CN114113191A - Tumor tissue magnetic imaging method with micron resolution - Google Patents

Tumor tissue magnetic imaging method with micron resolution Download PDF

Info

Publication number
CN114113191A
CN114113191A CN202111515040.6A CN202111515040A CN114113191A CN 114113191 A CN114113191 A CN 114113191A CN 202111515040 A CN202111515040 A CN 202111515040A CN 114113191 A CN114113191 A CN 114113191A
Authority
CN
China
Prior art keywords
diamond
tissue sample
tumor tissue
cover glass
tumor
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN202111515040.6A
Other languages
Chinese (zh)
Inventor
陈三友
李万和
石发展
杜江峰
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Science and Technology of China USTC
Original Assignee
University of Science and Technology of China USTC
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
Application filed by University of Science and Technology of China USTC filed Critical University of Science and Technology of China USTC
Priority to CN202111515040.6A priority Critical patent/CN114113191A/en
Publication of CN114113191A publication Critical patent/CN114113191A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N24/00Investigating or analyzing materials by the use of nuclear magnetic resonance, electron paramagnetic resonance or other spin effects
    • G01N24/10Investigating or analyzing materials by the use of nuclear magnetic resonance, electron paramagnetic resonance or other spin effects by using electron paramagnetic resonance

Landscapes

  • Physics & Mathematics (AREA)
  • High Energy & Nuclear Physics (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Investigating, Analyzing Materials By Fluorescence Or Luminescence (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

The invention provides a tumor tissue magnetic imaging method with micron resolution, which comprises the following steps: tagging a tissue sample with superparamagnetic nanoparticles, wherein the superparamagnetic nanoparticles are tagged on a tumor marker protein of the tissue sample; attaching diamond to the tissue sample to form a diamond-tumor tissue slice-cover glass structure; carrying out magnetic field measurement on the tissue sample by using an NV color center two-dimensional magnetic sensor in the diamond to obtain a magnetic field image of the tissue sample; and processing the magnetic field image through a deep learning model to obtain a micron resolution magnetic imaging image of the tissue sample. The tumor tissue magnetic imaging method provided by the invention has the advantages of high signal stability, low background, capability of realizing absolute quantification, capability of realizing magnetic and optical multi-mode correlation imaging of biological tissues, capability of realizing micron-sized spatial resolution of both magnetic and optical imaging, compatibility with a commercial optical microscope and easiness in popularization and application.

Description

Tumor tissue magnetic imaging method with micron resolution
Technical Field
The invention relates to the field of imaging, in particular to a tumor tissue magnetic imaging method with micron resolution.
Background
Cancer is one of the major diseases affecting human health, and the molecular mechanism research and accurate diagnosis of cancer are the basis for effective prevention and treatment of tumor diseases. The traditional tissue imaging methods mainly comprise HE staining, immunohistochemistry, immunofluorescence and the like. Multispectral imaging methods based on tyramine signal amplification, multichannel imaging based on mass spectrometric detection, and some other label-free spectroscopic imaging methods have also emerged in recent years.
Conventional optical imaging methods often suffer from background optical signals and signal instability and give only relative quantitative information of tumor markers, whereas various optical methods cannot be used simultaneously on the same piece of tissue, for example:
hematoxylin-eosin (HE) staining is chemical staining mainly based on differential combination of different cell components to dyes, diagnosis standards are different from person to person and depend on experience comparatively, and the HE staining lacks tumor marker information because of no specific immune marker, and the HE staining forms obvious color signals and simultaneously generates a large amount of autofluorescence, so that the HE staining cannot be used with optical methods such as immune tissues, immunofluorescence and the like.
Immunohistochemistry (IHC) can be divided into immunohistochemistry and immunofluorescence. Immunohistochemistry combines the specificity of immunoreaction and the visibility of histochemistry, images various antigen substances at the level of cells and subcells by means of an optical microscope, has the advantages of simple operation, stable signals, long-term preservation of samples, low cost and the like, but cannot perform accurate quantitative analysis of tumor markers, can only perform relative quantitative classification, depends on the experience of clinical pathologists, and is difficult to realize multi-target point common labeling; in tissues such as glands, livers and the like, due to the influence of endogenous enzyme activity, nonspecific binding or high background staining is easy to occur, so that judgment is interfered; in addition, immunohistochemistry is difficult to use with other optical methods on the same tissue section.
The immunofluorescence technique uses fluorescent molecules to label antigens, and can realize simultaneous labeling and multi-channel imaging of a plurality of tumor markers through spectral discrimination, but the immunofluorescence technique also has obvious defects, such as instability of fluorescence signals, background fluorescence in tissues, incapability of absolute quantification caused by dimensionless characteristics of fluorescence signal intensity, incapability of being used together with HE staining, immunohistochemistry and the like.
Conventional magnetic resonance imaging is widely used in clinical medicine, but its application at the tissue level is limited due to the low spatial resolution. Recently developed magnetic resonance techniques based on nitrogen vacancy colour centres (NV colour centres) in diamond provide a very attractive microscopic magnetic approach. The NV color center is used as a quantum magnetic sensor, can detect the magnetic field in a nearby sample with high sensitivity, high resolution and high stability, and is very suitable for detecting biological samples due to the good biocompatibility of diamond materials. Magnetic imaging of biological samples from nanometer to micrometer resolution has been achieved using NV colour centers, but due to some technical obstacles magnetic imaging (or magnetic resonance imaging) at tissue level with micrometer or sub-cellular resolution has not been achieved.
Disclosure of Invention
In view of the above, the main object of the present invention is to provide a solution to at least one of the above mentioned technical problems.
To achieve the above object, as one aspect of the present invention, there is provided a method for magnetic imaging of tumor tissue with micron resolution, comprising:
tagging a tissue sample with superparamagnetic nanoparticles, wherein the superparamagnetic nanoparticles are tagged on a tumor marker protein of the tissue sample;
attaching diamond to the tissue sample to form a diamond-tumor tissue section-cover glass structure, wherein the tumor tissue section belongs to the tissue sample;
carrying out magnetic field measurement on the tissue sample by using an NV color center two-dimensional magnetic sensor in the diamond to obtain a magnetic field image of the tissue sample; and
and processing the magnetic field image through a deep learning model to obtain a micron-sized resolution magnetic imaging image of the tissue sample.
According to an embodiment of the invention, said tagging of said tissue sample by superparamagnetic nanoparticles further comprises:
and carrying out immunomagnetic labeling treatment on the tissue sample, wherein the immunomagnetic labeling comprises a superparamagnetic nanoparticle incubation process so as to enable the tumor marker protein of the tissue sample to be specifically labeled and combined with the superparamagnetic nanoparticle.
According to an embodiment of the invention, further comprising:
treating the diamond to form a high density NV color center having a depth of 10nm to 110nm in a first surface of the diamond, the first surface of the diamond in contact with the tissue slice.
According to an embodiment of the invention, the attaching the diamond to the tissue sample further comprises:
dripping ultraviolet glue into the region to be detected of the tumor tissue slice in the diamond-tumor tissue slice-cover glass structure so as to enable the upper surface of the diamond to contact the region to be detected of the tumor tissue slice; and
and uniformly applying force to the diamond-tumor tissue slice-cover glass structure by using a metal clamp, wherein the force application direction is vertical to the second surface of the diamond and the cover glass surface which are in contact with the metal clamp, so that the pretreated tissue slice and the diamond are tightly attached, and the second surface of the diamond is a surface opposite to the first surface of the diamond.
According to an embodiment of the invention, further comprising:
integrally attaching the diamond-tumor tissue slice-cover glass structure to a dove prism, wherein the lower surface of the diamond is placed on the dove prism;
ultraviolet glue is used between the diamond-tumor tissue slice-cover glass structure and the dove prism for pasting; wherein the diamond-tumor tissue slice-cover glass structure is placed in the center of the dove prism; and
and (3) properly pressing the diamond-tumor tissue slice-cover glass structure to ensure that the diamond-tumor tissue slice-cover glass structure is attached to the dove prism in parallel.
According to an embodiment of the invention, the superparamagnetic nanoparticle labeled tissue sample further comprises:
and (3) pretreating the cover glass by polylysine before the operation of marking the tissue sample by the superparamagnetic nanoparticles, and placing the tumor tissue section on the cover glass so as to fix the tumor tissue section on the cover glass.
According to an embodiment of the invention, further comprising:
refracting laser light through the dove prism onto the diamond so as to excite an NV color center in the diamond.
According to an embodiment of the invention, further comprising:
the superparamagnetic nanoparticles are magnetized by an external magnetic field.
According to an embodiment of the invention, further comprising:
and regulating the spin state of the NV color center in the diamond by using microwaves.
According to an embodiment of the invention, further comprising:
and acquiring fluorescence data of the NV color center in the diamond by using a CMOS camera to obtain a magnetic field image.
According to an embodiment of the invention, further comprising:
the tissue sample to be tested may be a biological tissue sample of another type than a tumor tissue sample.
The tumor tissue magnetic imaging method provided by the invention has the advantages of high signal stability, low background, capability of realizing absolute quantification, capability of realizing magnetic and optical multi-mode correlation imaging of biological tissues, capability of realizing micron-sized spatial resolution of both magnetic and optical imaging, compatibility with a commercial optical microscope and easiness in popularization and application.
Drawings
The above and other objects, features and advantages of the present invention will become more apparent from the following description of the embodiments of the present invention with reference to the accompanying drawings, in which:
FIG. 1 schematically illustrates a flow diagram of an embodiment of the invention;
FIG. 2 is a schematic diagram showing the labeling of tumor marker proteins by superparamagnetic particles under the magnetic imaging method of tumor tissue of the present invention;
FIG. 3 schematically illustrates a schematic diagram of an NV color center two-dimensional magnetic sensor in a diamond for use in the present invention;
FIG. 4 schematically illustrates a schematic diagram of the diamond-neoplasmic tissue slice-cover glass structure of the present invention attached to a dove prism;
FIG. 5 schematically shows a schematic diagram of the weak magnetic field generated after the superparamagnetic nanoparticles used in the present invention are magnetized;
fig. 6 schematically shows a schematic view of an apparatus for imaging using a magnetic imaging method of tumor tissue according to the present invention.
Detailed Description
In order that the objects, technical solutions and advantages of the present invention will become more apparent, the present invention will be further described in detail with reference to the accompanying drawings in conjunction with the following specific embodiments.
The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. The terms "comprises," "comprising," and the like, as used herein, specify the presence of stated features, steps, operations, and/or components, but do not preclude the presence or addition of one or more other features, steps, operations, or components.
All terms (including technical and scientific terms) used herein have the same meaning as commonly understood by one of ordinary skill in the art unless otherwise defined. It is noted that the terms used herein should be interpreted as having a meaning that is consistent with the context of this specification and should not be interpreted in an idealized or overly formal sense.
Where a convention analogous to "at least one of A, B and C, etc." is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., "a system having at least one of A, B and C" would include but not be limited to systems that have a alone, B alone, C alone, a and B together, a and C together, B and C together, and/or A, B, C together, etc.).
The invention provides a tumor tissue magnetic imaging method, and a flow chart of an embodiment of the invention is schematically shown in FIG. 1.
As shown in FIG. 1, the method includes operations S101-104.
In operation S101, a tissue sample is labeled with superparamagnetic nanoparticles, wherein the superparamagnetic nanoparticles are labeled on a tumor marker protein of the tissue sample.
In operation S102, attaching a diamond to the tissue slice to form a diamond-tumor tissue slice-cover glass structure; wherein, the conventional glass slide is too thick, which can affect the microwave application and light transmission effect, and the thickness of the cover glass is smaller than that of the conventional glass slide, so that the tumor tissue section is attached on the cover glass for subsequent imaging.
In operation S103, a magnetic field measurement is performed on the tissue sample using an NV color center two-dimensional magnetic sensor in the diamond, resulting in a magnetic field image of the tissue sample.
In operation S104, the magnetic field image is processed through a deep learning model to obtain a magnetic imaging map of the tissue sample.
According to an embodiment of the present invention, before performing operation S101, the cover glass is pretreated by polylysine, and the tumor tissue section is placed on the cover glass so that the tumor tissue section is fixed on the cover glass.
More specifically, the tissue sample can be prepared by a process of frozen section or paraffin embedded section; for the frozen section, carrying out frozen section by using the conventional OCT embedded tumor tissue, and selecting the section thickness of 5-10 mu m; for paraffin embedded sections, a tissue sample needs to be subjected to conventional fixing, dehydration and embedding, then is sliced, the thickness of the section is selected to be 5 mu m, and the paraffin embedded section also needs to be subjected to antigen retrieval to carry out tumor tissue magnetic imaging operation; the tumor tissue section is attached to the cover glass, the thickness of the cover glass can be 0.16-0.19 mm, the cover glass needs to be pretreated by polylysine, and the tumor tissue section can be fixed on the cover glass only by soaking the cover glass for half an hour by using 0.1mg/ml poly-L-lysine.
According to one embodiment of the present invention, the tissue sample to be tested may be a biological tissue sample of another type than a tumor tissue sample.
More specifically, other types of biological tissue samples need to contain corresponding biomarkers, the biomarkers can be characteristic proteins, the corresponding biomarkers can be subjected to immunomagnetic labeling, and other types of biological tissue samples can be subjected to magnetic imaging by using the micron-resolution tumor tissue magnetic imaging method disclosed by the invention.
Fig. 2 schematically shows a schematic diagram of tumor marker proteins labeled by superparamagnetic particles under a tumor tissue magnetic imaging method according to an embodiment of the present invention. The preparation process of the immunomagnetic labeling tissue sample comprises the following steps:
(1) prepared tissue samples were placed in petri dishes or wet boxes and washed 3 times with Phosphate Buffered Saline (PBS).
(2) Fixing: fixation was done in 4% Paraformaldehyde (PFA) for 15 minutes at room temperature or 5 minutes with 100% methanol prefreezed at-20 ℃.
(3) The cells were washed 3 times with 5 minutes of Phosphate Buffered Saline (PBS) for 15 minutes.
(4) And (3) sealing: sealing for 30 minutes at room temperature, wherein the formula of the sealing liquid is as follows: 2% Bovine Serum Albumin (BSA), 0.1% Triton X-100(Triton X-100), 1 XPBS, and the pH of the blocking solution was maintained at 7.4.
(5) Primary antibody incubation: the blocked tissue samples were incubated in primary antibody solution overnight at 4 ℃. The antibody incubation liquid formula is as follows: 1% BSA, 0.1% Triton X-100,1 XPBS, pH 7.4.
(6) The following day, the cells were washed 5 times with Phosphate Buffered Saline (PBS) for 5 minutes each, and the total time was 25 minutes.
(7) Biotin-secondary antibody incubation: tissue samples were incubated in biotin (biotin) -modified secondary antibody solutions for 1 hour at room temperature. The biotin secondary antibody can be goat anti-mouse IgG H & L (Biotin) (Abcam, ab 6788). The concentration of the antibody working solution is 2 mug/ml, namely 1:1000 dilution. The antibody incubation liquid formula is as follows: 1% BSA, 0.1% Triton X-100,1 XPBS, pH 7.4.
(8) The cells were washed 5 times with 5 min Phosphate Buffered Saline (PBS) and 25 min.
After the operations (1) to (8) are finished, a labeled intermediate product is obtained, wherein the tumor marker protein of the tissue sample is combined with the primary antibody factor, the primary antibody factor is combined with the secondary antibody factor, and the secondary antibody factor is modified with biotin.
(9) Incubation of magnetic particles: tissue samples were incubated in streptavidin (streptavidin) coated superparamagnetic nanoparticle solution for 30 min at room temperature. Streptavidin (Streptavidin) -coated superparamagnetic nanoparticles such as Streptavidin-coated superparamagnetic MNPs (Ocean Nanotech,20-nm magnetic core, SHS-20-05) with a magnetic particle working solution concentration of 50 μ g Fe/ml, i.e., 1:20 dilution. The formula of the incubation liquid is as follows: 0.1% Triton X-100,1 XPBS, pH 7.4.
(10) Changing and washing 5 times with Phosphate Buffer Solution (PBS), 5 minutes each time, and recording for 25 minutes; during this time, the nuclei were stained with 4', 6-diamidino-2-phenylindole (DAPI) for 5 minutes, and optionally, the nuclei were stained with 4', 6-diamidino-2-phenylindole (DAPI) for 5 minutes at the 3 rd or 4 th time of the Phosphate Buffered Saline (PBS) change-over to make the nuclei sufficiently stained.
After the above operations (1) - (10) are finished, a schematic diagram of the tumor marker protein marked by the superparamagnetic particles is obtained as shown in fig. 2, in which the superparamagnetic nanoparticles are coated in streptavidin (streptavidin), and the streptavidin (streptavidin) and the biotin modified on the secondary antibody are specifically combined together through an antibody-antigen reaction, so that the superparamagnetic nanoparticles are marked on the tumor marker protein of the tissue sample.
According to one embodiment of the present invention, the diamond used in operation S102 may be a diamond having a high density NV color center formed in the first surface to a depth of 10nm to 110 nm.
More specifically, fig. 3 schematically shows a schematic diagram of a NV colour center two-dimensional magnetic sensor in a diamond for use in the present invention, where the first plane is set as xoy plane and the z-direction is set perpendicular to the xoy plane; purity of electronic grade and [100 ]]14N implantation into bulk diamond in crystal orientation+Ions are implanted into the diamond by 5 different energies with different ion concentrations to form 5 ion layers which are mutually connected, and the ion concentration implanted into each layer is 10^13/cm2Magnitude; passing through 4 small holes at 1000 deg.CAfter annealing, a high density of NV color centers is formed to a depth of 10nm to 110nm in the first surface of the diamond, the concentration of the NV color centers being about 1 x 10^12/cm2. The NV color center two-dimensional magnetic sensor in the diamond has better magnetic measurement sensitivity and measurement efficiency, and meanwhile, the thickness in the z direction of 100nm does not influence the realization of micron resolution.
In operation S102, according to an embodiment of the present invention, the diamond-tumor tissue slice-cover glass structure is formed by the following steps:
(1) rapidly cleaning the tissue sample marked by the superparamagnetic nano particles by using deionized water, and drying the tissue sample by blowing;
(2) dropping ultraviolet glue on the region to be detected of the tissue sample, buckling the first surface of the diamond on the region to be detected of the tissue sample, uniformly applying force to the diamond-tumor tissue slice-cover glass structure by using a metal clamp, wherein the force application direction is vertical to the second surface of the diamond and the cover glass surface which are contacted with the metal clamp, so that the pretreated tissue slice is tightly attached to the diamond, and the second surface of the diamond is a surface opposite to the first surface of the diamond;
(3) placing the bonded diamond and the tissue piece under an ultraviolet lamp, and irradiating for 5 minutes by ultraviolet light to cure the ultraviolet glue;
(4) coating ultraviolet glue at the center of the upper surface of the dove prism, covering the second surface of the diamond at the ultraviolet glue, adjusting the position of the diamond and properly pressing the diamond-tumor tissue slice-cover glass structure to ensure that the diamond-tumor tissue slice-cover glass structure is attached to the dove prism in parallel and is positioned at the center of the dove prism.
(5) And (3) placing the dove prism adhered with the diamond-tumor tissue slice-cover glass structure under an ultraviolet lamp, and irradiating for 5 minutes by using ultraviolet light to solidify the ultraviolet glue.
(6) And (4) cutting off the cover glass on the periphery of the diamond by using a glass cutter, and cleaning broken glass by using a brush and a suction ball.
Fig. 4 schematically shows a structural diagram of the diamond-tumor tissue slice-cover glass structure of the present invention attached to a dove prism. The superiority of the two-dimensional magnetic sensor based on the diamond NV color center is mainly derived from the sub-nanometer size, high sensitivity, high resolution, high stability and good biocompatibility of the NV color center quantum magnetic sensor, and in order to exert the superiority, a tissue sample needs to be close to the magnetic sensor. After the diamond-tumor tissue slice-cover glass structure is formed, the tissue slice is tightly attached to the first surface of the diamond, and the attaching state is not changed at least in the whole measuring process, so that superparamagnetic nano-particles on tumor marker proteins are easily detected by an NV color center two-dimensional magnetic sensor in the diamond.
According to an embodiment of the present invention, in operation S103, the method further includes:
laser light is refracted through the dove prism onto the diamond, exciting the NV colour centre in the diamond.
More specifically, the laser with the wavelength of 532nm is used for exciting the NV color center in the diamond, and the larger imaging area can provide richer and global tumor tissue information, so that the larger area of the region to be detected of the tissue sample requires larger power of the laser for exciting the NV color center, and therefore the laser cannot be transmitted through the objective lens, and needs to be refracted onto the diamond through the dove prism to excite the NV color center in the diamond for magnetism measurement.
According to an embodiment of the present invention, in operation S103, the method further includes:
the superparamagnetic nanoparticles are magnetized by an external magnetic field.
More specifically, an external magnetic field is applied for magnetizing superparamagnetic nanoparticles, which are marked on the tissue sample, which magnetic field may be generated by a permanent magnet. The external magnetic field of the magnet and the magnetic field generated by the magnetized superparamagnetic nano-particles can increase the Zeeman effect of NV color centers in the diamond, and the external magnetic field is subtracted from the total static magnetic field obtained by measurement, so that the weak magnetic field of the tissue sample can be obtained.
Fig. 5 schematically shows a schematic diagram of a weak magnetic field generated after the superparamagnetic nanoparticle used in the present invention is magnetized, and a NV color center two-dimensional magnetic sensor in a diamond can perform magnetic field measurement on tumor tissue attached to the surface of the diamond, and the magnetic field causes zeeman splitting of NV color centers in the diamond and fluorescence count reduction at a resonance frequency of a continuous spectrum (CW spectrum), so that total static magnetic field information can be obtained by measuring the zeeman splitting resonance frequency. And respectively fitting and reconstructing the fluorescence signals of all pixels in the imaging area to obtain a magnetic field image of the tissue sample.
According to an embodiment of the present invention, in operation S103, the method further includes:
and regulating the spin state of the NV color center in the diamond by using microwaves.
More specifically, the microwave which can be regulated and controlled can be generated by placing microwave lines on the surface of a cover glass sheet above the diamond-tumor tissue slice-cover glass structure; the spin state of the NV color center is regulated and controlled by changing the microwave frequency, the fluorescence of the NV color center in different spin states is read, and a continuous wave spectrum (CW spectrum) is obtained, so that the measurement of the magnetic field sensed by the NV color center is realized.
According to the embodiment of the invention, a CMOS camera can be used for collecting data, so that higher sampling efficiency is ensured, and the realization of micron-scale imaging resolution is ensured by using the pixel scale of 100-300 nm. The core optical path of the magnetic imaging system uses the optical path of an optical microscope, wherein a multi-channel fluorescence imaging and bright field imaging system is configured, magnetic and optical multi-mode correlated imaging can be satisfied, the system can be used in combination with most conventional optical imaging methods, the imaging area can be in a submillimeter level, and the imaging resolution can be in a micrometer or submicrometer level.
According to the embodiment of the invention, single cell segmentation and statistical software in optical imaging can be used for reference to perform inverse solution reconstruction on a magnetic image, magnetic quantification of the tumor marker single cell level is carried out, and a 4', 6-diamidino-2-phenylindole (DAPI) marked cell nucleus is used as important auxiliary information for single cell analysis.
Fig. 6 schematically shows a schematic view of an apparatus for imaging using a magnetic imaging method of tumor tissue according to the present invention.
As shown in fig. 6, in operation S103, the method further includes:
(1) finding a tissue sample to-be-detected area in a diamond-tumor tissue section-cover glass structure through an optical microscope, focusing, and acquiring a fluorescence image of 4', 6-diamidino-2-phenylindole (DAPI), wherein the acquired fluorescence image of 4', 6-diamidino-2-phenylindole (DAPI) represents an acquired fluorescence image of a cell nucleus;
(2) switching the detection channel of the optical microscope to an NV fluorescence channel, and adjusting the size of laser and the angle of incidence on a dove prism to ensure that the fluorescence intensity in a visual field is larger and uniformly distributed;
(3) setting parameters such as a microwave frequency range, camera exposure time, data accumulation times and the like, and controlling the synchronization of the CMOS camera and a microwave source by using a LabVIEW program to finish the acquisition of NV color center fluorescence data;
(4) acquired NV color center fluorescence data are processed through an MATLAB program, data at each pixel point are accumulated, and a continuous wave spectrum (CW spectrum) of each pixel is obtained after accumulation;
(5) obtaining a corresponding NV color center frequency diagram by Lorentzian fitting continuous wave spectrum (CW spectrum);
(6) obtaining a fitting result of the total static magnetic field by fitting a frequency map through a polynomial, and deducting the frequency map of the external magnetic field to obtain a frequency map of the tissue sample;
(7) converting the frequency map of the tissue sample into a magnetic field image to obtain the magnetic field image of the tissue sample;
(8) the method comprises the steps of generating a deep learning model of an antagonistic network by using a condition, training the model, establishing a mapping relation between a magnetic field image and corresponding magnetic particle marker proteins, realizing identification and inverse solution of magnetic field signals, and obtaining a magnetic imaging graph of a tissue sample, wherein the magnetic imaging graph can obtain intuitive distribution and expression quantity information of tumor marker proteins of the tissue sample.
According to the tumor tissue magnetic imaging method disclosed by the embodiment of the invention, superparamagnetic nanoparticles are used as magnetic signal labels to replace fluorescent labels and enzyme labels in the traditional optical method, and are indirectly marked on tumor marker proteins of a tissue sample through antibody-antigen reaction; superparamagnetic nano-particles are detected by an NV color center two-dimensional magnetic sensor in the diamond, so that the magnetic imaging of tumor tissue with micron-scale resolution can be realized. The tumor tissue magnetic imaging method of the embodiment of the invention has high signal stability and low background, can realize absolute quantification, and can realize magnetic and optical multi-modal associated imaging of biological tissues because the magnetism is another physical quantity different from light, electricity and the like.
The flowchart and block diagrams in the figures illustrate the architecture, functionality, and operation of possible implementations of systems, methods and computer program products according to various embodiments of the present invention. In this regard, each block in the flowchart or block diagrams may represent a module, segment, or portion of code, which comprises one or more executable instructions for implementing the specified logical function(s). It should also be noted that, in some alternative implementations, the functions noted in the block may occur out of the order noted in the figures. For example, two blocks shown in succession may, in fact, be executed substantially concurrently, or the blocks may sometimes be executed in the reverse order, depending upon the functionality involved. It will also be noted that each block of the block diagrams or flowchart illustration, and combinations of blocks in the block diagrams or flowchart illustration, can be implemented by special purpose hardware-based systems which perform the specified functions or acts, or combinations of special purpose hardware and computer instructions.
It will be appreciated by a person skilled in the art that various combinations and/or combinations of features described in the various embodiments and/or in the claims of the invention are possible, even if such combinations or combinations are not explicitly described in the invention. In particular, various combinations and/or combinations of the features recited in the various embodiments and/or claims of the present invention may be made without departing from the spirit or teaching of the invention. All such combinations and/or associations fall within the scope of the present invention.
The embodiments of the present invention have been described above. However, these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Although the embodiments are described separately above, this does not mean that the measures in the embodiments cannot be used in advantageous combination. The scope of the invention is defined by the appended claims and equivalents thereof. Various alternatives and modifications can be devised by those skilled in the art without departing from the scope of the invention, and these alternatives and modifications are intended to fall within the scope of the invention.

Claims (10)

1. A method of magnetic imaging of tumor tissue with micron resolution, comprising:
tagging a tissue sample with superparamagnetic nanoparticles, wherein the superparamagnetic nanoparticles are tagged on a tumor marker protein of the tissue sample;
attaching diamond to the tissue sample to form a diamond-tumor tissue section-cover glass structure, wherein the tumor tissue section belongs to the tissue sample;
measuring a magnetic field of the tissue sample by using a nitrogen vacancy color center (NV color center) two-dimensional magnetic sensor in the diamond to obtain a magnetic field image of the tissue sample; and
and processing the magnetic field image through a deep learning model to obtain a micron resolution magnetic imaging image of the tissue sample.
2. The method of claim 1, wherein the labeling of the tissue sample with superparamagnetic nanoparticles further comprises:
and carrying out immunomagnetic marking treatment on the tissue sample, wherein the immunomagnetic marking comprises a superparamagnetic nanoparticle incubation process so as to enable tumor marker proteins in the tissue sample to be combined with the superparamagnetic nanoparticle specific marks.
3. The method of claim 1, further comprising:
treating the diamond to form a high density NV color center having a depth of 10nm to 110nm in a first surface of the diamond, the first surface of the diamond in contact with the tissue slice.
4. The method of claim 1, further comprising:
dripping ultraviolet glue into the region to be detected of the tumor tissue slice in the diamond-tumor tissue slice-cover glass structure so as to enable the first surface of the diamond to contact the region to be detected of the tumor tissue slice; and
and uniformly applying force to the diamond-tumor tissue slice-cover glass structure by using a metal clamp, wherein the force application direction is vertical to the second surface of the diamond and the cover glass surface which are contacted with the metal clamp, so that the pretreated tissue slice and the diamond are tightly attached, and the second surface of the diamond is a surface opposite to the first surface of the diamond.
5. The method of claim 1, further comprising:
integrally attaching the diamond-tumor tissue slice-cover glass structure to a dove prism, wherein the lower surface of the diamond is placed on the dove prism;
ultraviolet glue is used between the diamond-tumor tissue slice-cover glass structure and the dove prism for pasting; wherein the diamond-tumor tissue slice-cover glass structure is placed in the center of the dove prism; and
and (3) properly pressing the diamond-tumor tissue slice-cover glass structure to ensure that the diamond-tumor tissue slice-cover glass structure is attached to the dove prism in parallel.
6. The method of claim 1, further comprising:
and (3) preprocessing the cover glass by polylysine before the operation of marking the tissue sample by the superparamagnetic nanoparticles, and placing the tumor tissue section on the cover glass so as to fix the tumor tissue section on the cover glass.
7. The method of claim 5, further comprising:
refracting laser light through the dove prism onto the diamond to excite an NV colour centre in the diamond.
8. The method of claim 1, further comprising:
magnetizing the superparamagnetic nanoparticles by an external magnetic field using the external magnetic field;
and regulating the spin state of the NV color center in the diamond by using microwaves.
9. The method of claim 1, further comprising:
fluorescence data of NV colour centers in the diamond is collected using a Complementary Metal Oxide Semiconductor (CMOS) camera to obtain a magnetic field image.
10. The method of claims 1-9, further comprising:
the tissue sample to be tested may be a biological tissue sample of another type than a tumor tissue sample.
CN202111515040.6A 2021-12-13 2021-12-13 Tumor tissue magnetic imaging method with micron resolution Pending CN114113191A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111515040.6A CN114113191A (en) 2021-12-13 2021-12-13 Tumor tissue magnetic imaging method with micron resolution

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111515040.6A CN114113191A (en) 2021-12-13 2021-12-13 Tumor tissue magnetic imaging method with micron resolution

Publications (1)

Publication Number Publication Date
CN114113191A true CN114113191A (en) 2022-03-01

Family

ID=80365075

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111515040.6A Pending CN114113191A (en) 2021-12-13 2021-12-13 Tumor tissue magnetic imaging method with micron resolution

Country Status (1)

Country Link
CN (1) CN114113191A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114706023A (en) * 2022-06-06 2022-07-05 中国科学技术大学 Magnetic detection method for biomolecular interaction

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114706023A (en) * 2022-06-06 2022-07-05 中国科学技术大学 Magnetic detection method for biomolecular interaction

Similar Documents

Publication Publication Date Title
US7282180B2 (en) Devices and methods to image objects
AU2008249152B2 (en) Devices and methods to image objects
CN106994370B (en) Micro-fluidic chip based on magnetic cup compound particle
Smith Two microscopes are better than one
CN110337588B (en) Method and system for quantitative immunohistochemistry
US20100196914A1 (en) Rare cell detection using flat-panel imager and chemiluminescent or radioisotopic tags
EP3124969A1 (en) Biological-material quantitation method based on multiple-antigen immunostaining
JP6911855B2 (en) Biomaterial quantification method, image processing device, pathological diagnosis support system and program
CN105866410A (en) Immunochromatographic detection test strip for joint detection of NSE and CEA, and preparation method and application method thereof
CN111366563B (en) Digital plasma immunoadsorption kit and manufacturing and testing method thereof
Viter et al. Bioanalytical system for detection of cancer cells with photoluminescent ZnO nanorods
US20160216209A1 (en) Method for determining quantity of biological material in tissue section
CN114113191A (en) Tumor tissue magnetic imaging method with micron resolution
CN107656048A (en) A kind of immuno-chromatographic test paper strip and its application of half-quantitative detection antigen or antibody
CN101294903A (en) Method for detecting identical antigen expression on identical sample
CN103063849A (en) A method for simultaneous detection of cancer-associated fibroblasts and expressed proteins thereof
EP0300037B1 (en) Cell screening methods
US20180080936A1 (en) Method for detection of l523s expression in biological samples
CN111363789B (en) Kit and method for simultaneously detecting protein and RNA
Nethercott et al. Immunocytochemical analysis of human pluripotent stem cells
CN207396501U (en) A kind of immuno-chromatographic test paper strip of half-quantitative detection antigen or antibody
CN103353531B (en) Highly Sensitive System and method for analysis of troponin
US11885800B2 (en) Method and system for detecting analyte of interest using magnetic field sensor and magnetic particles
JP7160047B2 (en) Biological substance quantification method, image processing device and program
WO2021192910A1 (en) Image generation method, image generation device, and program

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination