PubMed PubMed Central

[5] Zemła J, Danilkiewicz J, Orzechowska B, Pabijan J, Seweryn S, Lekka M. Atomic force microscopy as a tool for assessing the cellular elasticity and adhesiveness to identify cancer cells and tissues. Semin Cell Dev Biol. 2018;73:115–24.10.1016/j.semcdb.2017.06.029Search in Google Scholar PubMed

[6] Nandi T, Ainavarapu SRK. Applications of atomic force microscopy in modern biology. Emerg Top Life Sci. 2021;5(1):103–11.10.1042/ETLS20200255Search in Google Scholar PubMed

[7] Binnig G, Quate CF, Gerber C. Atomic force microscope. Phys Rev Lett. 1986;56(9):930–3.10.1103/PhysRevLett.56.930Search in Google Scholar PubMed

[8] Li M, Liu L, Xi N, Wang Y, Dong Z, Xiao X, et al. Atomic force microscopy imaging of live mammalian cells. Sci China Life Sci. 2013;56(9):811–7.10.1007/s11427-013-4532-ySearch in Google Scholar PubMed

[9] Katan AJ, Dekker C. High-speed AFM reveals the dynamics of single biomolecules at the nanometer scale. Cell. 2011;147(5):979–82.10.1016/j.cell.2011.11.017Search in Google Scholar PubMed

[10] Dufrêne YF, Ando T, Garcia R, Alsteens D, Martinez-Martin D, Engel A, et al. Imaging modes of atomic force microscopy for application in molecular and cell biology. Nat Nanotechnol. 2017;12(4):295–307.10.1038/nnano.2017.45Search in Google Scholar PubMed

[11] Dufrêne YF. Atomic force microscopy and chemical force microscopy of microbial cells. Nat Protoc. 2008;3(7):1132–8.10.1038/nprot.2008.101Search in Google Scholar PubMed

[12] Wang J, Wan Z, Liu W, Li L, Ren L, Wang X, et al. Atomic force microscope study of tumor cell membranes following treatment with anti-cancer drugs. Biosens Bioelectron. 2009;25(4):721–7.10.1016/j.bios.2009.08.011Search in Google Scholar PubMed

[13] Simpson JD, Ray A, Koehler M, Mohammed D, Alsteens D. Atomic force microscopy applied to interrogate nanoscale cellular chemistry and supramolecular bond dynamics for biomedical applications. Chem Commun (Camb). 2022;58(33):5072–87.10.1039/D1CC07200ESearch in Google Scholar PubMed

[14] Cascione M, De Matteis V, Toma CC, Pellegrino P, Leporatti S, Rinaldi R. Morphomechanical and structural changes induced by ROCK inhibitor in breast cancer cells. Exp Cell Res. 2017;360(2):303–9.10.1016/j.yexcr.2017.09.020Search in Google Scholar PubMed

[15] Simon A, Durrieu MC. Strategies and results of atomic force microscopy in the study of cellular adhesion. Micron. 2006;37(1):1–13.10.1016/j.micron.2005.06.006Search in Google Scholar PubMed

[16] Li M, Liu L, Xi N, Wang Y. Atomic force microscopy studies on cellular elastic and viscoelastic properties. Sci China Life Sci. 2018;61(1):57–67.10.1007/s11427-016-9041-9Search in Google Scholar PubMed

[17] Wojcikiewicz EP, Zhang X, Moy VT. Force and compliance measurements on living cells using atomic force microscopy (AFM). Biol Proced Online. 2004;6:1–9.10.1251/bpo67Search in Google Scholar PubMed PubMed Central

[18] Merkel R, Nassoy P, Leung A, Ritchie K, Evans E. Energy landscapes of receptor-ligand bonds explored with dynamic force spectroscopy. Nature. 1999;397(6714):50–3.10.1038/16219Search in Google Scholar PubMed

[19] Lostao A, Lim K, Pallarés MC, Ptak A, Marcuello C. Recent advances in sensing the inter-biomolecular interactions at the nanoscale - A comprehensive review of AFM-based force spectroscopy. Int J Biol Macromol. 2023;238:124089.10.1016/j.ijbiomac.2023.124089Search in Google Scholar PubMed

[20] Marcuello C, de Miguel R, Lostao A. Molecular recognition of proteins through quantitative force maps at single molecule level. Biomolecules. 2022;12(4):594.10.3390/biom12040594Search in Google Scholar PubMed PubMed Central

[21] Hansma HG. Surface biology of DNA by atomic force microscopy. Annu Rev Phys Chem. 2001;52:71–92.10.1146/annurev.physchem.52.1.71Search in Google Scholar PubMed

[22] Sbrana F, Bongini L, Cappugi G, Fanelli D, Guarino A, Pazzagli L, et al. Atomic force microscopy images suggest aggregation mechanism in cerato-platanin. Eur Biophys J. 2007;36(7):727–32.10.1007/s00249-007-0159-xSearch in Google Scholar PubMed

[23] Ushiki T, Hitomi J, Ogura S, Umemoto T, Shigeno M. Atomic force microscopy in histology and cytology. Arch Histol Cytol. 1996;59(5):421–31.10.1679/aohc.59.421Search in Google Scholar PubMed

[24] Tilwani S, Gandhi K, Narayan S, Ainavarapu SRK, Dalal SN. Disruption of desmosome function leads to increased centrosome clustering in 14-3-3γ-knockout cells with supernumerary centrosomes. FEBS Lett. 2021;595(21):2675–90.10.1002/1873-3468.14204Search in Google Scholar PubMed

[25] Lewis NS, Chouhan G, Belapurkar V, Arora P, Ainavarapu SRK, Sonawane M. A new tension induction paradigm unravels tissue response and the importance of E-cadherin in the developing epidermis. Int J Dev Biol. 2020;64(4-5-6):343–52.10.1387/ijdb.190219msSearch in Google Scholar PubMed

[26] Heu C, Berquand A, Elie-Caille C, Nicod L. Glyphosate-induced stiffening of HaCaT keratinocytes, a peak force tapping study on living cells. J Struct Biol. 2012;178(1):1–7.10.1016/j.jsb.2012.02.007Search in Google Scholar PubMed

[27] Shibata M, Yamashita H, Uchihashi T, Kandori H, Ando T. High-speed atomic force microscopy shows dynamic molecular processes in photoactivated bacteriorhodopsin. Nat Nanotechnol. 2010;5(3):208–12.10.1038/nnano.2010.7Search in Google Scholar PubMed

[28] Kerr JF, Wyllie AH, Currie AR. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 1972;26(4):239–57.10.1038/bjc.1972.33Search in Google Scholar PubMed PubMed Central

[29] Afford S, Randhawa S. Apoptosis. Mol Pathol. 2000;53(2):55–63.10.1136/mp.53.2.55Search in Google Scholar PubMed PubMed Central

[30] Van der Meeren L, Verduijn J, Krysko DV, Skirtach AG. AFM analysis enables differentiation between apoptosis, necroptosis, and ferroptosis in murine cancer cells. iScience. 2020;23(12):101816.10.1016/j.isci.2020.101816Search in Google Scholar PubMed PubMed Central

[31] Liu Y, Zhang T, Zhou Y, Li J, Liang X, Zhou N, et al. Visualization of perforin/gasdermin/complement-formed pores in real cell membranes using atomic force microscopy. Cell Mol Immunol. 2019;16(6):611–20.10.1038/s41423-018-0165-1Search in Google Scholar PubMed PubMed Central

[32] Efimova I, Catanzaro E, Van der Meeren L, Turubanova VD, Hammad H, Mishchenko TA, et al. Vaccination with early ferroptotic cancer cells induces efficient antitumor immunity. J Immunother Cancer. 2020;8(2):e001369.10.1136/jitc-2020-001369Search in Google Scholar PubMed PubMed Central

[33] Edlich F, Banerjee S, Suzuki M, Cleland MM, Arnoult D, Wang C, et al. Bcl-x(L) retrotranslocates Bax from the mitochondria into the cytosol. Cell. 2011;145(1):104–16.10.1016/j.cell.2011.02.034Search in Google Scholar PubMed PubMed Central

[34] Cosentino K, García-Sáez AJ. Bax and bak pores: Are we closing the circle? Trends Cell Biol. 2017;27(4):266–75.10.1016/j.tcb.2016.11.004Search in Google Scholar PubMed PubMed Central

[35] Lovell JF, Billen LP, Bindner S, Shamas-Din A, Fradin C, Leber B, et al. Membrane binding by tBid initiates an ordered series of events culminating in membrane permeabilization by Bax. Cell. 2008;135(6):1074–84.10.1016/j.cell.2008.11.010Search in Google Scholar PubMed

[36] Deng J, Gutiérrez LG, Stoll G, Motiño O, Martins I, Núñez L, et al. Paradoxical implication of BAX/BAK in the persistence of tetraploid cells. Cell Death Dis. 2021;12(11):1039.10.1038/s41419-021-04321-3Search in Google Scholar PubMed PubMed Central

[37] Cookson BT, Brennan MA. Pro-inflammatory programmed cell death. Trends Microbiol. 2001;9(3):113–4.10.1016/S0966-842X(00)01936-3Search in Google Scholar

[38] Wang Y, Gao W, Shi X, Ding J, Liu W, He H, et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature. 2017;547(7661):99–103.10.1038/nature22393Search in Google Scholar PubMed

[39] Kayagaki N, Stowe IB, Lee BL, O’Rourke K, Anderson K, Warming S, et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature. 2015;526(7575):666–71.10.1038/nature15541Search in Google Scholar PubMed

[40] Li N, Wang Y, Wang X, Sun N, Gong YH. Pathway network of pyroptosis and its potential inhibitors in acute kidney injury. Pharmacol Res. 2022;175:106033.10.1016/j.phrs.2021.106033Search in Google Scholar PubMed

[41] Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N, et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol. 2005;1(2):112–9.10.1038/nchembio711Search in Google Scholar PubMed

[42] Zhang J, Yang Y, He W, Sun L. Necrosome core machinery: MLKL. Cell Mol Life Sci. 2016;73(11–12):2153–63.10.1007/s00018-016-2190-5Search in Google Scholar PubMed

[43] Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature. 2015;517(7534):311–20.10.1038/nature14191Search in Google Scholar PubMed

[44] Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–72.10.1016/j.cell.2012.03.042Search in Google Scholar PubMed PubMed Central

[45] Linkermann A, Bräsen JH, Darding M, Jin MK, Sanz AB, Heller JO, et al. Two independent pathways of regulated necrosis mediate ischemia-reperfusion injury. Proc Natl Acad Sci U S A. 2013;110(29):12024–9.10.1073/pnas.1305538110Search in Google Scholar PubMed PubMed Central

[46] Li M, Dang D, Xi N, Wang Y, Liu L. Nanoscale imaging and force probing of biomolecular systems using atomic force microscopy: from single molecules to living cells. Nanoscale. 2017;9(45):17643–66.10.1039/C7NR07023CSearch in Google Scholar

[47] Shan Y, Wang H. The structure and function of cell membranes examined by atomic force microscopy and single-molecule force spectroscopy. Chem Soc Rev. 2015;44(11):3617–38.10.1039/C4CS00508BSearch in Google Scholar PubMed

[48] Arizono M, Idziak A, Quici F, Nägerl UV. Getting sharper: the brain under the spotlight of super-resolution microscopy. Trends Cell Biol. 2023;33(2):148–61.10.1016/j.tcb.2022.06.011Search in Google Scholar PubMed

[49] Liu S, Hoess P, Ries J. Super-resolution microscopy for structural cell biology. Annu Rev Biophy. 2022;51:301–26.10.1146/annurev-biophys-102521-112912Search in Google Scholar PubMed

[50] Li W, Kaminski Schierle GS, Lei B, Liu Y, Kaminski CF. Fluorescent nanoparticles for super-resolution imaging. Chem Rev. 2022;122(15):12495–543.10.1021/acs.chemrev.2c00050Search in Google Scholar PubMed PubMed Central

[51] Bond C, Santiago-Ruiz AN, Tang Q, Lakadamyali M. Technological advances in super-resolution microscopy to study cellular processes. Mol Cell. 2022;82(2):315–32.10.1016/j.molcel.2021.12.022Search in Google Scholar PubMed PubMed Central

[52] Rizvi A, Mulvey JT, Carpenter BP, Talosig R, Patterson JP. A close look at molecular self-assembly with the transmission electron microscope. Chem Rev. 2021;121(22):14232–80.10.1021/acs.chemrev.1c00189Search in Google Scholar PubMed

[53] Starodubtseva M, Starodubtsev I, Yegorenkov N, Kuzhel N, Konstantinova E, Chizhik S. Physical-mechanical image of the cell surface on the base of AFM data in contact mode. IOP Conference Series: Materials Science and Engineering. IOP Publishing; 2017. p. 012016.10.1088/1757-899X/256/1/012016Search in Google Scholar

[54] Chang K-C, Chiang Y-W, Yang C-H, Liou J-W. Atomic force microscopy in biology and biomedicine. Tzu Chi Med J. 2012;24(4):162–9.10.1016/j.tcmj.2012.08.002Search in Google Scholar

[55] Müller DJ, Sass HJ, Müller SA, Büldt G, Engel A. Surface structures of native bacteriorhodopsin depend on the molecular packing arrangement in the membrane. J Mol Biol. 1999;285(5):1903–9.10.1006/jmbi.1998.2441Search in Google Scholar PubMed

[56] Engel A, Müller DJ. Observing single biomolecules at work with the atomic force microscope. Nat Struct Biol. 2000;7(9):715–8.10.1038/78929Search in Google Scholar PubMed

[57] Liang W, Shi H, Yang X, Wang J, Yang W, Zhang H, et al. Recent advances in AFM-based biological characterization and applications at multiple levels. Soft Matter. 2020;16(39):8962–84.10.1039/D0SM01106ASearch in Google Scholar PubMed

[58] Almonte L, Colchero J. True non-contact atomic force microscopy imaging of heterogeneous biological samples in liquids: topography and material contrast. Nanoscale. 2017;9(8):2903–15.10.1039/C6NR07967ASearch in Google Scholar

[59] Li M, Xi N, Liu L. Peak force tapping atomic force microscopy for advancing cell and molecular biology. Nanoscale. 2021;13(18):8358–75.10.1039/D1NR01303CSearch in Google Scholar PubMed

[60] Ido S, Kimura K, Oyabu N, Kobayashi K, Tsukada M, Matsushige K, et al. Beyond the helix pitch: direct visualization of native DNA in aqueous solution. ACS Nano. 2013;7(2):1817–22.10.1021/nn400071nSearch in Google Scholar PubMed

[61] Ido S, Kimiya H, Kobayashi K, Kominami H, Matsushige K, Yamada H. Immunoactive two-dimensional self-assembly of monoclonal antibodies in aqueous solution revealed by atomic force microscopy. Nat Mater. 2014;13(3):264–70.10.1038/nmat3847Search in Google Scholar PubMed

[62] Wegmann S, Jung YJ, Chinnathambi S, Mandelkow EM, Mandelkow E, Muller DJ. Human Tau isoforms assemble into ribbon-like fibrils that display polymorphic structure and stability. J Biol Chem. 2010;285(35):27302–13.10.1074/jbc.M110.145318Search in Google Scholar PubMed PubMed Central

[63] El Kirat K, Burton I, Dupres V, Dufrene YF. Sample preparation procedures for biological atomic force microscopy. J Microscopy. 2005;218(Pt 3):199–207.10.1111/j.1365-2818.2005.01480.xSearch in Google Scholar PubMed

[64] Matzke R, Jacobson K, Radmacher M. Direct, high-resolution measurement of furrow stiffening during division of adherent cells. Nat Cell Biol. 2001;3(6):607–10.10.1038/35078583Search in Google Scholar PubMed

[65] Puntheeranurak T, Wildling L, Gruber HJ, Kinne RK, Hinterdorfer P. Ligands on the string: Single-molecule AFM studies on the interaction of antibodies and substrates with the Na+ -glucose co-transporter SGLT1 in living cells. J Cell Sci. 2006;119(Pt 14):2960–7.10.1242/jcs.03035Search in Google Scholar PubMed

[66] Harris AR, Charras GT. Experimental validation of atomic force microscopy-based cell elasticity measurements. Nanotechnology. 2011;22(34):345102.10.1088/0957-4484/22/34/345102Search in Google Scholar PubMed

[67] Hockenbery D. Defining apoptosis. Am J Pathol. 1995;146(1):16–9.Search in Google Scholar

[68] Kane AB. Redefining cell death. Am J Pathol. 1995;146(1):1–2.10.1136/gut.36.1.146Search in Google Scholar PubMed PubMed Central

[69] Elkady AS. Scanning transmitted and reflected light microscopy: A novel microscopy for visualizing biomaterials at interfaces. Micron. 2007;38(8):848–53.10.1016/j.micron.2007.06.004Search in Google Scholar PubMed

[70] Chen H, Lü JH, Liang WQ, Huang YH, Zhang WJ, Zhang DB. Purification of the recombinant hepatitis B virus core antigen (rHBcAg) produced in the yeast Saccharomyces cerevisiae and comparative observation of its particles by transmission electron microscopy (TEM) and atomic force microscopy (AFM). Micron. 2004;35(5):311–8.10.1016/j.micron.2003.12.003Search in Google Scholar PubMed

[71] Taatjes DJ, Sobel BE, Budd RC. Morphological and cytochemical determination of cell death by apoptosis. Histochem Cell Biol. 2008;129(1):33–43.10.1007/s00418-007-0356-9Search in Google Scholar PubMed PubMed Central

[72] Ryan J, Gerhold AR, Boudreau V, Smith L, Maddox PS. Introduction to modern methods in light microscopy. Methods Mol Biol (Clifton, NJ). 2017;1563:1–15.10.1007/978-1-4939-6810-7_1Search in Google Scholar PubMed

[73] Nienhaus K, Nienhaus GU. Where do we stand with super-resolution optical microscopy? J Mol Biol. 2016;428(2 Pt A):308–22.10.1016/j.jmb.2015.12.020Search in Google Scholar PubMed

[74] Kubiak A, Zieliński T, Pabijan J, Lekka M. Nanomechanics in monitoring the effectiveness of drugs targeting the cancer cell cytoskeleton. Int J Mol Sci. 2020;21(22):8786.10.3390/ijms21228786Search in Google Scholar PubMed PubMed Central

[75] Schneider SW, Matzke R, Radmacher M, Oberleithner H. Shape and volume of living aldosterone-sensitive cells imaged with the atomic force microscope. Methods Mol Biol (Clifton, NJ). 2004;242:255–79.10.1385/1-59259-647-9:255Search in Google Scholar PubMed

[76] Braet F, Vermijlen D, Bossuyt V, De Zanger R, Wisse E. Early detection of cytotoxic events between hepatic natural killer cells and colon carcinoma cells as probed with the atomic force microscope. Ultramicroscopy. 2001;89(4):265–73.10.1016/S0304-3991(01)00092-4Search in Google Scholar

[77] Hessler JA, Budor A, Putchakayala K, Mecke A, Rieger D, Banaszak Holl MM, et al. Atomic force microscopy study of early morphological changes during apoptosis. Langmuir. 2005;21(20):9280–6.10.1021/la051837gSearch in Google Scholar PubMed

[78] Wang DC, Chen KY, Tsai CH, Chen GY, Chen CH. AFM membrane roughness as a probe to identify oxidative stress-induced cellular apoptosis. J Biomech. 2011;44(16):2790–4.10.1016/j.jbiomech.2011.08.021Search in Google Scholar PubMed

[79] Jin H, Zhao H, Liu L, Jiang J, Wang X, Ma S, et al. Apoptosis induction of K562 cells by lymphocytes: an AFM study. Scanning. 2013;35(1):7–11.10.1002/sca.21028Search in Google Scholar PubMed

[80] Kim KS, Cho CH, Park EK, Jung MH, Yoon KS, Park HK. AFM-detected apoptotic changes in morphology and biophysical property caused by paclitaxel in Ishikawa and HeLa cells. PLoS One. 2012;7(1):e30066.10.1371/journal.pone.0030066Search in Google Scholar PubMed PubMed Central

[81] Su X, Zhou H, Bao G, Wang J, Liu L, Zheng Q, et al. Nanomorphological and mechanical reconstruction of mesenchymal stem cells during early apoptosis detected by atomic force microscopy. Biol Open. 2020;9(3):bio048108.10.1242/bio.048108Search in Google Scholar PubMed PubMed Central

[82] Hu M, Wang J, Zhao H, Dong S, Cai J. Nanostructure and nanomechanics analysis of lymphocyte using AFM: From resting, activated to apoptosis. J Biomech. 2009;42(10):1513–9.10.1016/j.jbiomech.2009.03.051Search in Google Scholar PubMed

[83] Jin H, Liang Q, Chen T, Wang X. Resveratrol protects chondrocytes from apoptosis via altering the ultrastructural and biomechanical properties: an AFM study. PLoS One. 2014;9(3):e91611.10.1371/journal.pone.0091611Search in Google Scholar PubMed PubMed Central

[84] Mukherjee C, Bera M, Koti Ainavarapu SR, Sengupta K. Pulling the springs of a cell by single-molecule force spectroscopy. Emerg Top Life Sci. 2021;5(1):77–87.10.1042/ETLS20200254Search in Google Scholar PubMed

[85] Pelling AE, Veraitch FS, Pui-Kei Chu C, Nicholls BM, Hemsley AL, Mason C, et al. Mapping correlated membrane pulsations and fluctuations in human cells. J Mol Recognit. 2007;20(6):467–75.10.1002/jmr.832Search in Google Scholar PubMed

[86] Zhang L, Yang F, Cai JY, Yang PH, Liang ZH. In-situ detection of resveratrol inhibition effect on epidermal growth factor receptor of living MCF-7 cells by Atomic Force Microscopy. Biosens Bioelectron. 2014;56:271–7.10.1016/j.bios.2014.01.024Search in Google Scholar PubMed

[87] Rotsch C, Radmacher M. Drug-induced changes of cytoskeletal structure and mechanics in fibroblasts: An atomic force microscopy study. Biophys J. 2000;78(1):520–35.10.1016/S0006-3495(00)76614-8Search in Google Scholar PubMed PubMed Central

[88] Pelling AE, Veraitch FS, Chu CP, Mason C, Horton MA. Mechanical dynamics of single cells during early apoptosis. Cell Motil Cytoskelet. 2009;66(7):409–22.10.1002/cm.20391Search in Google Scholar PubMed

[89] Henderson E, Haydon PG, Sakaguchi DS. Actin filament dynamics in living glial cells imaged by atomic force microscopy. Science (New York, NY). 1992;257(5078):1944–6.10.1126/science.1411511Search in Google Scholar PubMed

[90] McArthur K, Whitehead LW, Heddleston JM, Li L, Padman BS, Oorschot V, et al. BAK/BAX macropores facilitate mitochondrial herniation and mtDNA efflux during apoptosis. Science (New York, NY). 2018;359(6378):eaao6047.10.1126/science.aao6047Search in Google Scholar PubMed

[91] Mulvihill E, Sborgi L, Mari SA, Pfreundschuh M, Hiller S, Müller DJ. Mechanism of membrane pore formation by human gasdermin-D. Embo J. 2018;37(14):e98321.10.15252/embj.201798321Search in Google Scholar PubMed PubMed Central

[92] Parsons ES, Stanley GJ, Pyne ALB, Hodel AW, Nievergelt AP, Menny A, et al. Single-molecule kinetics of pore assembly by the membrane attack complex. Nat Commun. 2019;10(1):2066.10.1038/s41467-019-10058-7Search in Google Scholar PubMed PubMed Central

[93] Salvador-Gallego R, Mund M, Cosentino K, Schneider J, Unsay J, Schraermeyer U, et al. Bax assembly into rings and arcs in apoptotic mitochondria is linked to membrane pores. Embo J. 2016;35(4):389–401.10.15252/embj.201593384Search in Google Scholar PubMed PubMed Central

[94] Cosentino K, Hertlein V, Jenner A, Dellmann T, Gojkovic M, Peña-Blanco A, et al. The interplay between BAX and BAK tunes apoptotic pore growth to control mitochondrial-DNA-mediated inflammation. Mol Cell. 2022;82(5):933–49.e9.10.1016/j.molcel.2022.01.008Search in Google Scholar PubMed PubMed Central

[95] Leung C, Hodel AW, Brennan AJ, Lukoyanova N, Tran S, House CM, et al. Real-time visualization of perforin nanopore assembly. Nat Nanotechnol. 2017;12(5):467–73.10.1038/nnano.2016.303Search in Google Scholar PubMed

[96] Schwarz G, Robert CH. Kinetics of pore-mediated release of marker molecules from liposomes or cells. Biophys Chem. 1992;42(3):291–6.10.1016/0301-4622(92)80021-VSearch in Google Scholar PubMed

[97] Baran K, Dunstone M, Chia J, Ciccone A, Browne KA, Clarke CJ, et al. The molecular basis for perforin oligomerization and transmembrane pore assembly. Immunity. 2009;30(5):684–95.10.1016/j.immuni.2009.03.016Search in Google Scholar PubMed

[98] Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25(3):486–541.10.1038/s41418-018-0102-ySearch in Google Scholar PubMed PubMed Central

[99] Flores-Romero H, Ros U, Garcia-Saez AJ. Pore formation in regulated cell death. Embo J. 2020;39(23):e105753.10.15252/embj.2020105753Search in Google Scholar PubMed PubMed Central

[100] Bayly-Jones C, Bubeck D, Dunstone MA. The mystery behind membrane insertion: a review of the complement membrane attack complex. Philos Trans R Soc Lond Ser B, Biol Sci. 2017;372(1726):20160221.10.1098/rstb.2016.0221Search in Google Scholar PubMed PubMed Central

[101] Morgan BP, Harris CL. Complement, a target for therapy in inflammatory and degenerative diseases. Nat Rev Drug Discov. 2015;14(12):857–77.10.1038/nrd4657Search in Google Scholar PubMed PubMed Central

[102] Mari SA, Pluhackova K, Pipercevic J, Leipner M, Hiller S, Engel A, et al. Gasdermin-A3 pore formation propagates along variable pathways. Nat Commun. 2022;13(1):2609.10.1038/s41467-022-30232-8Search in Google Scholar PubMed PubMed Central

[103] Sborgi L, Rühl S, Mulvihill E, Pipercevic J, Heilig R, Stahlberg H, et al. GSDMD membrane pore formation constitutes the mechanism of pyroptotic cell death. Embo J. 2016;35(16):1766–78.10.15252/embj.201694696Search in Google Scholar PubMed PubMed Central

[104] Leung C, Dudkina NV, Lukoyanova N, Hodel AW, Farabella I, Pandurangan AP, et al. Stepwise visualization of membrane pore formation by suilysin, a bacterial cholesterol-dependent cytolysin. Elife. 2014;3:e04247.10.7554/eLife.04247Search in Google Scholar PubMed PubMed Central

[105] Frederix PL, Bosshart PD, Engel A. Atomic force microscopy of biological membranes. Biophys J. 2009;96(2):329–38.10.1016/j.bpj.2008.09.046Search in Google Scholar PubMed PubMed Central

[106] Hodel AW, Hammond K, Hoogenboom BW. AFM imaging of pore forming proteins. Methods Enzymol. 2021;649:149–88.10.1016/bs.mie.2021.01.002Search in Google Scholar PubMed

[107] Jiao F, Ruan Y, Scheuring S. High-speed atomic force microscopy to study pore-forming proteins. Methods Enzymol. 2021;649:189–217.10.1016/bs.mie.2021.01.033Search in Google Scholar PubMed

[108] Unsay JD, García-Sáez AJ. AFM to Study Pore-Forming Proteins. Methods Mol Biol (Clifton, NJ). 2019;1886:191–202.10.1007/978-1-4939-8894-5_10Search in Google Scholar PubMed

[109] Metkar SS, Marchioretto M, Antonini V, Lunelli L, Wang B, Gilbert RJ, et al. Perforin oligomers form arcs in cellular membranes: a locus for intracellular delivery of granzymes. Cell Death Differ. 2015;22(1):74–85.10.1038/cdd.2014.110Search in Google Scholar PubMed PubMed Central

[110] Law RH, Lukoyanova N, Voskoboinik I, Caradoc-Davies TT, Baran K, Dunstone MA, et al. The structural basis for membrane binding and pore formation by lymphocyte perforin. Nature. 2010;468(7322):447–51.10.1038/nature09518Search in Google Scholar PubMed

[111] Xia S, Hollingsworth LRT, Wu H. Mechanism and regulation of gasdermin-mediated cell death. Cold Spring Harb Perspect Biol. 2020;12(3):a036400.10.1101/cshperspect.a036400Search in Google Scholar PubMed PubMed Central

[112] Broz P, Pelegrín P, Shao F. The gasdermins, a protein family executing cell death and inflammation. Nat Rev Immunol. 2020;20(3):143–57.10.1038/s41577-019-0228-2Search in Google Scholar PubMed

[113] Ding J, Shao F. Growing a gasdermin pore in membranes of pyroptotic cells. Embo J. 2018;37(15):e100067.10.15252/embj.2018100067Search in Google Scholar PubMed PubMed Central

[114] Wang X, Lu D, Liu Y, Wang W, Ren R, Li M, et al. Electrochemical signal amplification strategies and their use in olfactory and taste evaluation. Biosensors. 2022;12(8):566.10.3390/bios12080566Search in Google Scholar PubMed PubMed Central

[115] Li J, Liu Y, Yuan Y, Huang B. Applications of atomic force microscopy in immunology. Front Med. 2021;15(1):43–52.10.1007/s11684-020-0769-6Search in Google Scholar PubMed

[116] Rief M, Oesterhelt F, Heymann B, Gaub HE. Single molecule force spectroscopy on polysaccharides by atomic force microscopy. Science (New York, NY). 1997;275(5304):1295–7.10.1126/science.275.5304.1295Search in Google Scholar PubMed

[117] Alsteens D, Müller DJ, Dufrêne YF. Multiparametric atomic force microscopy imaging of biomolecular and cellular systems. Acc Chem Res. 2017;50(4):924–31.10.1021/acs.accounts.6b00638Search in Google Scholar PubMed

[118] Hinterdorfer P, Dufrêne YF. Detection and localization of single molecular recognition events using atomic force microscopy. Nat Methods. 2006;3(5):347–55.10.1038/nmeth871Search in Google Scholar PubMed

[119] Zhang W, Lü X, Zhang W, Shen J. EMSA and single-molecule force spectroscopy study of interactions between Bacillus subtilis single-stranded DNA-binding protein and single-stranded DNA. Langmuir. 2011;27(24):15008–15.10.1021/la203752ySearch in Google Scholar PubMed

[120] Zhang W, Dillingham MS, Thomas CD, Allen S, Roberts CJ, Soultanas P. Directional loading and stimulation of PcrA helicase by the replication initiator protein RepD. J Mol Biol. 2007;371(2):336–48.10.1016/j.jmb.2007.05.050Search in Google Scholar PubMed

[121] Burmistrova A, Fresch B, Sluysmans D, De Pauw E, Remacle F, Duwez AS. Force measurements reveal how small binders perturb the dissociation mechanisms of DNA duplex sequences. Nanoscale. 2016;8(22):11718–26.10.1039/C6NR02201DSearch in Google Scholar

[122] Brown AE, Litvinov RI, Discher DE, Weisel JW. Forced unfolding of coiled-coils in fibrinogen by single-molecule AFM. Biophys J. 2007;92(5):L39–41.10.1529/biophysj.106.101261Search in Google Scholar PubMed PubMed Central

[123] Dammer U, Hegner M, Anselmetti D, Wagner P, Dreier M, Huber W, et al. Specific antigen/antibody interactions measured by force microscopy. Biophys J. 1996;70(5):2437–41.10.1016/S0006-3495(96)79814-4Search in Google Scholar PubMed PubMed Central

[124] Yu J, Jiang Y, Ma X, Lin Y, Fang X. Energy landscape of aptamer/protein complexes studied by single-molecule force spectroscopy. Chem Asian J. 2007;2(2):284–9.10.1002/asia.200600230Search in Google Scholar PubMed

[125] Pérez-Domínguez S, Caballero-Mancebo S, Marcuello C, Martínez-Júlvez M, Medina M. Lostao Nanomechanical A. Study of Enzyme: Coenzyme Complexes: Bipartite Sites in Plastidic Ferredoxin-NADP(+) Reductase for the Interaction with NADP(). Antioxidants (Basel, Switz). 2022;11(3):537.10.3390/antiox11030537Search in Google Scholar PubMed PubMed Central

[126] Pfreundschuh M, Alsteens D, Wieneke R, Zhang C, Coughlin SR, Tampé R, et al. Identifying and quantifying two ligand-binding sites while imaging native human membrane receptors by AFM. Nat Commun. 2015;6:8857.10.1038/ncomms9857Search in Google Scholar PubMed PubMed Central

[127] Praper T, Besenicar MP, Istinic H, Podlesek Z, Metkar SS, Froelich CJ, et al. Human perforin permeabilizing activity, but not binding to lipid membranes, is affected by pH. Mol Immunol. 2010;47(15):2492–504.10.1016/j.molimm.2010.06.001Search in Google Scholar PubMed

[128] Lv J, Liu Y, Cheng F, Li J, Zhou Y, Zhang T, et al. Cell softness regulates tumorigenicity and stemness of cancer cells. Embo J. 2021;40(2):e106123.10.15252/embj.2020106123Search in Google Scholar PubMed PubMed Central

[129] Liu Y, Zhang T, Zhang H, Li J, Zhou N, Fiskesund R, et al. Cell softness prevents cytolytic T-cell killing of tumor-repopulating cells. Cancer Res. 2021;81(2):476–88.10.1158/0008-5472.CAN-20-2569Search in Google Scholar PubMed

[130] Zhang KL, Han L, Chen LY, Shi ZD, Yang M, Ren Y, et al. Blockage of a miR-21/EGFR regulatory feedback loop augments anti-EGFR therapy in glioblastomas. Cancer Lett. 2014;342(1):139–49.10.1016/j.canlet.2013.08.043Search in Google Scholar PubMed

[131] Stewart JR, O’Brian CA. Resveratrol antagonizes EGFR-dependent Erk1/2 activation in human androgen-independent prostate cancer cells with associated isozyme-selective PKC alpha inhibition. Invest N Drugs. 2004;22(2):107–17.10.1023/B:DRUG.0000011787.75522.ecSearch in Google Scholar PubMed

[132] Müller DJ, Dufrêne YF. Atomic force microscopy: a nanoscopic window on the cell surface. Trends Cell Biol. 2011;21(8):461–9.10.1016/j.tcb.2011.04.008Search in Google Scholar PubMed

[133] Guillaume-Gentil O, Potthoff E, Ossola D, Franz CM, Zambelli T, Vorholt JA. Force-controlled manipulation of single cells: from AFM to FluidFM. Trends Biotechnol. 2014;32(7):381–8.10.1016/j.tibtech.2014.04.008Search in Google Scholar PubMed

[134] Liu H, Wen J, Xiao Y, Liu J, Hopyan S, Radisic M, et al. In situ mechanical characterization of the cell nucleus by atomic force microscopy. ACS Nano. 2014;8(4):3821–8.10.1021/nn500553zSearch in Google Scholar PubMed

[135] Ando T, Uchihashi T, Scheuring S. Filming biomolecular processes by high-speed atomic force microscopy. Chem Rev. 2014;114(6):3120–88.10.1021/cr4003837Search in Google Scholar PubMed PubMed Central

[136] Casuso I, Khao J, Chami M, Paul-Gilloteaux P, Husain M, Duneau JP, et al. Characterization of the motion of membrane proteins using high-speed atomic force microscopy. Nat Nanotechnol. 2012;7(8):525–9.10.1038/nnano.2012.109Search in Google Scholar PubMed

[137] Miyagi A, Ando T, Lyubchenko YL. Dynamics of nucleosomes assessed with time-lapse high-speed atomic force microscopy. Biochemistry. 2011;50(37):7901–8.10.1021/bi200946zSearch in Google Scholar PubMed

[138] Chopinet L, Formosa C, Rols MP, Duval RE, Dague E. Imaging living cells surface and quantifying its properties at high resolution using AFM in QI™ mode. Micron. 2013;48:26–33.10.1016/j.micron.2013.02.003Search in Google Scholar PubMed

[139] Chtcheglova LA, Hinterdorfer P. Simultaneous AFM topography and recognition imaging at the plasma membrane of mammalian cells. Semin Cell Dev Biol. 2018;73:45–56.10.1016/j.semcdb.2017.08.025Search in Google Scholar PubMed

Received: 2023-08-24
Revised: 2024-01-09
Accepted: 2024-01-25
Published Online: 2024-02-15

© 2024 the author(s), published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Research Articles
  2. Tension buckling and postbuckling of nanocomposite laminated plates with in-plane negative Poisson’s ratio
  3. Polyvinylpyrrolidone-stabilised gold nanoparticle coatings inhibit blood protein adsorption
  4. Energy and mass transmission through hybrid nanofluid flow passing over a spinning sphere with magnetic effect and heat source/sink
  5. Surface treatment with nano-silica and magnesium potassium phosphate cement co-action for enhancing recycled aggregate concrete
  6. Numerical investigation of thermal radiation with entropy generation effects in hybrid nanofluid flow over a shrinking/stretching sheet
  7. Enhancing the performance of thermal energy storage by adding nano-particles with paraffin phase change materials
  8. Using nano-CaCO3 and ceramic tile waste to design low-carbon ultra high performance concrete
  9. Numerical analysis of thermophoretic particle deposition in a magneto-Marangoni convective dusty tangent hyperbolic nanofluid flow – Thermal and magnetic features
  10. Dual numerical solutions of Casson SA–hybrid nanofluid toward a stagnation point flow over stretching/shrinking cylinder
  11. Single flake homo p–n diode of MoTe2 enabled by oxygen plasma doping
  12. Electrostatic self-assembly effect of Fe3O4 nanoparticles on performance of carbon nanotubes in cement-based materials
  13. Multi-scale alignment to buried atom-scale devices using Kelvin probe force microscopy
  14. Antibacterial, mechanical, and dielectric properties of hydroxyapatite cordierite/zirconia porous nanocomposites for use in bone tissue engineering applications
  15. Time-dependent Darcy–Forchheimer flow of Casson hybrid nanofluid comprising the CNTs through a Riga plate with nonlinear thermal radiation and viscous dissipation
  16. Durability prediction of geopolymer mortar reinforced with nanoparticles and PVA fiber using particle swarm optimized BP neural network
  17. Utilization of zein nano-based system for promoting antibiofilm and anti-virulence activities of curcumin against Pseudomonas aeruginosa
  18. Antibacterial effect of novel dental resin composites containing rod-like zinc oxide
  19. An extended model to assess Jeffery–Hamel blood flow through arteries with iron-oxide (Fe2O3) nanoparticles and melting effects: Entropy optimization analysis
  20. Comparative study of copper nanoparticles over radially stretching sheet with water and silicone oil
  21. Cementitious composites modified by nanocarbon fillers with cooperation effect possessing excellent self-sensing properties
  22. Confinement size effect on dielectric properties, antimicrobial activity, and recycling of TiO2 quantum dots via photodegradation processes of Congo red dye and real industrial textile wastewater
  23. Biogenic silver nanoparticles of Moringa oleifera leaf extract: Characterization and photocatalytic application
  24. Novel integrated structure and function of Mg–Gd neutron shielding materials
  25. Impact of multiple slips on thermally radiative peristaltic transport of Sisko nanofluid with double diffusion convection, viscous dissipation, and induced magnetic field
  26. Magnetized water-based hybrid nanofluid flow over an exponentially stretching sheet with thermal convective and mass flux conditions: HAM solution
  27. A numerical investigation of the two-dimensional magnetohydrodynamic water-based hybrid nanofluid flow composed of Fe3O4 and Au nanoparticles over a heated surface
  28. Development and modeling of an ultra-robust TPU-MWCNT foam with high flexibility and compressibility
  29. Effects of nanofillers on the physical, mechanical, and tribological behavior of carbon/kenaf fiber–reinforced phenolic composites
  30. Polymer nanocomposite for protecting photovoltaic cells from solar ultraviolet in space
  31. Study on the mechanical properties and microstructure of recycled concrete reinforced with basalt fibers and nano-silica in early low-temperature environments
  32. Synergistic effect of carbon nanotubes and polyvinyl alcohol on the mechanical performance and microstructure of cement mortar
  33. CFD analysis of paraffin-based hybrid (Co–Au) and trihybrid (Co–Au–ZrO2) nanofluid flow through a porous medium
  34. Forced convective tangent hyperbolic nanofluid flow subject to heat source/sink and Lorentz force over a permeable wedge: Numerical exploration
  35. Physiochemical and electrical activities of nano copper oxides synthesised via hydrothermal method utilising natural reduction agents for solar cell application
  36. A homotopic analysis of the blood-based bioconvection Carreau–Yasuda hybrid nanofluid flow over a stretching sheet with convective conditions
  37. In situ synthesis of reduced graphene oxide/SnIn4S8 nanocomposites with enhanced photocatalytic performance for pollutant degradation
  38. A coarse-grained Poisson–Nernst–Planck model for polyelectrolyte-modified nanofluidic diodes
  39. A numerical investigation of the magnetized water-based hybrid nanofluid flow over an extending sheet with a convective condition: Active and passive controls of nanoparticles
  40. The LyP-1 cyclic peptide modified mesoporous polydopamine nanospheres for targeted delivery of triptolide regulate the macrophage repolarization in atherosclerosis
  41. Synergistic effect of hydroxyapatite-magnetite nanocomposites in magnetic hyperthermia for bone cancer treatment
  42. The significance of quadratic thermal radiative scrutinization of a nanofluid flow across a microchannel with thermophoretic particle deposition effects
  43. Ferromagnetic effect on Casson nanofluid flow and transport phenomena across a bi-directional Riga sensor device: Darcy–Forchheimer model
  44. Performance of carbon nanomaterials incorporated with concrete exposed to high temperature
  45. Multicriteria-based optimization of roller compacted concrete pavement containing crumb rubber and nano-silica
  46. Revisiting hydrotalcite synthesis: Efficient combined mechanochemical/coprecipitation synthesis to design advanced tunable basic catalysts
  47. Exploration of irreversibility process and thermal energy of a tetra hybrid radiative binary nanofluid focusing on solar implementations
  48. Effect of graphene oxide on the properties of ternary limestone clay cement paste
  49. Improved mechanical properties of graphene-modified basalt fibre–epoxy composites
  50. Sodium titanate nanostructured modified by green synthesis of iron oxide for highly efficient photodegradation of dye contaminants
  51. Green synthesis of Vitis vinifera extract-appended magnesium oxide NPs for biomedical applications
  52. Differential study on the thermal–physical properties of metal and its oxide nanoparticle-formed nanofluids: Molecular dynamics simulation investigation of argon-based nanofluids
  53. Heat convection and irreversibility of magneto-micropolar hybrid nanofluids within a porous hexagonal-shaped enclosure having heated obstacle
  54. Numerical simulation and optimization of biological nanocomposite system for enhanced oil recovery
  55. Laser ablation and chemical vapor deposition to prepare a nanostructured PPy layer on the Ti surface
  56. Cilostazol niosomes-loaded transdermal gels: An in vitro and in vivo anti-aggregant and skin permeation activity investigations towards preparing an efficient nanoscale formulation
  57. Linear and nonlinear optical studies on successfully mixed vanadium oxide and zinc oxide nanoparticles synthesized by sol–gel technique
  58. Analytical investigation of convective phenomena with nonlinearity characteristics in nanostratified liquid film above an inclined extended sheet
  59. Optimization method for low-velocity impact identification in nanocomposite using genetic algorithm
  60. Analyzing the 3D-MHD flow of a sodium alginate-based nanofluid flow containing alumina nanoparticles over a bi-directional extending sheet using variable porous medium and slip conditions
  61. A comprehensive study of laser irradiated hydrothermally synthesized 2D layered heterostructure V2O5(1−x)MoS2(x) (X = 1–5%) nanocomposites for photocatalytic application
  62. Computational analysis of water-based silver, copper, and alumina hybrid nanoparticles over a stretchable sheet embedded in a porous medium with thermophoretic particle deposition effects
  63. A deep dive into AI integration and advanced nanobiosensor technologies for enhanced bacterial infection monitoring
  64. Effects of normal strain on pyramidal I and II 〈c + a〉 screw dislocation mobility and structure in single-crystal magnesium
  65. Computational study of cross-flow in entropy-optimized nanofluids
  66. Significance of nanoparticle aggregation for thermal transport over magnetized sensor surface
  67. A green and facile synthesis route of nanosize cupric oxide at room temperature
  68. Effect of annealing time on bending performance and microstructure of C19400 alloy strip
  69. Chitosan-based Mupirocin and Alkanna tinctoria extract nanoparticles for the management of burn wound: In vitro and in vivo characterization
  70. Electrospinning of MNZ/PLGA/SF nanofibers for periodontitis
  71. Photocatalytic degradation of methylene blue by Nd-doped titanium dioxide thin films
  72. Shell-core-structured electrospinning film with sequential anti-inflammatory and pro-neurogenic effects for peripheral nerve repairment
  73. Flow and heat transfer insights into a chemically reactive micropolar Williamson ternary hybrid nanofluid with cross-diffusion theory
  74. One-pot fabrication of open-spherical shapes based on the decoration of copper sulfide/poly-O-amino benzenethiol on copper oxide as a promising photocathode for hydrogen generation from the natural source of Red Sea water
  75. A penta-hybrid approach for modeling the nanofluid flow in a spatially dependent magnetic field
  76. Advancing sustainable agriculture: Metal-doped urea–hydroxyapatite hybrid nanofertilizer for agro-industry
  77. Utilizing Ziziphus spina-christi for eco-friendly synthesis of silver nanoparticles: Antimicrobial activity and promising application in wound healing
  78. Plant-mediated synthesis, characterization, and evaluation of a copper oxide/silicon dioxide nanocomposite by an antimicrobial study
  79. Effects of PVA fibers and nano-SiO2 on rheological properties of geopolymer mortar
  80. Investigating silver and alumina nanoparticles’ impact on fluid behavior over porous stretching surface
  81. Potential pharmaceutical applications and molecular docking study for green fabricated ZnO nanoparticles mediated Raphanus sativus: In vitro and in vivo study
  82. Effect of temperature and nanoparticle size on the interfacial layer thickness of TiO2–water nanofluids using molecular dynamics
  83. Characteristics of induced magnetic field on the time-dependent MHD nanofluid flow through parallel plates
  84. Flexural and vibration behaviours of novel covered CFRP composite joints with an MWCNT-modified adhesive
  85. Experimental research on mechanically and thermally activation of nano-kaolin to improve the properties of ultra-high-performance fiber-reinforced concrete
  86. Analysis of variable fluid properties for three-dimensional flow of ternary hybrid nanofluid on a stretching sheet with MHD effects
  87. Biodegradability of corn starch films containing nanocellulose fiber and thymol
  88. Toxicity assessment of copper oxide nanoparticles: In vivo study
  89. Some measures to enhance the energy output performances of triboelectric nanogenerators
  90. Reinforcement of graphene nanoplatelets on water uptake and thermomechanical behaviour of epoxy adhesive subjected to water ageing conditions
  91. Optimization of preparation parameters and testing verification of carbon nanotube suspensions used in concrete
  92. Max-phase Ti3SiC2 and diverse nanoparticle reinforcements for enhancement of the mechanical, dynamic, and microstructural properties of AA5083 aluminum alloy via FSP
  93. Advancing drug delivery: Neural network perspectives on nanoparticle-mediated treatments for cancerous tissues
  94. PEG-PLGA core–shell nanoparticles for the controlled delivery of picoplatin–hydroxypropyl β-cyclodextrin inclusion complex in triple-negative breast cancer: In vitro and in vivo study
  95. Conduction transportation from graphene to an insulative polymer medium: A novel approach for the conductivity of nanocomposites
  96. Review Articles
  97. Developments of terahertz metasurface biosensors: A literature review
  98. Overview of amorphous carbon memristor device, modeling, and applications for neuromorphic computing
  99. Advances in the synthesis of gold nanoclusters (AuNCs) of proteins extracted from nature
  100. A review of ternary polymer nanocomposites containing clay and calcium carbonate and their biomedical applications
  101. Recent advancements in polyoxometalate-functionalized fiber materials: A review
  102. Special contribution of atomic force microscopy in cell death research
  103. A comprehensive review of oral chitosan drug delivery systems: Applications for oral insulin delivery
  104. Cellular senescence and nanoparticle-based therapies: Current developments and perspectives
  105. Cyclodextrins-block copolymer drug delivery systems: From design and development to preclinical studies
  106. Micelle-based nanoparticles with stimuli-responsive properties for drug delivery
  107. Critical assessment of the thermal stability and degradation of chemically functionalized nanocellulose-based polymer nanocomposites
  108. Research progress in preparation technology of micro and nano titanium alloy powder
  109. Nanoformulations for lysozyme-based additives in animal feed: An alternative to fight antibiotic resistance spread
  110. Incorporation of organic photochromic molecules in mesoporous silica materials: Synthesis and applications
  111. A review on modeling of graphene and associated nanostructures reinforced concrete
  112. A review on strengthening mechanisms of carbon quantum dots-reinforced Cu-matrix nanocomposites
  113. Review on nanocellulose composites and CNFs assembled microfiber toward automotive applications
  114. Nanomaterial coating for layered lithium rich transition metal oxide cathode for lithium-ion battery
  115. Application of AgNPs in biomedicine: An overview and current trends
  116. Nanobiotechnology and microbial influence on cold adaptation in plants
  117. Hepatotoxicity of nanomaterials: From mechanism to therapeutic strategy
  118. Applications of micro-nanobubble and its influence on concrete properties: An in-depth review
  119. A comprehensive systematic literature review of ML in nanotechnology for sustainable development
  120. Exploiting the nanotechnological approaches for traditional Chinese medicine in childhood rhinitis: A review of future perspectives
  121. Twisto-photonics in two-dimensional materials: A comprehensive review
  122. Current advances of anticancer drugs based on solubilization technology
  123. Recent process of using nanoparticles in the T cell-based immunometabolic therapy
  124. Future prospects of gold nanoclusters in hydrogen storage systems and sustainable environmental treatment applications
  125. Preparation, types, and applications of one- and two-dimensional nanochannels and their transport properties for water and ions
  126. Microstructural, mechanical, and corrosion characteristics of Mg–Gd–x systems: A review of recent advancements
  127. Functionalized nanostructures and targeted delivery systems with a focus on plant-derived natural agents for COVID-19 therapy: A review and outlook
  128. Mapping evolution and trends of cell membrane-coated nanoparticles: A bibliometric analysis and scoping review
  129. Nanoparticles and their application in the diagnosis of hepatocellular carcinoma
  130. In situ growth of carbon nanotubes on fly ash substrates
  131. Structural performance of boards through nanoparticle reinforcement: An advance review
  132. Reinforcing mechanisms review of the graphene oxide on cement composites
  133. Seed regeneration aided by nanomaterials in a climate change scenario: A comprehensive review
  134. Surface-engineered quantum dot nanocomposites for neurodegenerative disorder remediation and avenue for neuroimaging
  135. Graphitic carbon nitride hybrid thin films for energy conversion: A mini-review on defect activation with different materials
  136. Nanoparticles and the treatment of hepatocellular carcinoma
  137. Special Issue on Advanced Nanomaterials and Composites for Energy Conversion and Storage - Part II
  138. Highly safe lithium vanadium oxide anode for fast-charging dendrite-free lithium-ion batteries
  139. Recent progress in nanomaterials of battery energy storage: A patent landscape analysis, technology updates, and future prospects
  140. Special Issue on Advanced Nanomaterials for Carbon Capture, Environment and Utilization for Energy Sustainability - Part II
  141. Calcium-, magnesium-, and yttrium-doped lithium nickel phosphate nanomaterials as high-performance catalysts for electrochemical water oxidation reaction
  142. Low alkaline vegetation concrete with silica fume and nano-fly ash composites to improve the planting properties and soil ecology
  143. Mesoporous silica-grafted deep eutectic solvent-based mixed matrix membranes for wastewater treatment: Synthesis and emerging pollutant removal performance
  144. Electrochemically prepared ultrathin two-dimensional graphitic nanosheets as cathodes for advanced Zn-based energy storage devices
  145. Enhanced catalytic degradation of amoxicillin by phyto-mediated synthesised ZnO NPs and ZnO-rGO hybrid nanocomposite: Assessment of antioxidant activity, adsorption, and thermodynamic analysis
  146. Incorporating GO in PI matrix to advance nanocomposite coating: An enhancing strategy to prevent corrosion
  147. Synthesis, characterization, thermal stability, and application of microporous hyper cross-linked polyphosphazenes with naphthylamine group for CO2 uptake
  148. Engineering in ceramic albite morphology by the addition of additives: Carbon nanotubes and graphene oxide for energy applications
  149. Nanoscale synergy: Optimizing energy storage with SnO2 quantum dots on ZnO hexagonal prisms for advanced supercapacitors
  150. Aging assessment of silicone rubber materials under corona discharge accompanied by humidity and UV radiation
  151. Tuning structural and electrical properties of Co-precipitated and Cu-incorporated nickel ferrite for energy applications
  152. Sodium alginate-supported AgSr nanoparticles for catalytic degradation of malachite green and methyl orange in aqueous medium
  153. An environmentally greener and reusability approach for bioenergy production using Mallotus philippensis (Kamala) seed oil feedstock via phytonanotechnology
  154. Micro-/nano-alumina trihydrate and -magnesium hydroxide fillers in RTV-SR composites under electrical and environmental stresses
  155. Mechanism exploration of ion-implanted epoxy on surface trap distribution: An approach to augment the vacuum flashover voltages
  156. Nanoscale engineering of semiconductor photocatalysts boosting charge separation for solar-driven H2 production: Recent advances and future perspective
  157. Excellent catalytic performance over reduced graphene-boosted novel nanoparticles for oxidative desulfurization of fuel oil
  158. Special Issue on Advances in Nanotechnology for Agriculture
  159. Deciphering the synergistic potential of mycogenic zinc oxide nanoparticles and bio-slurry formulation on phenology and physiology of Vigna radiata
  160. Nanomaterials: Cross-disciplinary applications in ornamental plants
  161. Special Issue on Catechol Based Nano and Microstructures
  162. Polydopamine films: Versatile but interface-dependent coatings
  163. In vitro anticancer activity of melanin-like nanoparticles for multimodal therapy of glioblastoma
  164. Poly-3,4-dihydroxybenzylidenhydrazine, a different analogue of polydopamine
  165. Chirality and self-assembly of structures derived from optically active 1,2-diaminocyclohexane and catecholamines
  166. Advancing resource sustainability with green photothermal materials: Insights from organic waste-derived and bioderived sources
  167. Bioinspired neuromelanin-like Pt(iv) polymeric nanoparticles for cancer treatment
  168. Special Issue on Implementing Nanotechnology for Smart Healthcare System
  169. Intelligent explainable optical sensing on Internet of nanorobots for disease detection
  170. Special Issue on Green Mono, Bi and Tri Metallic Nanoparticles for Biological and Environmental Applications
  171. Tracking success of interaction of green-synthesized Carbopol nanoemulgel (neomycin-decorated Ag/ZnO nanocomposite) with wound-based MDR bacteria
  172. Green synthesis of copper oxide nanoparticles using genus Inula and evaluation of biological therapeutics and environmental applications
  173. Biogenic fabrication and multifunctional therapeutic applications of silver nanoparticles synthesized from rose petal extract
  174. Metal oxides on the frontlines: Antimicrobial activity in plant-derived biometallic nanoparticles
  175. Controlling pore size during the synthesis of hydroxyapatite nanoparticles using CTAB by the sol–gel hydrothermal method and their biological activities
  176. Special Issue on State-of-Art Advanced Nanotechnology for Healthcare
  177. Applications of nanomedicine-integrated phototherapeutic agents in cancer theranostics: A comprehensive review of the current state of research
  178. Smart bionanomaterials for treatment and diagnosis of inflammatory bowel disease
  179. Beyond conventional therapy: Synthesis of multifunctional nanoparticles for rheumatoid arthritis therapy
Downloaded on 13.12.2025 from https://www.degruyterbrill.com/document/doi/10.1515/ntrev-2023-0208/html
Scroll to top button