WO2011115189A1 - Dispositif d'analyse d'activité de cellules, procédé d'analyse d'activité de cellules et procédé d'analyse de cellules - Google Patents

Dispositif d'analyse d'activité de cellules, procédé d'analyse d'activité de cellules et procédé d'analyse de cellules Download PDF

Info

Publication number
WO2011115189A1
WO2011115189A1 PCT/JP2011/056304 JP2011056304W WO2011115189A1 WO 2011115189 A1 WO2011115189 A1 WO 2011115189A1 JP 2011056304 W JP2011056304 W JP 2011056304W WO 2011115189 A1 WO2011115189 A1 WO 2011115189A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
image
intensity
cells
living
Prior art date
Application number
PCT/JP2011/056304
Other languages
English (en)
Japanese (ja)
Inventor
道広 秀
雄輝 柳瀬
隆明 平郡
Original Assignee
国立大学法人広島大学
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from JP2010061710A external-priority patent/JP2011193752A/ja
Application filed by 国立大学法人広島大学 filed Critical 国立大学法人広島大学
Publication of WO2011115189A1 publication Critical patent/WO2011115189A1/fr

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54366Apparatus specially adapted for solid-phase testing
    • G01N33/54373Apparatus specially adapted for solid-phase testing involving physiochemical end-point determination, e.g. wave-guides, FETS, gratings
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/17Systems in which incident light is modified in accordance with the properties of the material investigated
    • G01N21/55Specular reflectivity
    • G01N21/552Attenuated total reflection
    • G01N21/553Attenuated total reflection and using surface plasmons
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects

Definitions

  • the present invention relates to a cell activity analyzer, a cell activity analysis method, and a cell analysis method for analyzing the reaction activity of living cells.
  • Surface plasmon resonance consists of evanescent light that is generated when light is incident on a metal thin film under total reflection conditions, and surface plasmon that is a close-packed wave of free electrons propagating on the interface between the metal thin film and the object to be measured. It is a phenomenon that resonates.
  • surface plasmon resonance occurs, at least part of the energy of the incident light is transferred to surface plasmon resonance and the intensity of the totally reflected light is reduced.
  • the incident angle at which the light intensity decreases most is called the resonance angle.
  • This resonance angle changes according to the change in the dielectric constant of the measurement object.
  • the change in the dielectric constant of the measurement object can be observed by measuring the change in the resonance angle.
  • Examples of the object measured using the principle of the surface plasmon resonance method include enzymes, antibodies, DNA, cells, and the like.
  • a method has been proposed in which a cell is an object to be measured, and an external stimulation activity for a living cell is evaluated using a surface plasmon resonance apparatus (see Patent Document 1).
  • a surface plasmon resonance apparatus is used to evaluate the activity of external stimuli on live cells using as an index the secondary signal that appears after the primary signal observed when live cells are exposed to external stimuli.
  • the signal is a dielectric constant, refractive index, or resonance angle measured in the same manner as a normal SPR measurement for living cells (hereinafter, appropriately described by any one of dielectric constant, refractive index, and resonance angle). Indicates a change.
  • Patent Document 2 describes a method and system for simply calculating the number contained in a cell population fixed on a plate.
  • the method is a method for analyzing a cell, the step of measuring reflected light intensity caused by a cell fixed on a plate using surface plasmon resonance imaging, and the reflected light intensity from the cell A step of calculating a parameter relating to.
  • a method for analyzing a cell-related parameter such as the number of cells, cell adhesion area, or cell size is described.
  • Non-Patent Document 1 describes a method for analyzing parameters relating to cells such as cell adhesion density using a surface plasmon resonance apparatus.
  • Patent Document 1 can evaluate the average value of the activity of external stimuli on a plurality of living cells in real time, it analyzes and evaluates the activity of external stimuli on individual living cells. It is difficult.
  • the method and apparatus for analyzing cells described in Patent Document 2 and the method described in Non-Patent Document 1 are parameters related to cells such as the number of cells, the adhesion area of cells, or the size of cells. It is the method and apparatus (system) aiming at calculation and analysis of. Therefore, when such a method and apparatus (system) is used, it is impossible to directly analyze and evaluate stimulus responses including parameters other than those described above for living cells. Difficult to do.
  • the present invention has been made in view of the above circumstances, and an object of the present invention is to provide a cell activity analyzer, a cell activity analysis method, and a cell analysis method capable of analyzing the activity of external stimuli for individual living cells.
  • a cell activity analyzer comprises: A cell activity analyzer that analyzes the activity of external stimuli on living cells using the surface plasmon resonance phenomenon, A metal thin film in contact with the living cell on one side; A refractive optical element having an interface substantially in contact with the other surface of the metal thin film; Incident means for causing a P-polarized parallel light beam to enter the refractive optical element and to enter the interface at a predetermined incident angle that causes the surface plasmon resonance phenomenon; A magnifying optical system for enlarging an intensity image corresponding to a two-dimensional intensity distribution of the reflected light of the parallel light beam incident on the interface to a predetermined magnification; Imaging means for capturing the intensity image magnified by the magnification optical system; Image acquisition means for sampling image data of the intensity image captured by the imaging means; Selecting means for selecting at least a partial image of the living cells as a measurement target from the image data of the intensity image sampled by the image acquisition means; The brightness value of the
  • the selection means can specify a plurality of measurement objects
  • the calculation means extracts a luminance value of each of the plurality of selected measurement objects from the image data of the intensity image, and calculates information regarding a change in intensity of reflected light of the measurement object for each measurement object. .
  • the selection means selects a plurality of different locations in the same live cell image as the measurement target.
  • the calculation means is based on a difference between a luminance value of the measurement target before applying the external stimulus to the living cells and a luminance value of the measurement target after applying the external stimulus. Then, information on the change in the intensity of the reflected light of the measurement target is calculated.
  • the calculation unit corrects information related to a change in intensity of reflected light of the measurement target based on a luminance value component of a portion where the living cells do not exist in the image data of the intensity image.
  • the predetermined incident angle is equal to a resonance angle when the living cell is not in contact with the metal thin film.
  • it further comprises analysis means for analyzing the living cells based on information on a change in intensity of reflected light of the measurement target calculated by the calculation means.
  • the analysis means extracts a characteristic of a change pattern with time of the dielectric constant of the living cell to be measured.
  • the analysis means determines whether the time-dependent change pattern of the dielectric constant of the living cell corresponds to a monophasic, biphasic, triphasic or other atypical pattern.
  • a microscope is further provided for observing the living cells in contact with the metal thin film from the one surface side.
  • the selection means includes Display means for displaying an image based on the image data of the intensity image sampled by the image acquisition means; An operation means for designating at least a partial image of the living cell as the selected measurement object from image data of the intensity image sampled by the image acquisition means by an operation input; Is further provided.
  • a plurality of the thin metal films can be arranged by separating a group of living cells including the living cells.
  • it further includes external stimulus applying means for applying different external stimuli to each of the plurality of living cell groups.
  • the cell activity analysis method comprises: A cell activity analysis method for analyzing the activity of external stimuli on living cells using surface plasmon resonance phenomenon, An arrangement step of arranging the living cells so as to contact one surface of the metal thin film; An incident step in which a P-polarized parallel light beam is incident on a refractive optical element having an interface substantially in contact with the other surface of the metal thin film, and is incident on the interface at a predetermined incident angle that causes the surface plasmon resonance phenomenon; , An enlargement step of enlarging an intensity image corresponding to a two-dimensional intensity distribution of the reflected light of the parallel light beam incident on the interface to a predetermined magnification by an enlargement optical system; An imaging step of capturing the intensity image magnified by the magnification optical system; An image acquisition step of sampling image data of the intensity image imaged in the imaging step; From the image data of the intensity image sampled by the image acquisition step, a selection step of selecting at least a partial image of the living cell as a measurement
  • a plurality of measurement objects can be specified.
  • a luminance value of each of the plurality of selected measurement objects is extracted from the image data of the intensity image, and information regarding a change in intensity of reflected light of the measurement object is calculated for each measurement object. .
  • a plurality of different locations in the same live cell image are selected as the measurement target.
  • the calculation step based on a difference between a luminance value of the measurement target before applying the external stimulus to the living cells and a luminance value of the measurement target after applying the external stimulus. Then, information on the change in the intensity of the reflected light of the measurement target is calculated.
  • information on a change in intensity of reflected light of the measurement target is corrected based on a luminance value component of a portion where the living cells do not exist in the image data of the intensity image.
  • the predetermined incident angle is equal to a resonance angle when the living cell is not in contact with the metal thin film.
  • the selection step comprises A display step of displaying an image based on the image data of the intensity image sampled in the image acquisition step; By an operation input, an operation step of designating at least a part of the live cell image as the selected measurement object from the image data of the intensity image sampled in the image acquisition step; Further included.
  • a plurality of living cell groups including the living cells are arranged apart from each other.
  • a different external stimulus is applied to each of the plurality of living cell groups.
  • the cell analysis method according to the third aspect of the present invention comprises: The living cells are analyzed based on information relating to changes in the intensity of reflected light of the living cells obtained by analysis using the cell activity analysis method of the present invention.
  • the living cells are analyzed by extracting characteristics of a change pattern of the dielectric constant of the living cells over time based on information on a change in intensity of reflected light of the measurement target.
  • the time-dependent change pattern of the dielectric constant of the living cells corresponds to a monophasic, biphasic, triphasic or other atypical pattern.
  • cancer cells are the subject of analysis.
  • cancer cells are the subject of analysis.
  • the cancer cell is any of a stomach cancer cell, a prostate cancer cell, or an angiosarcoma cell.
  • the living cells exposed to the external stimulus by cytokines are analyzed.
  • the living cells exposed to the external stimulus by EGF are analyzed.
  • the image data of the reflection intensity image of the P-polarized parallel light beam incident on the interface substantially in contact with the metal thin film with which the living cells are in contact is acquired by sampling. Then, from the acquired image data, at least a part of the image of the living cell is selected as the measurement target, and information on the change in the intensity of the reflected light of the measurement target is calculated based on the change in the luminance of the selected image.
  • a living cell to be measured is a dielectric, and its dielectric constant changes depending on a response to an external stimulus. As a result, the resonance angle of the surface plasmon resonance phenomenon changes, and the intensity of the reflected reflected light changes there.
  • the activity of the external stimulus for each living cell related to the reflected light can be analyzed based on the information.
  • it is possible to evaluate and analyze the properties of individual living cells and / or a part of each living cell, or to isolate specific types of living cells. It becomes possible to detect that the reaction is different, and it becomes possible to analyze individual cells without isolation, and to analyze each individual place in each individual cell. As a result, it is possible to detect the behavior of activity for each individual living cell.
  • FIG. 3A to 3H are diagrams illustrating an example of an image of a reflection intensity image captured when the incident angle is changed by 1 °.
  • FIGS. 4A to 4C are diagrams illustrating an example of temporal changes in the image of the reflection intensity image when the living cells (RBL-2H3 cells) are not stimulated.
  • FIG. 5A to FIG. 5C are diagrams showing an example of temporal changes in the image of the reflection intensity image when a living cell (RBL-2H3 cell) is stimulated.
  • FIG. 3A to 3H are diagrams illustrating an example of an image of a reflection intensity image captured when the incident angle is changed by 1 °.
  • FIGS. 4A to 4C are diagrams illustrating an example of temporal changes in the image of the reflection intensity image when the living cells (RBL-2H3 cells) are not stimulated.
  • FIG. 5A to FIG. 5C are diagrams showing an example of temporal changes in the image of the reflection intensity image when a living cell (RBL-2H3 cell) is stimulate
  • FIG. 6A is a graph illustrating an example of a temporal change in the intensity of reflected light of a measurement target that is not stimulated.
  • FIG. 6B is a graph showing an example of a temporal change in the intensity of reflected light of a measurement target stimulated with living cells (RBL-2H3 cells). It is a block diagram which shows the detailed structure of the image processing part of FIG. It is a figure for demonstrating the various components contained in the intensity
  • FIGS. 9A to 9C are diagrams showing an example of changes in the intensity of reflected light in different parts of a living cell (RBL-2H3 cell). It is a flowchart of an example of a cell activity analysis method (analyte subject cell analysis).
  • FIG. 12A is a perspective view of a part of a cell activity analyzer equipped with a multiwell chamber
  • FIG. 12B shows an image captured by the cell activity analyzer of FIG.
  • 13A is a diagram showing an example of an image of a reflection intensity image when the PAM212 cell is stimulated
  • FIG. 13B is a diagram when the A431 cell is stimulated
  • FIG. 14A is a graph showing an example of temporal change in intensity of reflected light when PAM212 cells are stimulated and FIG. 14B is stimulated with A431 cells.
  • FIG. 16 is a graph showing an example of a temporal change in intensity of reflected light when stimulated with DNP-HSA and PMA in the sensor chip shown in FIG.
  • FIGS. 17A to 17C are diagrams illustrating an example of a temporal change in the image of the reflection intensity image when the sensor chip illustrated in FIG. 15 is stimulated with DNP-HSA and PMA.
  • FIG. 17A to 17C are diagrams illustrating an example of a temporal change in the image of the reflection intensity image when the sensor chip illustrated in FIG. 15 is stimulated with DNP-HSA and PMA.
  • FIG. 4 is a diagram showing a sensor chip on which RBL-2H3 cells bound with anti-DNP-mouse IgE and RBL-3D4 cells bound with a human IgE antibody are arranged.
  • FIG. 19 is a graph showing an example of a temporal change in intensity of reflected light when stimulated with DNP-HSA and an anti-human IgE antibody in the sensor chip shown in FIG.
  • FIGS. 20A to 20C are diagrams illustrating an example of a temporal change in the image of the reflection intensity image when the sensor chip illustrated in FIG. 18 is stimulated with DNP-HSA and anti-human IgE.
  • FIGS. 22A and 22B are diagrams showing an example of the temporal change in reflection intensity when the sensor chip shown in FIG. 21 is stimulated with DNP-HSA and EGF.
  • FIG. 23A and FIG. 23B are diagrams illustrating an example of a temporal change in an image of a reflection intensity image when stimulated with DNP-HSA and EGF in the sensor chip illustrated in FIG. It is a figure which shows the change of the resonance angle with time by EGF stimulation in the CHO cell which forcedly expressed wild type human EGFR.
  • FIG. 33 (A) is a diagram showing the results of Western blotting using an anti-phosphorylation-specific EGFR antibody and an anti-EGFR antibody of a human hemangiosarcoma cell line.
  • FIG. 33 (B) is a graph showing changes in resonance angle over time by EGF stimulation in a human hemangiosarcoma cell line.
  • the cell activity analyzer 100 is an apparatus that analyzes the activity observed when a living cell as a subject is exposed to an external stimulus using a surface plasmon resonance phenomenon.
  • the temperature of the space in which the cell activity analyzer 100 is installed is preferably adjusted to 37 ° C. by a thermostat (not shown), but is not limited thereto.
  • the cell activity analyzer 100 includes a light source 1, a polarizing plate 2, a prism 3, a glass substrate 4, a metal thin film 5, an objective lens 6, an imaging unit 7, and a computer 8. , A flow cell 9, a liquid supply unit 10, and a microscope 11.
  • the optical axis of the optical system including the light source 1, the polarizing plate 2, the prism 3, the glass substrate 4, the metal thin film 5, the objective lens 6, and the imaging unit 7 is defined as AX. .
  • the light source 1 is, for example, a semiconductor laser. This semiconductor laser oscillates and outputs laser light having a wavelength of 635 nm, for example.
  • the laser beam output from the light source 1 is converted into a parallel light beam by a collimator lens (not shown) or the like and enters the polarizing plate 2.
  • a collimator lens not shown
  • the light source 1 you may use red, white LED (Light * Emitting * Diode), etc.
  • the polarizing plate 2 converts the incident laser light into a linearly polarized parallel light beam and emits it. This linearly polarized light becomes P polarized light with respect to an interface F between a glass substrate 4 and a metal thin film 5 described later.
  • the light source 1 and the polarizing plate 2 correspond to the incident means.
  • the prism 3 for example, S-LAL-10 glass is employed.
  • the refractive index of this glass is 1.72.
  • the prism 3 receives the parallel light flux that has been changed to P-polarized light by the polarizing plate 2.
  • the glass substrate 4 for example, S-LAL-10 glass is adopted. That is, the glass substrate 4 and the prism 3 have the same refractive index. Both are bonded by a matching oil having a refractive index of 1.72. Thereby, the laser light (P-polarized light) incident on the prism 3 is incident on the glass substrate 4 and goes straight as it is.
  • the prism 3 and the glass substrate 4 correspond to a refractive optical element.
  • a metal thin film 5 is deposited on the glass substrate 4.
  • the metal thin film 5 is, for example, a gold film.
  • thin films such as Ag, Cu, Zn, Al, and K can also be used as the metal thin film 5.
  • the thickness of the metal thin film 5 is, for example, 50 nm.
  • the metal thin film 5 is formed on the glass substrate 4 by vapor deposition, for example.
  • the laser light reflected by the interface F is emitted from the glass substrate 4 and the prism 3 and enters the objective lens 6.
  • the objective lens 6 refracts and emits laser light.
  • a lens having a longer rear focal length than a front focal length is used. Therefore, the objective lens 6 enlarges the object image at a predetermined magnification and forms an image on the image plane.
  • the laser light emitted from the objective lens 6 reaches the imaging surface of the imaging unit 7.
  • the imaging unit 7 is, for example, a CCD (Charge-Coupled Device) image sensor or a CMOS (Complementary Metal-Oxide Semiconductor) image sensor.
  • the imaging unit 7 receives the laser light reflected by the interface F.
  • the imaging surface of the imaging unit 7 and the interface F are in a conjugate relationship. Therefore, an intensity image corresponding to the two-dimensional intensity distribution of the reflected light of the parallel light beam incident on the interface F of the prism 3, that is, a reflection intensity image is formed on the imaging surface of the imaging unit 7.
  • This reflection intensity image is magnified by, for example, 2 to 40 times by the objective lens 6.
  • the imaging unit 7 captures the reflection intensity image.
  • the imaging unit 7 outputs an image signal corresponding to the reflected intensity image.
  • the image signal output from the imaging unit 7 is input to the computer 8.
  • the computer 8 has a CPU and a memory (both not shown). When the CPU executes the program stored in the memory, the functions of the image acquisition unit 21, the image processing unit 22, the display unit 23, and the operation unit 24 illustrated in FIG. 1 are realized.
  • the image acquisition unit 21 samples the input image signal at regular time intervals, and outputs the image data obtained by the sampling to the image processing unit 22.
  • the image processing unit 22 outputs the image data acquired by the image acquisition unit 21 to the display unit 23.
  • the display unit 23 displays an image based on the input image data.
  • the operation unit 24 is a user interface that receives user operation input, and includes, for example, a keyboard, a touch panel, and a mouse.
  • the operation unit 24 is operated by a user who views an image displayed on the display unit 23.
  • the user operates the operation unit 24 to specify a specific measurement target in the image of the reflection intensity image displayed on the display unit 23, for example.
  • the position coordinates of the measurement target in the designated image are input to the image processing unit 22.
  • the image processing unit 22 determines the luminance value of the measurement target specified by the operation input of the operation unit 24 from the sampled reflection intensity image (if the measurement target is a region, the average value of the luminance values of that region). , And information on the temporal change in the intensity of the reflected light of the measurement target is calculated based on the extracted temporal change in the luminance value of the measurement target. This time change in the intensity of the reflected light corresponds to a time change in the dielectric constant of the measurement target.
  • the image processing unit 22 graphs the information related to the temporal change in the calculated reflected light intensity of the measurement target, and outputs the image data of the graph to the display unit 23.
  • the display unit 23 displays simultaneously the image of the reflection intensity image obtained by sampling and the image of the graph showing the temporal change in the intensity of the reflected light in the designated measurement target.
  • the user can analyze the activity of the external stimulus with respect to the living cells while referring to the graph showing the temporal change of the intensity of the reflected light.
  • live cells C1 and C2 which are measurement targets of the active reaction, are attached.
  • the method of attaching the living cells C1 and C2 to the metal thin film 5 is performed by using, for example, an appropriate spacer (poly-L-lysine or the like) between the living cells C1 and C2 and the metal thin film 5, and living cells C1 and C2.
  • an appropriate spacer poly-L-lysine or the like
  • any method can be used as long as it is known in the art, any method may be used.
  • a method of utilizing the affinity of the cell membrane for lipid, a method of covalently tethering, or a method of tethering with a positive charge developed by the present inventors Yamamoto e et al. 2007. Biosensors Bioelectron. 23, 562-567 and JP 2007-14327 A).
  • a flow cell 9 is provided as a flow path for flowing a liquid.
  • the flow cell 9 is a flow path for flowing a liquid to be exposed to the living cells C1 and C2 set on the metal thin film 5.
  • the flow cell 9 is connected to the liquid supply unit 10.
  • a liquid to be exposed to the living cells C1 and C2 is supplied from the liquid supply unit 10 into the flow cell 9.
  • the liquid flowing through the flow cell 9 contains, for example, an antigen that may bind to the antibodies on the living cells C1 and C2.
  • the living cells C1 and C2 are stimulated and activated (see, for example, Patent Document 1). That is, since the external stimulation is applied to the living cells C1 and C2 by exposing the living cells C1 and C2 to the liquid by the flow cell 9 and the liquid supply unit 10, in this embodiment, the flow cell 9 and the liquid supply unit 10 are stimulated.
  • the flow cell 9 and the liquid supply unit 10 correspond to an exposure unit that exposes a living cell as a subject to be exposed to an external stimulus.
  • the exposure means holds the living cells to be examined as a subject in an environment of a predetermined condition, and exposes the living cells of the subject to a given external stimulus.
  • the microscope 11 is installed to observe the living cells C1 and C2 set on the metal thin film 5 from the opposite side of the interface F.
  • the metal thin film 5 is provided with alignment marks (not shown) in advance. This mark is provided at a position that falls within both the imaging field of the microscope 11 and the imaging field of the imaging unit 7. With this mark, the positional relationship between a specific portion in the observation image of the microscope 11 and a specific portion of the image captured by the imaging unit 7 becomes clear. With reference to the image of the mark appearing in both images, the position of the specific living cells C1 and C2 that appear in the image of the reflection intensity image captured by the imaging unit 7 within the observation field of the microscope 11 is specified, for example, pipetting operation, etc. In addition, the living cells C1 and C2 can be taken out using a method known in the art.
  • the graph of FIG. 2 shows an example of the incident angle dependence of the intensity of the reflected light received by the imaging unit 7.
  • the horizontal axis indicates the incident angle ⁇ of the parallel light flux to the interface F
  • the vertical axis indicates the intensity of the reflected light at the incident angle ⁇ .
  • FIG. 2 shows three characteristic curves (a) to (c).
  • the characteristic curve (a) is a curve showing the incident angle dependence of the intensity of the reflected light in a state where nothing is placed on the metal thin film 5 (the state where there are no living cells C1 and C2). According to this, the intensity of reflected light is most attenuated at an incident angle of 56 ° due to the surface plasmon resonance phenomenon. This incident angle of 56 ° is called a resonance angle. In the present embodiment, the incident angle of the laser beam to the interface F is set to 56 ° so that the intensity of the reflected light when the living cells C1 and C2 are not in contact is the darkest.
  • the characteristic curve (b) is a curve showing the incident angle dependence of the reflected light intensity when the living cells C1 and C2 are set on the metal thin film 5 and the living cells C1 and C2 are not yet stimulated. Since the living cells C1 and C2 are dielectrics, the dielectric constant changes around the metal thin film 5 where the living cells C1 and C2 come into contact, and the incident angle dependence of the reflected light intensity is characteristic from the characteristic curve (a). The curve (b) is shifted and the resonance angle is also shifted from 56 ° to ⁇ 1.
  • the characteristic curve (c) shows that the living cells C1 and C2 are set on the metal thin film 5, and the living cells C1 and C2 are stimulated by the antigen contained in the liquid flowing through the flow cell 9, and in response to the stimulation. It is a curve which shows the incident angle dependence of reflected light intensity.
  • the living cells C1 and C2 are stimulated and respond to the stimulation, the dielectric constants of the living cells C1 and C2 are further changed. Therefore, the incident angle dependence of the intensity of the reflected light is changed from the characteristic curve (b) to the characteristic curve (c).
  • the resonance angle is also shifted from ⁇ 1 to ⁇ 2.
  • the incident angle of the parallel light flux on the interface F is fixed at 56 °. Therefore, focusing attention on an incident angle of 56 °, the intensity of the reflected light is I1 and is the darkest when the living cells C1 and C2 are not in contact with the metal thin film 5. Further, when the living cells C1 and C2 adhere to the metal thin film 5, the intensity of the reflected light becomes I2, which is higher by ⁇ I1 than I1. Further, when the living cells C1 and C2 attached to the metal thin film 5 are stimulated by an antigen or the like and respond to the stimulation, the intensity of the reflected light increases from I2 by ⁇ I2 to I3, and becomes stronger.
  • the place where the living cells C1 and C2 do not exist becomes dark, the place where the cells exist becomes bright, and the living cells C1 and C2 become bright.
  • the activated area becomes brighter.
  • FIGS. 3A to 3H show images of reflection intensity images when the incident angle ⁇ of the laser beam is changed by 1 ° from 53 ° to 60 °.
  • the contrast between the place where the live cells C1 and C2 are present and the place where the live cells C1 and C2 are not present is the highest in the incident angle.
  • 56 °.
  • FIGS. 4A to 4C show changes in the image of the reflection intensity image after 0 minutes, 10 minutes, and 20 minutes in a state where the living cells C1 and C2 are not stimulated.
  • . 5A to 5C show changes in the reflection intensity image after 0 minutes, 10 minutes, and 20 minutes after stimulating the living cells C1 and C2.
  • FIGS. 5A to 5C it is clear that when the living cells C1 and C2 react to an external stimulus, the living cells C1 and C2 It can be seen that the luminance of the portion corresponding to is changing.
  • the user While viewing the reflection intensity image displayed on the display unit 23 of the computer 8, the user operates the operation unit 24 (for example, a mouse) to specify a specific measurement target in the image of the reflection intensity image displayed on the display unit 23.
  • This measurement target may be specified as a point (for example, a part of one living cell) or may be specified as a region (for example, an entire region of one living cell).
  • the user can designate a plurality of living cells at a time by operating the operation unit 24 and selecting, for example, several bright portions shown in FIG.
  • the image processing unit 22 creates an image of a graph of the change in the intensity of the reflected light of the portion corresponding to the designated living cell and causes the display unit 23 to display the image.
  • FIG. 6 (A) and 6 (B) show the temporal change in the intensity of the reflected light of the portion corresponding to each of several living cells specified in this way and the time of the average value of the intensity of the reflected light. Changes are shown.
  • FIG. 6 (A) the time change etc. of the intensity
  • the time change of the intensity of the reflected light of each part in the reacted state is shown.
  • FIG. 6 (A) and FIG. 6 (B) when live cells are stimulated, reacted, and activated, the intensity of the reflected light at that portion changes greatly. Note that the horizontal line at the bottom of the graph in FIG. 6B indicates that at that time, a solution (a solution containing DNP-HSA) is caused to flow through the flow cell 9 and the living cells are stimulated. 4A to FIG. 4C, FIG. 5A to FIG. 5C, FIG. 6A, and FIG. 6B will be described in detail in the embodiments described later. Explained.
  • FIG. 7 shows a further detailed configuration of the image processing unit 22 of the computer 8.
  • the image processing unit 22 includes an operation content analysis unit 30, a measurement target extraction unit 31, an initial value holding unit 32, a dark component extraction unit 33, difference units 34 and 35, and waveform generation. Part 36.
  • the operation content analysis unit 30 analyzes the operation content input from the operation unit 24.
  • the operation content obtained as a result of analysis includes the position coordinates in the specified measurement target image, the position coordinates in the image at the position specified as a part where no living cells exist, the measurement start command of waveform data, etc. include.
  • the operation content analysis unit 30 outputs the position coordinates in the designated measurement target image to the measurement target extraction unit 31. Further, the operation content analysis unit 30 outputs a waveform data measurement start command to the measurement target extraction unit 31, the initial value holding unit 32, and the dark component extraction unit 33. Further, the operation content analysis unit 30 outputs the position coordinates in the image at the position designated as a part where no living cells exist to the dark component extraction unit 33.
  • the measurement target extraction unit 31 extracts the luminance value of the measurement target for each measurement target based on the position coordinates of the measurement target input from the operation content analysis unit 30. Output. Note that only the luminance value of the first measurement target at the time when the waveform data measurement start command is input is output to the initial value holding unit 32.
  • the initial value holding unit 32 holds and outputs the luminance value of the measurement target output from the measurement target extraction unit 31 for each measurement target when the waveform data measurement start command is input. This output is held as an initial value during measurement and continues to be output.
  • the dark component extraction unit 33 extracts and outputs the amount of change in the luminance value of the portion where there is no live cell input from the operation content analysis unit 30.
  • the initial value of this change amount is zero. After that, the luminance value obtained at the next sampling time is calculated as the amount of change.
  • the difference unit 34 subtracts the initial value of the measurement target luminance value output from the initial value holding unit 32 from the measurement target luminance value output from the measurement target extraction unit 31 and outputs the result.
  • the difference unit 35 subtracts the luminance value output from the dark component extraction unit 33 from the luminance value output from the difference unit 34 and outputs the result.
  • the waveform generation unit 36 converts the luminance value output from the difference unit 35 into the intensity of the reflected light, and arranges the intensity of the reflected light obtained so far in time series, thereby changing the intensity of the reflected light over time. Generate and output graph image data.
  • the reflected light intensity I in the reflection intensity image includes (A) a change due to the reaction of the living cell and (B) a default intensity component of the living cell before giving the stimulus. And (C) a component of intensity change in a region where no living cells exist.
  • the luminance value output from the measurement target extraction unit 31 corresponds to the intensity I of the reflected light
  • the luminance value output from the initial value holding unit 32 corresponds to the component (B)
  • the luminance value output from the extraction unit 33 corresponds to the component (C).
  • the components (B) and (C) consist of background and noise, respectively. Therefore, the difference units 34 and 35 subtract the luminance value output from the initial value holding unit 32 and the luminance value output from the dark component extraction unit 33 from the luminance value output from the measurement target extraction unit 31. If the luminance value is corrected, the waveform data of the component (A) to be originally measured can be acquired with high accuracy.
  • live cells C 1 and C 2 are set on the metal thin film 5.
  • emission of laser light is started, a reflection intensity image is captured by the imaging unit 7, and an image of the reflection intensity image is displayed on the display unit 23.
  • the portion where the living cells C1 and C2 are present is displayed brightly. Therefore, the user who has viewed the image displayed on the display unit 23 operates the operation unit 24 to display the image in the image.
  • the living cells C1 and C2 are designated as measurement targets, and the portion where no living cells exist is designated.
  • This step is an exposure step in which live cells to be examined are exposed to an external stimulus.
  • the exposure method may be a method suitable for each external stimulus. For example, in the case of giving stimulation with EGF, the target living cells may be infiltrated into an EGF solution having an appropriate concentration to give the stimulation.
  • This step is a dielectric constant measurement step for measuring a change with time in the dielectric constant of the cells C1 and C2 exposed to the external stimulus in the exposure step.
  • the measurement result of the dielectric constant of the living cell is stored together with the measured time.
  • the time information to be measured may be a relative time from the start of measurement.
  • the measured dielectric constant and the information on the time constitute a time-dependent change pattern of the dielectric constant of the living cells of the subject.
  • the user analyzes the activity of the external stimulus for the living cells C1 and C2 based on the change pattern of the dielectric constant with time, that is, the time change. For example, it is possible to detect that the living cell C1 is activated but the living cell C2 is not activated, and the like that the reaction is different for each living cell.
  • the waveform generation unit 36 of the image processing unit 22 stores waveform data indicating the temporal change characteristics of reflected light intensity in several living cells as analysis means. Then, the waveform generator 36 obtains the correlation between the waveform of the living cell to be measured currently being created and the waveform data already stored, and analyzes the cell having the maximum correlation as the living cell being measured. can do.
  • each of the living cells displayed on the display unit 23 is measured. If it designates as a target, it can analyze without isolating every living cell of each shape.
  • the user can collect the living cells in a live state by pipetting operation or the like while observing the living cells with the microscope 11.
  • the cell activity analyzer 100 may be further provided with a pipette device (not shown) that automatically collects live cells, and the live cells may be automatically collected using the pipette device.
  • a plurality of different portions of the same living cell can be designated as a measurement target.
  • the measurement points P1 to P13 which are a plurality of measurement objects randomly extracted, are designated within one living cell by the operation input of the operation unit 24.
  • P1 to P5 are measurement points near the center of the living cell, that is, near the nucleus
  • P6 to P12 are measurement points near the edge of the living cell, that is, near the cell membrane.
  • P13 is a measurement point where there is no cell.
  • FIG. 9C shows the change in the average value of the intensity of reflected light at the measurement points P1 to P5, that is, the measurement points near the center of the living cell, and the average value of the intensity of reflected light at the measurement points P6 to P12. Changes.
  • the change in reflected light intensity is larger at the measurement point near the center of the living cell, that is, near the nucleus than at the measurement point near the edge of the living cell, that is, near the cell membrane.
  • an image of the reflection intensity image of the P-polarized parallel light beam incident on the interface F substantially in contact with the metal thin film 5 in contact with the living cells C1 and C2 is obtained by sampling. Is done. Then, from the acquired image data, at least a part of the images of the living cells C1 and C2 is selected as the measurement target, and information on the change in the intensity of the reflected light of the measurement target is obtained based on the change in the luminance of the selected image. Calculated.
  • the living cells C1 and C2 to be measured are dielectrics, and the dielectric constant changes due to a reaction caused by an external stimulus.
  • the dielectric constants of the living cells C1 and C2 change, the dielectric constant around them changes, and as a result, the resonance angle of the surface plasmon resonance phenomenon changes, and the intensity of the reflected light reflected thereby changes. Therefore, if information on the change in the intensity of the reflected light of the portion related to the selected individual living cells C1 and C2 is calculated, the activity of the external stimulus for the individual living cells C1 and C2 is analyzed based on the information. be able to.
  • the waveform generation unit 36 uses the waveform data to be measured as an extraction means to determine the characteristics of the temporal change pattern of the dielectric constant observed when the living cells to be examined are exposed to an external stimulus. You may make it extract. In this way, it is possible to analyze living cells using the characteristics of the temporal change pattern of the dielectric constant as an index.
  • the process performed by the waveform generation unit 36 at this time is an extraction process for extracting characteristics of the temporal change pattern of the dielectric constant of the living cells to be examined.
  • diagnosis and analysis of malignant tumors, etc. are based on visual observation, X-ray, CT (Computed Tomography), or image information by ultrasound, and finally, a tissue structure using a pathological tissue specimen is observed microscopically. It was done based on that. Sometimes, in addition to these, information on the presence or absence of gene abnormality in the suspected tissue or the expression of a marker substance related to cancer may be added.
  • the presence or absence of expression of a specific gene or other substance can be grasped by a concept slightly different from structural disturbance, but after a biological tissue or cell suspected of having cancer is chemically fixed, In terms of analyzing the amount of quantity, it is still an analysis of the structure of cells or living tissues at a certain moment.
  • normal cells and cancer cells exhibit characteristics of temporal change patterns of different resonance angles with respect to EGF (epidermal growth factor) stimulation by an SPR device. It was elucidated. Furthermore, histopathologically, it was shown that even the same type of cancer or cancer cell line can exhibit different temporal changes in resonance angle characteristics in response to EGF stimulation.
  • EGF epidermal growth factor
  • EGFR epidermal Growth factor receptor, Epidermal Growth
  • Factor ⁇ ⁇ Receptor which is not expressed in normal vascular component cells. Similarly, it has been elucidated that it shows the characteristics of the change pattern with time of different resonance angles.
  • the waveform generation unit 36 analyzes the living cells that are the object of the subject, using the extracted characteristic of the change pattern of the dielectric constant over time as an index. More specifically, the waveform generation unit 36 extracts the characteristics of the measured change pattern of the dielectric constant of living cells over time, for example, monophasic, biphasic, triphasic or other atypical patterns. It is determined which of the conditions is met. The waveform generation unit 36 may perform a more detailed analysis of living cells by comparing with a characteristic of a temporal change pattern of a predetermined dielectric constant.
  • the characteristics of a predetermined dielectric constant change pattern over time include, for example, normal cells (cells without abnormality) that have been measured in advance, and various cancer cells (epithelial / It means the characteristics of the time-dependent change pattern of the dielectric constant of various tumor cells (non-epithelial / fluid), various cancer cells (epithelial malignant tumors) or various cancer cell lines.
  • the waveform generation unit 36 determines whether or not the living cell that is the subject of the subject is a normal cell, whether or not the target living cell is a cancer cell or a cancer cell, or in the case of a cancer cell Diagnosis / analysis of cancer cells of such types and cell lines (for example, gastric cancer cells, prostate cancer cells, hemangiosarcoma cells (see Examples described later), etc.), etc.
  • the diagnosis / analysis result that is, the information on the characteristics of the extracted change pattern of the dielectric constant with time (information on the state of the living cells) is sent to the display unit 23.
  • the display unit 23 displays live cell state information sent from the waveform generation unit 36.
  • the waveform generation unit 36 may further include a database in which conditions are stored. In this case, for example, the waveform generation unit 36 performs the determination by comparing the conditions stored in the database with the extracted characteristics.
  • Such conditions include, for example, the presence or absence of local maximum and local minimum values of the change pattern, the order of local maximum and local minimum values, the initial value, the maximum and minimum values, the range of time for taking local maximum and local minimum values, Conditions for identifying the characteristics of the change pattern, such as the sign and size of the change rate, the change rate of the change rate, etc., can be set.
  • FIG. 10 is a flowchart showing an example of a cell activity analysis method. As shown in FIG. 10, for example, when analysis of a living cell of a subject is started, an exposure test is performed (step S11). During the exposure test, the dielectric constant is measured (step S12).
  • the characteristics of the dielectric constant change pattern over time are extracted (step S13).
  • the extracted characteristics are monophasic, biphasic, triphasic, or other atypical patterns. It is determined which condition is met (step S14). The extraction and the determination thereof are performed in the waveform generation unit 36 as analysis means. Thereafter, the state of the cell with the characteristics of the time-dependent change pattern of the dielectric constant of the extracted characteristics (for example, monophasic, biphasic, triphasic or other atypical patterns) is the object of the subject. The state of the living cell is analyzed and sent to the display unit 23 for display (step S15).
  • an analysis system for cancer cells may be used as an analysis system for cancer cells or an analysis system for cancer cells (for example, gastric cancer cells, prostate cancer cells or angiosarcoma cells).
  • an analysis system for cancer cells for example, gastric cancer cells, prostate cancer cells or angiosarcoma cells.
  • the state of cells can be comprehensively and directly analyzed at the level of individual living cells.
  • normal cells and cancers can be obtained only by dynamically analyzing the stimulation response patterns of cells without fixing cells or tissues as in the prior art and evaluating malignant tumors as a potential.
  • the ability to analyze the types of cells, cancer types and cancer cell lines is extremely high in technical value.
  • the display unit 23 and the operation unit 24 correspond to the selection unit, but the present invention is not limited to this.
  • the luminance of the region where the living cells are present is the luminance of the region where the living cells are not present.
  • the image processing unit 22 may automatically select a living cell to be measured as a selection unit by using the fact that it is larger than that.
  • the image processing unit 22 extracts a point where a change in spatial luminance is equal to or greater than a predetermined threshold, that is, a change point (edge) in the captured reflection intensity image, and sets the image data surrounded by the edge. May be selected as a measurement target.
  • the optical system of the Kretschmann arrangement in which the reflection surface of the laser beam is the interface F between the metal thin film 5, the glass substrate 4, and the metal thin film 5, is not limited to this.
  • An optical system with an otto arrangement may be employed.
  • the interface F needs to be disposed at a distance substantially in contact with the nanometer order such that near-field light (evanescent light) is generated with respect to the metal thin film 5.
  • the metal thin film 5 does not need to be provided on the entire surface of the imaging field, and as shown in FIG. 11, small metal thin films 15 may be arranged on the glass substrate 4 in a matrix, for example. In this case, different live cells may be arranged on each metal thin film 15. In this way, the response of various living cells to the same external stimulus can be measured at a time.
  • the stimulus applying means is the flow cell 9 and the liquid supply unit 10, but the present invention is not limited to this.
  • a droplet discharge device having a nozzle for dropping a droplet containing an antigen that gives an external stimulus to living cells on the metal thin film 5 may be used as the stimulus applying means.
  • a plurality of nozzles may be provided, a cluster of living cells (for example, a collection of 1 to 100 living cells) may be arranged, and droplets containing different antibodies or the like may be discharged to each cluster.
  • a cell activity analyzer 100 that omits the flow cell 9 and the liquid supply unit 10 is used.
  • a liquid known in the art such as a pipette or an injector.
  • An embodiment in which a person operates the drop ejection device to give an external stimulus is also possible.
  • each living cell is based on the temporal change (change before and after applying the stimulus) of the calculated reflected light intensity. And / or the properties of a part of individual living cells can be evaluated.
  • the properties of individual living cells can be analyzed and isolated, living cells with certain activities can be screened, specific biomolecules related to cell activity (external stimuli) can be screened, It can be used for various cell studies such as investigating in which part of live cells activation occurs.
  • the cell activity analysis apparatus 100 and the cell activity analysis method according to the present embodiment can be used for medical diagnosis apparatuses, diagnosis methods, and the like. For example, it can be used for a rapid allergic reaction test of living cells (blood or biopsy material) collected from a living body. It is also possible to stimulate a lesion cell such as cancer with a growth factor or the like and quickly determine whether it is a normal cell or a malignant cell. Furthermore, analysis of the required drug amount that varies depending on the individual (analysis of drug reactivity of peripheral blood cells (lymphocytes, basophils, eosinophils, antigen-presenting cells, etc.) for each individual) can be performed. In addition, it can be used for analysis of drugs that cause drug allergy.
  • peripheral blood cells lymphocytes, basophils, eosinophils, antigen-presenting cells, etc.
  • the cell activity analyzer 100 and the cell activity analysis method according to the present embodiment it is possible to analyze individual living cells and / or a part of each living cell, and thus the above-described sensitivity is higher than that in the past. Such diagnosis, evaluation or analysis can be performed.
  • the cell activity analyzer 100 and the cell activity analysis method of the present embodiment can also be used as a high-throughput screening apparatus for clinical diagnosis, for example, a high-throughput allergy diagnosis apparatus.
  • a solution containing basophils is obtained from blood collected from a living body using micro magnetic beads or the like (others may be any if including living cells).
  • the basophil solution is described in a multi-well chamber 40 as shown in FIG. 12A (for example, in which wells are vacated in a matrix so that the injected solution is separated) at the time of discharging an external stimulus. It is injected by a droplet discharge device such as that described above or pipetting.
  • an allergen administration multi-chamber 41 designed to fit the multi-well chamber 40 into which the basophil solution is injected is also prepared.
  • the allergen administration multi-chamber 41 is also configured as a droplet discharge device as described above, and different allergens are preferably administered simultaneously to each well into which blood has been injected. Therefore, the cell activity analyzer 100 and the cell activity analysis method of this embodiment are used.
  • An image captured by the imaging unit 7 of the cell activity analyzer 100 described above is displayed on the display unit 23 of the computer 8. Then, an image as shown in FIG. 12B is displayed, and a highly reliable diagnosis can be performed in a shorter time than a conventional diagnosis method.
  • the allergen may be the same and the specimen from which the basophil solution is collected may be different.
  • Such an apparatus can be used as various high-throughput screening apparatuses such as a high-throughput cancer diagnostic apparatus in addition to an allergy diagnostic apparatus.
  • the term “cell” is defined in the same way as the broadest meaning used in the field, and may be any kind or animal cell. Moreover, it may be a naturally occurring cell or an artificially modified cell (for example, a fused cell or a genetically modified cell). “Live cell” refers to a living cell. Of these, cells derived from humans (Homo sapience) are preferred, and mast cells, keratinocytes, human basophils or human B cells are preferred, but not limited thereto.
  • predetermined magnification means the magnification of an intensity image by a magnifying optical system such as the objective lens 6.
  • the predetermined magnification needs to be a magnification at which the image data sampled by the image acquisition means can be identified by individual living cells.
  • the predetermined magnification is, for example, 2 to 40 times as described above.
  • “external stimulation” refers to binding of a ligand to a cell surface receptor (for example, a biomolecule such as an antigen described in the present embodiment), environmental changes such as temperature or pH, or mechanical It means a stimulus or an electrical stimulus, and includes all stimuli that act on the activity of a cell (for example, activation of an information transmission system in the cell).
  • a cell surface receptor for example, a biomolecule such as an antigen described in the present embodiment
  • environmental changes such as temperature or pH, or mechanical
  • It means a stimulus or an electrical stimulus, and includes all stimuli that act on the activity of a cell (for example, activation of an information transmission system in the cell).
  • ligand binding to cell surface receptors is preferred, and external stimulation with cytokines is more preferred.
  • the cytokines in this specification include all types of cytokines known in the art.
  • interleukin, chemokine, interferon, hematopoietic factor, cell growth factor and the like can be mentioned, among which cell growth factor is preferable.
  • the cell growth factor include EGF, fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), hepatocyte growth factor (HGF) or transforming growth factor (TGF) described above. Of these, EGF is most preferred.
  • exposure means that each external stimulus as described above is given to living cells by an appropriate exposure method.
  • characteristic of a change pattern with time means a characteristic of a change rate with time.
  • the initial value, the maximum value and / or the minimum value, the time from the exposure to an external stimulus until reaching the maximum value and / or the minimum value, and the change over time are monophasic, biphasic, It means characteristics such as compatibility or other atypical patterns (see Examples).
  • the “dielectric change over time” or “dielectric change over time pattern” used in this specification depends on changes in the dielectric constant in addition to changes in the dielectric constant with time.
  • the change of the rate of change accompanying the time change of the value (for example, the refractive index or the resonance angle) is also included.
  • Analysis of a living cell in this specification can be defined as including various meanings such as evaluation, identification, classification and diagnosis of a living cell.
  • extraction of the characteristics of the temporal change pattern is the characteristics of the measured temporal change pattern among the characteristics of the temporal change pattern as described above. Detection, determination, and / or determination.
  • the image obtained by the CMOS camera was subjected to luminance analysis using Image-Pro (manufactured by Media Cybernetics) corresponding to the image processing software program executed by the computer 8 according to the above embodiment.
  • RBL-2H3 cell (Rat Basophilic Leukemia cell, rat basophilic leukemia cell line) is used as a living cell, and DNP-HSA (Dinitrophenyl-Human Serum Albumin (manufactured by Sigma-Aldrich Japan, Tokyo, Japan)) antigen as an external stimulus.
  • DNP-HSA Dinitrophenyl-Human Serum Albumin (manufactured by Sigma-Aldrich Japan, Tokyo, Japan)) antigen as an external stimulus.
  • DNP-HSA Dinitrophenyl-Human Serum Albumin (manufactured by Sigma-Aldrich Japan, Tokyo, Japan)) antigen as an external stimulus.
  • RBL-2H3 cells have granules containing histamine in the cells and express IgE receptors on the cell surface, the cells can be activated by antigen-IgE stimulation.
  • RBL-2H3 cells were cultured in RPMI (Roswell Park Memorial Institute) medium supplemented with 10% fetal calf serum (FCS), 100 U / ml penicillin and 100 ⁇ g streptomycin, and trypsin was added on the day before the experiment. Used to recover.
  • FCS fetal calf serum
  • the collected RBL-2H3 cells were cultured overnight (37 ° C.) on a sensor chip (gold thin film-deposited glass substrate) in the presence of 50 ng / ml anti-DNP-IgE (Sigma-Aldrich Japan, Tokyo, Japan). ).
  • the chip was mounted on the cell activity analyzer 100 described above, and a running buffer (PIPES buffer) was run. Thereafter, DNP-HSA (50 ng / ml) was injected, and the running buffer was allowed to flow as it was.
  • a running buffer PPES buffer
  • DNP-HSA 50 ng / ml
  • the running buffer was allowed to flow as it was.
  • an intensity image of the reflected light magnified by the objective lens (4 times) was obtained every 10 seconds. It was imaged with a CMOS camera.
  • luminance analysis based on the time change with individual living cells as a selected region was performed from the image by Image-Pro. As a control, imaging and luminance analysis were also performed for those in which DNP-HSA was not injected under the same conditions.
  • FIGS. 4A to 4C are RBL-2H3 cells not stimulated with DNP-HSA
  • FIGS. 5A to 5C are CMOS cameras of RBL-2H3 cells stimulated with DNP-HSA. The image at is shown.
  • the refractive index of each RBL-2H3 cell did not change for 20 minutes when not stimulated with DNP-HSA (antigen).
  • FIG. 5 (A) to FIG. 5 (C) it clearly increased when stimulated with DNP-HSA.
  • FIG. 6A is from the images in FIGS. 4A to 4C
  • FIG. 6B is from the images in FIGS. 5A to 5C.
  • Five RBL-2H3 cells are removed. Changes in luminance values (refractive index) measured using Image-Pro and average values thereof are plotted every 10 seconds.
  • the lower line in the graph of FIG. 6B shows the time during which stimulation is performed by DNP-HSA. For the same graph described below, the line indicates the stimulation time.
  • the refractive index of RBL-2H3 cells hardly changed during 20 minutes not stimulated with DNP-HSA, and obviously increased after stimulation with DNP-HSA.
  • the activity of external stimulation stimulation by, for example, DNP-HSA (antigen)
  • DNP-HSA antigen
  • each individual living cell can be analyzed without labeling with any of the above substances.
  • the intensity of the reflected light that is, the degree of the reaction of the living cells due to the external stimulus is different. This is because the present invention, which can analyze the activity of external stimuli for each individual living cell, can detect with higher sensitivity than the conventional method of evaluating the average value of the stimulus response to a plurality of living cells. Is shown.
  • the cell activity analyzer 100 it is possible to analyze a dielectric constant of a partial region of the cell, that is, a reaction in (a plurality of different) regions in one cell. It is. 9C is similar to FIGS. 4A to 4C, FIG. 5A to FIG. 5C, FIG. 6A, and FIG. 6B in DNP-HSA. This is an example in which the dielectric constant of RBL-2H3 cells, i.e., the cell response, was analyzed, and the activity of external stimuli and the cellular response were analyzed for a plurality of different regions within one cell.
  • PAM212 cells and A431 cells as living cells and EGF (Epidermal® Growth Factor) (10 ng / ml, manufactured by R & D® system, Minneapolis, MN) as external stimuli
  • EGF Epidermal® Growth Factor
  • PAM212 cells mouse keratinocyte cell line
  • A431 cells Human epithelial carcinoma cell line, human squamous cell carcinoma line
  • the method for culturing live cells, imaging, and luminance analysis are the same as those in the RBL-2H3 cells described above, and will not be described.
  • FIG. 13A is an example of an image of a reflection intensity image when a PAM212 cell is stimulated and FIG. FIG. 14A is a graph showing an example of a temporal change in the intensity of reflected light to be measured when the PAM212 cell is stimulated, and FIG.
  • Each of Track 1 to Track 5 shown in FIG. 13 (A) and FIG. 13 (B) shows five living cells or living cell groups selected at random, and the time change of the intensity of these reflected lights is shown in FIG. It is graphed in FIG. 14 (A) and FIG. 14 (B).
  • Track 6 is a background, that is, a place where there are no living cells, and is used for correcting information related to a change in intensity of reflected light to be measured.
  • the thick lines in FIGS. 14A and 14B indicate average values.
  • activation by stimulation can be observed in any kind of living cells, It has been found that the intensity of the reflected light (ie, the dielectric constant) may decrease after activation by stimulation depending on the type of cell or stimulation, compared to before stimulation.
  • both cells have a similar graph pattern, but both cells can be distinguished from the intensity of the reflected light. Was also shown.
  • the measurement target is a large number of living cells as shown in FIGS. 13 (A), 13 (B), 14 (A), and 14 (B) from a part of one living cell as described above. It has also been found that selection and measurement can be made up to the area of the cell group where the cells have gathered.
  • PMA Calbiochem, California, which induces activation of DNP-HSA and RBL-2H3 cells as external stimuli
  • FIG. 15 is a view showing a sensor chip on which RBL-2H3 cells bound with anti-DNP-mouse IgE and RBL-2H3 cells not bound are arranged.
  • RBL-2H3 cells binding anti-DNP-mouse IgE are circular cells previously cultured in Hydrocell (Cellcellse Inc, Tokyo, Japan) to bind IgE.
  • RBL-2H3 cells not bound to anti-DNP-mouse IgE are spindle shaped cells.
  • FIG. 16 is a graph showing an example of a temporal change in intensity of reflected light when stimulated with DNP-HSA and PMA in the sensor chip shown in FIG. a is a graph of RBL-2H3 cells bound with anti-DNP-mouse IgE, and b is a graph of RBL-2H3 cells not bound with anti-DNP-mouse IgE.
  • FIGS. 17A to 17C are examples of temporal changes in the image of the reflection intensity image when the sensor chip shown in FIG. 15 is stimulated with DNP-HSA and PMA.
  • FIG. 16 after stimulation of DNP-HSA, only RBL-2H3 cells bound with anti-DNP-mouse IgE were activated, and RBL-2H3 cells not bound with anti-DNP-mouse IgE were stimulated with PMA. It turns out that it is activated.
  • FIGS. 17 (A) to 17 (C) only DNP-HSA stimulation at 20 min brightly images mainly circular cells (indicated by arrows in the figure), and at 40 min after PMA stimulation. Spindle-shaped cells are also brightly imaged (indicated by arrows in the figure). Note that graphs a and b in FIG. 16 show average values of changes over time in the intensity of reflected light of both cells indicated by these arrows.
  • RBL-2H3 and RBL-3D4 cells are cells established by the present inventors using genetic engineering techniques for RBL-2H3 cells, and express human IgE receptors. Since RBL-2H3 cells are rat-derived cells, only IgE derived from rats and mice can bind. On the other hand, since RBL-3D4 cells also express human IgE receptor, human-derived IgE can be bound and stimulated with anti-human IgE antibody to activate the cells.
  • FIG. 18 is a diagram showing a sensor chip in which RBL-2H3 cells and RBL-3D4 cells are arranged.
  • RBL-2H3 cells are spindle-shaped cells bound with anti-DNP-mouse IgE (anti-DNP mIgE), and RBL-3D4 cells are round-shaped cells bound with human IgE antibody (hIgE).
  • FIG. 19 is a graph showing an example of temporal changes in the intensity of reflected light when the sensor chip shown in FIG. 18 is stimulated with DNP-HSA and an anti-human IgE antibody (manufactured by BETYL, Montgomery, Texas).
  • a is a graph of RBL-2H3 cells
  • b is a graph of RBL-3D4 cells.
  • FIGS. 20 (A) to 20 (C) are diagrams illustrating an example of temporal change in the image of the reflection intensity image when the sensor chip shown in FIG. 18 is stimulated with DNP-HSA and anti-human IgE.
  • the arrow in FIG. 20 (B) shows spindle-shaped RBL-2H3 cells stimulated and activated by DNP-HSA, and the arrow in FIG. 20 (C) is a circle stimulated and activated by anti-human IgE antibody.
  • Shaped RBL-3D4 cells are shown.
  • Graphs a and b in FIG. 19 show the average values of the temporal changes in the intensity of the reflected light of both cells indicated by these arrows. As shown in FIGS.
  • FIG. 21 is a diagram showing a sensor chip in which RBL-2H3 cells and A431 cells (and A431 cell group (A431 cluster)) are arranged.
  • RBL-2H3 cells are spindle-shaped cells with anti-DNP-mouse IgE
  • A431 cells are circular cells.
  • FIGS. 22A and 22B are graphs showing an example of a temporal change in the intensity of reflected light when stimulated with DNP-HSA and EGF in the sensor chip shown in FIG. As shown in FIG. 22, stimulation with DNP-HSA and EGF was performed simultaneously.
  • FIG. 23A and FIG. 23B are diagrams illustrating an example of a temporal change in an image of a reflection intensity image when stimulated with DNP-HSA and EGF in the sensor chip illustrated in FIG.
  • FIG. 23 (A) shows an image (cont) before giving a stimulus
  • FIG. 23 (B) shows an image while giving a stimulus with DNP-HSA and EGF 30 minutes later.
  • the arrow in a figure shows five cells selected at random among each cell.
  • a triangular arrowhead indicates RBL-2H3 cells
  • a square shape indicates A431 cells.
  • a is a graph showing changes in these five RBL-2H3 cells
  • b is a graph showing changes in five A431 cells.
  • FIG. 22A it can be seen that even in the same type of cells, there is a difference in the temporal change of the intensity of the reflected light, but a and b are clearly different graph patterns.
  • a represents the average value of changes in RBL-2H3 cells
  • b represents the average value of changes in A431 cells
  • c represents the average value of RBL-2H3 cells and A431 cells
  • d represents The average value in the whole image is shown.
  • the graph of a RBL-2H3 cell average
  • the intensity of the reflected light remains increased over time.
  • the graph of b (A431 cell average) is lower than that before stimulation after a certain amount of time has passed. Therefore, the change of the graph of d which is the average value of the whole image has become weak.
  • Example 2 In Example 2, the change in resonance angle over time when EGF stimulation is performed on CHO (Chinese Hamster Ovary) cells in which wild-type human EGFR is forcibly expressed will be described in detail.
  • EGFR is well known to exhibit important functions in the proliferation of various cells in the body and the development / formation of organs, and is also known to be overexpressed in various cancer cells.
  • an empty vector pCMV-Tag4 (manufactured by Stratagene) and a vector in which wild-type human EGFR gene is incorporated into pCMV-Tag4 are cultivated in Ham's F-12 containing 10% fetal calf serum (FCS, Fetal calf serum). Genes were introduced into CHO cells maintained in solution using electroporation. The wild-type human EGFR gene incorporated into pCMV-Tag4 was amplified by PCR using the forward primer and reverse primer of the base sequences shown in SEQ ID NO: 1 and SEQ ID NO: 2 in the sequence listing. After gene introduction, drug selection was performed in the presence of 10 mg / ml G418.
  • a CHO cell line introduced with an empty vector and a CHO cell line stably expressing wild-type human EGFR at a high level are dropped on the sensor chip so as to be 1.2 ⁇ 10 4 cells / 60 ⁇ l, and cultured overnight. did.
  • the cells on the sensor chip were perfused with Hepes buffer, stimulated with 10 ng / ml recombinant human EGF (hEGF, manufactured by R & D) for 10 minutes, and changes in the resonance angle over time after stimulation were measured using SPR- It measured using CELLIA (Mortex company make).
  • hEGF human EGF
  • CELLIA CELLIA
  • FIG. 24 is a graph showing changes in resonance angle over time by EGF stimulation in CHO cells in which wild-type human EGFR according to Example 2 was forcibly expressed.
  • the vertical axis indicates the change in resonance angle (Change of AR (Angle of Resonance) (degree)) by the SPR device, and the horizontal axis indicates the time (Time (sec)) from the EGF stimulation.
  • Change of AR Angle of Resonance
  • time Time
  • the wild-type human EGFR-expressing CHO cell line As shown in FIG. 24, when compared with the CHO cell line (mock) into which an empty vector was introduced, the wild-type human EGFR-expressing CHO cell line (EGFR-WT) changes with time in different resonance angles with respect to EGF stimulation. It turns out that it shows a pattern. Specifically, it was confirmed that the wild-type human EGFR-expressing CHO cell line exhibits a typical three-phase resonance angle change pattern over time (a fluctuation pattern that rises from the initial level and rises again after rising). It was.
  • Example 3 In this Example 3, an example relating to EGF stimulation in a CHO cell line expressing human EGFR with a mutated ATP binding domain will be described in detail.
  • EGFR-K721M Choen, WS, Lazar, CS, Poenie, M., Tsien, RY, Gill, GN, Rosenfeld. MG, 1987, Nature 28 328 (6133), 820-823. See)
  • Genes were generated using QuickChange® Site® Directed® Mutagenesis® Kit (Stratagene) and amplified by PCR.
  • the base sequences of the forward primer and reverse primer used in the PCR are shown in SEQ ID NO: 3 and SEQ ID NO: 4 in the sequence listing.
  • the prepared ATP-binding domain mutant EGFR gene was introduced into CHO cells by electroporation in the same manner as described in Example 1, and drug selection was performed to obtain a stable expression cell line.
  • the wild-type human EGFR-expressing CHO cell line of Example 2 and the ATP-binding domain mutant EGFR-expressing CHO cell line described above were seeded in a 6-well plate at a concentration of 0.2 ⁇ 10 6 / ml. Cultured overnight in Ham's F-12 culture medium containing 1% fetal bovine serum. Thereafter, the sample was prepared by stimulation with 10 ng / ml hEGF (R & D) for 5 minutes, treatment with a cell lysate containing 1% NP-40, and separation by SDS-PAGE (SDS-polyacrylamide gel electrophoresis). .
  • FIG. 25 is a diagram showing the results of Western blotting using anti-phosphorylation specific EGFR antibody, anti-EGFR antibody and anti-FLAG antibody in CHO cells expressing human EGFR with a mutated ATP binding domain according to Example 2. is there.
  • ATP-binding domain mutant EGFR-expressing CHO cells (K721M) were able to confirm the expression of EGFR protein at an equivalent level or higher when compared to wild-type human EGFR-expressing CHO cells (WT). Tyrosine phosphorylation hardly occurred even after stimulation.
  • FIG. 26 is a diagram showing measurement results of the expression level of EGFR on the surface of CHO cells in which human EGFR having a mutated ATP binding domain according to Example 3 was expressed.
  • the black-colored ones indicate isotype control antibodies (Control), and the solid-line ones indicate the EGFR cell surface expression level (EGFR-WT) in wild-type human EGFR-expressing CHO cells.
  • the dotted line indicates the EGFR cell surface expression level (EGFR-K721M) in the ATP-binding domain mutant EGFR-expressing CHO cells.
  • the cell surface expression level was equivalent to the cell surface expression level of wild-type human EGFR-expressing CHO cells.
  • the present inventors seeded the aforementioned ATP-binding domain mutant EGFR-expressing CHO cell line and wild-type human EGFR-expressing CHO cell line on a sensor chip, and the next day, 10 ng / ml hEGF (manufactured by R & D) was used. The sample was stimulated for 10 minutes, and the change in resonance angle over time was measured with SPR-CELLIA (Mortex).
  • FIG. 27 is a diagram showing changes in resonance angle over time by EGF stimulation in CHO cells in which human EGFR with a mutated ATP binding domain according to Example 2 was expressed.
  • the wild-type human EGFR-expressing CHO cell line (EGFR-WT) showed a typical three-phase change in resonance angle similar to the result of Example 1.
  • the resonance angle was hardly changed by stimulation with EGF.
  • Example 4 In Example 4, examples relating to changes in resonance angle with respect to EGF stimulation of various cancer cell lines will be described in detail.
  • gastric cancer cell lines MKN-1, MKN-7 and MK28
  • prostate cancer cell lines DU145 and LNCap
  • FIG. 28 is a graph showing changes in the resonance angle over time by EGF stimulation in the gastric cancer cell line MKN-1 according to Example 4.
  • FIG. 29 is a graph showing changes in resonance angle over time by EGF stimulation in gastric cancer cell line MKN-7 according to Example 4.
  • FIG. 30 is a diagram showing changes in the resonance angle over time by EGF stimulation in the gastric cancer cell line MK28 according to Example 4.
  • FIG. 31 is a graph showing changes in resonance angle over time by EGF stimulation in prostate cancer cell line DU145 according to Example 4.
  • FIG. FIG. 32 is a diagram showing changes in resonance angle over time by EGF stimulation in the prostate cancer cell line LNCap according to Example 4.
  • the biphasic change pattern of resonance angle over time (1) rises and (2) falls by EGF stimulation. A fluctuation pattern was observed.
  • FIGS. 31 and 32 in the prostate cancer cell lines (DU145 and LNCap), the change pattern of the uniphasic resonance angle over time by stimulation with EGF (FIG. 31 shows the change pattern of (1) rise only) In FIG. 32, (2) fluctuation pattern of only falling) was observed.
  • the prostate cancer cell line LNCap did not contain a phase with increasing resonance angle, unlike other cell lines.
  • Example 4 From the results of this Example 4 and the typical three-phase resonance angle change pattern of the wild-type human EGFR-expressing CHO cell line described in Example 1 and Example 2, cancers against normal cells
  • the characteristics of the change pattern of the resonance angle of the cell over time that is, the change pattern of the dielectric constant over time, as well as the type of cancer cell line as well as the cell (for example, monophasic, biphasic, triphasic)
  • the comprehensive state of living cells can be diagnosed and analyzed by distinguishing by other atypical patterns or the like.
  • Example 5 an example relating to the relationship between the human hemangiosarcoma cell line and the change in resonance angle with respect to EGFR and EGF stimulation will be described in detail.
  • human vascular sarcoma cells which are human soft tissue sarcoma cells, and EGFR
  • EJSO European Journal of Surgical Oncology
  • a human hemangiosarcoma cell line (ISO-HAS) is suspended in DMEM (Dulbecco's Modified Eagle Medium) containing 15% FCS, seeded on a 6-well plate at a concentration of 0.2 ⁇ 10 6 / ml, and overnight. Culture was performed. Thereafter, stimulation with hEGF (final concentration of 100 ng / ml, using R & D) for 0 to 15 minutes, treatment with cell lysate, anti-phosphorylation specific EGFR antibody (Cell signaling) and anti-EGFR antibody ( Western blotting was performed in the same manner as described in Example 3 and Example 4 using Cell Signaling).
  • the ISO-HAS cells were seeded on a sensor chip, stimulated with hEGF (10 ng / ml, manufactured by R & D) on the next day, and the resonance angle over time was the same as in Examples 3 and 4 described above.
  • SPR-CELLIA SPR-CELLIA (Mortex).
  • FIG. 33 (A) shows the results of western blotting of the human hemangiosarcoma cell line according to Example 5 using anti-phosphorylation-specific EGFR antibody and anti-EGFR antibody.
  • FIG. 33 (B) is a graph showing changes in the resonance angle over time by EGF stimulation in the human hemangiosarcoma cell line according to Example 5.
  • the upper graph shows the case where EGF stimulation is performed on ISO-HAS cells (EGF) and the case where EGF stimulation is not performed on ISO-HAS cells.
  • the change in resonance angle over time due to (Control) is shown, and the lower graph shows the difference in change in resonance angle over time.
  • the human hemangiosarcoma cell line (ISO-HAS) according to Example 5 expresses EGFR and phosphorylates EGFR upon stimulation with EGF at a concentration of 100 ng / ml.
  • EGF EGF at a concentration of 100 ng / ml.
  • FIG. 33 (B) in the human hemangiosarcoma cell line (ISO-HAS) according to Example 5, an atypical and temporal change pattern of the resonance angle ((1) rising by EGF stimulation) (2) Random (atypical fluctuation patterns) that fell incompletely and fluctuated up and down were observed.
  • a sample containing living cells can be used as an object of analysis of the present invention as it is.
  • the possibility that the contained cells can be evaluated and analyzed is also suggested. Specifically, for example, it is suggested that by collecting a body fluid such as blood from a subject in advance and directly analyzing it with an SPR device, it may lead to a technique that can evaluate whether or not any tumor / cancer cell is included.
  • a cell activity analyzing apparatus and a cell activity analyzing method capable of analyzing the activity of external stimuli for individual living cells.
  • these cell activity analyzers and cell activity analysis methods it is possible to evaluate and analyze the properties of individual living cells and / or a part of each living cell, or to isolate specific types of living cells. it can.
  • live cells with a specific activity are screened, specific biomolecules related to cell activity (external stimuli) are screened, and further, in which part of live cells the activation occurs mainly Can be used for research on various cells.
  • a medical diagnostic apparatus for example, a high-throughput allergy diagnostic apparatus.
  • the present inventors have clarified that normal cells and cancer cells exhibit characteristics of temporal change patterns of different resonance angles with respect to EGF stimulation using a surface plasmon resonance device (SPR device). Furthermore, it was also shown that different cancer and cancer cell line types exhibit different resonance angle characteristics over time for EGF stimulation. In addition, it was confirmed that the human hemangiosarcoma cell line also has a characteristic of a temporal change pattern of different resonance angles with respect to EGF stimulation.
  • SPR device surface plasmon resonance device
  • the state of cells can be comprehensively and directly analyzed at the level of individual living cells.
  • it is a technology that can analyze cell status, cancer cells, types of cancer and types of cancer cell lines without fixing cells or tissues and evaluating malignant tumors as a potential possibility. High value.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Toxicology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Investigating Or Analysing Materials By Optical Means (AREA)

Abstract

L'invention porte sur un dispositif d'analyse d'activité de cellules (100), qui comporte : un film mince en métal (5) venant en contact avec des cellules vivantes (C1, C2); un prisme (3) comportant une interface (F) sensiblement en contact avec le film mince en métal (5); une source de lumière (1) qui irradie le prisme (3) à l'aide des faisceaux parallèles de lumière polarisée (P), et qui irradie ensuite l'interface (F) selon un angle d'incidence prescrit qui induit un phénomène de résonance plasmonique de surface; un objectif (6) qui agrandit d'un rapport prescrit une image d'intensité correspondant à la distribution d'intensité en deux dimensions de la lumière réfléchie à partir de ladite interface (F); une unité de réalisation d'image (7) qui réalise une image de l'image d'intensité agrandie; une unité d'acquisition d'image (21) qui échantillonne les données d'image de l'image d'intensité; une unité d'affichage (23) et une unité d'actionnement (24) pour sélectionner pour la mesure l'image d'au moins une partie des cellules vivantes (C1, C2) à partir des données d'image de l'image d'intensité; et une unité de traitement d'image (22) qui extrait des valeurs de luminosité de l'objet de mesure, et qui, sur la base de changements de la valeur de luminosité de l'objet de mesure avant et après l'application d'un stimulus externe aux cellules vivantes (C1, C2), calcule des informations associées au changement d'intensité de la lumière réfléchie à partir de l'objet de mesure.
PCT/JP2011/056304 2010-03-17 2011-03-16 Dispositif d'analyse d'activité de cellules, procédé d'analyse d'activité de cellules et procédé d'analyse de cellules WO2011115189A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2010-061710 2010-03-17
JP2010061710A JP2011193752A (ja) 2010-03-17 2010-03-17 細胞活性分析装置及び細胞活性分析方法、並びに細胞同定方法
JP2010-278131 2010-12-14
JP2010278131 2010-12-14

Publications (1)

Publication Number Publication Date
WO2011115189A1 true WO2011115189A1 (fr) 2011-09-22

Family

ID=44649272

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2011/056304 WO2011115189A1 (fr) 2010-03-17 2011-03-16 Dispositif d'analyse d'activité de cellules, procédé d'analyse d'activité de cellules et procédé d'analyse de cellules

Country Status (1)

Country Link
WO (1) WO2011115189A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPWO2018062215A1 (ja) * 2016-09-30 2019-07-18 オリンパス株式会社 観察装置
US10739260B2 (en) 2015-01-26 2020-08-11 Hitachi High-Tech Corporation Optical analyzing device
US11460682B2 (en) 2017-05-29 2022-10-04 Evident Corporation Observation device
WO2023166655A1 (fr) * 2022-03-03 2023-09-07 オリンパス株式会社 Unité d'éclairage et système d'imagerie

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
MASAE HORII ET AL.: "Response measurement of individual mast cells using a 2D-SPR sensor and its application to allergen sensing", CHEM. SENS., vol. 24, no. SUPPLE, 29 March 2008 (2008-03-29), pages 145 - 147 *
MICHIHIRO HIDE ET AL.: "Hyomen Plasmon Kyomei Biosensor ni yoru Saibo Kino Sokutei Gijutsu no Kaihatsu", ANNUAL REPORT, NAKATANI FOUNDATION OF ELECTRONIC MEASURING TECHNOLOGY ADVANCEMENT, vol. 18, 15 August 2004 (2004-08-15), pages 66 - 70 *
YASUNORI IRIBE ET AL.: "Nijigen Imaging ni yoru Mouse B, T Lymph-kyu no Saibo Oto Kenshutsu", ABSTRACTS OF AUTUMN MEETING OF THE ELECTROCHEMICAL SOCIETY OF JAPAN, vol. 2009, 10 September 2009 (2009-09-10), pages 88 *
YASUNORI IRIBE ET AL.: "Nijigen SPR Sensor ni yoru Kogen Ketsugo Ji no B Lymph-kyu Oto no Kenshutsu", SOCIETY FOR CHEMISTRY AND MICRO- NANO SYSTEMS KOEN YOSHISHU, vol. 17TH, 20 May 2008 (2008-05-20), pages 40 *
YUKI YANASE ET AL.: "Imaging SPR o Riyo shita 1 Saibo Oto Kaiseki", EXTENDED ABSTRACTS, JAPAN SOCIETY OF APPLIED PHYSICS AND RELATED SOCIETIES, vol. 57, 3 March 2010 (2010-03-03) *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10739260B2 (en) 2015-01-26 2020-08-11 Hitachi High-Tech Corporation Optical analyzing device
JPWO2018062215A1 (ja) * 2016-09-30 2019-07-18 オリンパス株式会社 観察装置
US11226476B2 (en) 2016-09-30 2022-01-18 Olympus Corporation Specimen observation apparatus
US11460682B2 (en) 2017-05-29 2022-10-04 Evident Corporation Observation device
WO2023166655A1 (fr) * 2022-03-03 2023-09-07 オリンパス株式会社 Unité d'éclairage et système d'imagerie

Similar Documents

Publication Publication Date Title
DK2271657T3 (en) GEL-MICRO-DROP COMPOSITION AND METHOD OF USING IT
JP2011193752A (ja) 細胞活性分析装置及び細胞活性分析方法、並びに細胞同定方法
JP2020511126A (ja) 生物細胞を研究するための方法及びシステム
US20130252848A1 (en) Cellomics system
JP2013531787A (ja) 粒子の運動度および/または細胞の分散を求めるためのホログラフィック変動顕微鏡装置および方法
JP5953293B2 (ja) 発光タンパクによる長期モニタリング方法および解析方法
CN102667484A (zh) 识别黑素瘤肿瘤细胞的生物标记
JP2013531787A5 (ja) 粒子の運動度および/または細胞の分散を求めるためのホログラフィック変動顕微鏡装置、方法並びにプログラム
CN107209178A (zh) 基于拴系颗粒的生物传感器
US20220299421A1 (en) Systems, devices and methods for three-dimensional imaging of moving particles
WO2011115189A1 (fr) Dispositif d'analyse d'activité de cellules, procédé d'analyse d'activité de cellules et procédé d'analyse de cellules
Li et al. AFM analysis of the multiple types of molecular interactions involved in rituximab lymphoma therapy on patient tumor cells and NK cells
CN103913444A (zh) 基于蓝光光镊的单光子激发荧光多通道定量检测装置及检测方法
EP2972337B1 (fr) Technique d'imagerie nanoplasmonique pour la cartographie spatio-temporelle de sécrétions cellulaires isolées en temps réel
JP7140103B2 (ja) 治療有効性の予測方法
US8709828B2 (en) Method for the analysis of solid objects
JP2018526625A (ja) 血液細胞の細胞表面受容体の評価方法
JP2004271337A (ja) 表面プラズモン共鳴現象を利用した細胞の多検体同時解析装置
Chen et al. Cellular analyses for label-free and rapid HER2-positive cancer diagnosis based on SPRi-modified with nanobody
WO2009110614A1 (fr) Procédé de mesure de la fonction de cellules effectrices, kit de mesure et système de mesure
JP2010537195A (ja) バイオセンサ抗体機能マッピング
CN108693144B (zh) 基于sprm技术进行复用性单细胞蛋白质组学监测方法
Gora et al. Analysis of the H-Ras mobility pattern in vivo shows cellular heterogeneity inside epidermal tissue
JP4336812B2 (ja) 細胞に発現する表面抗原を迅速に同定するための分析方法
JP2009232736A (ja) 補体活性検査方法

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11756373

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11756373

Country of ref document: EP

Kind code of ref document: A1