JP7443762B2 - How to retain cells contained in the sample - Google Patents
How to retain cells contained in the sample Download PDFInfo
- Publication number
- JP7443762B2 JP7443762B2 JP2019234568A JP2019234568A JP7443762B2 JP 7443762 B2 JP7443762 B2 JP 7443762B2 JP 2019234568 A JP2019234568 A JP 2019234568A JP 2019234568 A JP2019234568 A JP 2019234568A JP 7443762 B2 JP7443762 B2 JP 7443762B2
- Authority
- JP
- Japan
- Prior art keywords
- cells
- holding
- cell
- holding part
- sample
- 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.)
- Active
Links
- 239000002904 solvent Substances 0.000 claims description 35
- 239000000758 substrate Substances 0.000 claims description 34
- 238000000034 method Methods 0.000 claims description 22
- 239000002480 mineral oil Substances 0.000 claims description 6
- 235000010446 mineral oil Nutrition 0.000 claims description 6
- 238000007599 discharging Methods 0.000 claims description 5
- 229940057995 liquid paraffin Drugs 0.000 claims description 5
- 238000006073 displacement reaction Methods 0.000 claims 1
- 210000004027 cell Anatomy 0.000 description 157
- 238000001514 detection method Methods 0.000 description 26
- 108090000623 proteins and genes Proteins 0.000 description 7
- 102000004169 proteins and genes Human genes 0.000 description 7
- 239000000243 solution Substances 0.000 description 7
- 125000006850 spacer group Chemical group 0.000 description 7
- PRDFBSVERLRRMY-UHFFFAOYSA-N 2'-(4-ethoxyphenyl)-5-(4-methylpiperazin-1-yl)-2,5'-bibenzimidazole Chemical group C1=CC(OCC)=CC=C1C1=NC2=CC=C(C=3NC4=CC(=CC=C4N=3)N3CCN(C)CC3)C=C2N1 PRDFBSVERLRRMY-UHFFFAOYSA-N 0.000 description 6
- 210000003855 cell nucleus Anatomy 0.000 description 6
- 239000012128 staining reagent Substances 0.000 description 6
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 5
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 5
- 229930006000 Sucrose Natural products 0.000 description 5
- 210000004369 blood Anatomy 0.000 description 5
- 239000008280 blood Substances 0.000 description 5
- 238000010586 diagram Methods 0.000 description 5
- 239000012212 insulator Substances 0.000 description 5
- 239000007788 liquid Substances 0.000 description 5
- 239000002609 medium Substances 0.000 description 5
- 230000005012 migration Effects 0.000 description 5
- 238000013508 migration Methods 0.000 description 5
- 238000010186 staining Methods 0.000 description 5
- 239000005720 sucrose Substances 0.000 description 5
- 230000000717 retained effect Effects 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 3
- 239000007864 aqueous solution Substances 0.000 description 3
- 229940098773 bovine serum albumin Drugs 0.000 description 3
- 239000000356 contaminant Substances 0.000 description 3
- 230000000694 effects Effects 0.000 description 3
- 239000000644 isotonic solution Substances 0.000 description 3
- 230000014759 maintenance of location Effects 0.000 description 3
- 238000011084 recovery Methods 0.000 description 3
- 238000005406 washing Methods 0.000 description 3
- 208000005443 Circulating Neoplastic Cells Diseases 0.000 description 2
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 2
- 239000007995 HEPES buffer Substances 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 239000012503 blood component Substances 0.000 description 2
- 230000012292 cell migration Effects 0.000 description 2
- 238000012258 culturing Methods 0.000 description 2
- 238000004720 dielectrophoresis Methods 0.000 description 2
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 2
- 230000005684 electric field Effects 0.000 description 2
- 239000012091 fetal bovine serum Substances 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 239000011859 microparticle Substances 0.000 description 2
- 239000008188 pellet Substances 0.000 description 2
- 239000002953 phosphate buffered saline Substances 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 239000006228 supernatant Substances 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 206010003445 Ascites Diseases 0.000 description 1
- UXVMQQNJUSDDNG-UHFFFAOYSA-L Calcium chloride Chemical compound [Cl-].[Cl-].[Ca+2] UXVMQQNJUSDDNG-UHFFFAOYSA-L 0.000 description 1
- 108091007741 Chimeric antigen receptor T cells Proteins 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 101000958041 Homo sapiens Musculin Proteins 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- RVGRUAULSDPKGF-UHFFFAOYSA-N Poloxamer Chemical compound C1CO1.CC1CO1 RVGRUAULSDPKGF-UHFFFAOYSA-N 0.000 description 1
- 239000004793 Polystyrene Substances 0.000 description 1
- 206010036790 Productive cough Diseases 0.000 description 1
- 210000004381 amniotic fluid Anatomy 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 239000003792 electrolyte Substances 0.000 description 1
- 210000001671 embryonic stem cell Anatomy 0.000 description 1
- 238000001704 evaporation Methods 0.000 description 1
- 210000003608 fece Anatomy 0.000 description 1
- 210000004700 fetal blood Anatomy 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 238000002073 fluorescence micrograph Methods 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- 238000001215 fluorescent labelling Methods 0.000 description 1
- 238000012252 genetic analysis Methods 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 102000046949 human MSC Human genes 0.000 description 1
- 238000007654 immersion Methods 0.000 description 1
- 210000004263 induced pluripotent stem cell Anatomy 0.000 description 1
- XUGNVMKQXJXZCD-UHFFFAOYSA-N isopropyl palmitate Chemical compound CCCCCCCCCCCCCCCC(=O)OC(C)C XUGNVMKQXJXZCD-UHFFFAOYSA-N 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 238000004020 luminiscence type Methods 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 210000002901 mesenchymal stem cell Anatomy 0.000 description 1
- 239000011259 mixed solution Substances 0.000 description 1
- 230000000877 morphologic effect Effects 0.000 description 1
- 210000003463 organelle Anatomy 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- 239000012466 permeate Substances 0.000 description 1
- 239000002504 physiological saline solution Substances 0.000 description 1
- 210000002381 plasma Anatomy 0.000 description 1
- 229920001993 poloxamer 188 Polymers 0.000 description 1
- 229920002223 polystyrene Polymers 0.000 description 1
- 210000003296 saliva Anatomy 0.000 description 1
- 238000004062 sedimentation Methods 0.000 description 1
- 210000000582 semen Anatomy 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 238000002791 soaking Methods 0.000 description 1
- 210000003802 sputum Anatomy 0.000 description 1
- 208000024794 sputum Diseases 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 210000001519 tissue Anatomy 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
Landscapes
- Sampling And Sample Adjustment (AREA)
- Apparatus Associated With Microorganisms And Enzymes (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Description
本発明は、試料中に含まれる細胞の保持方法に関する。 The present invention relates to a method for retaining cells contained in a sample.
ライフサイエンス研究分野では、複数の細胞種を含む試料中より、標的の細胞を、単一細胞かつ生きた状態で検出および回収することが求められる。方法の一例として、微小粒子を保持可能な保持部を設けた基板を用いる方法が挙げられる(例えば、特許文献1)。 In the field of life science research, it is required to detect and recover target cells in a single living state from samples containing multiple cell types. An example of the method is a method using a substrate provided with a holding portion capable of holding microparticles (for example, Patent Document 1).
特許文献1に記載の基板では、細胞保持空間に誘電泳動力を発生させる目的で、保持部と並行に配置した天板面に電極を設けている。そのため、保持部に保持された標的細胞をキャピラリ等による吸引で回収しようとする際、電極(天板)を外す工程が必要であり、当該工程で発生する振動等により保持部に保持された細胞が当該保持部外または別の保持部へ移動してしまうという課題があった。
さらに前述した方法では、細胞の生死にかかわらず保持部に保持されるため、生細胞を検出および回収しようする際、死細胞が混入する課題もあった。
本発明の目的は、死細胞の混入を抑え、かつ細胞の保持部への保持状態を安定的に維持することにある。
In the substrate described in Patent Document 1, electrodes are provided on the top plate surface arranged in parallel with the holding part for the purpose of generating dielectrophoretic force in the cell holding space. Therefore, when attempting to collect target cells held in the holding unit by suction using a capillary, etc., a step is required to remove the electrode (top plate), and the cells held in the holding unit are There is a problem in that the holder moves outside the holding section or to another holding section.
Furthermore, in the above-described method, since cells are retained in the holding portion regardless of whether they are alive or dead, there is also the problem that dead cells may be mixed in when trying to detect and collect living cells.
An object of the present invention is to suppress the contamination of dead cells and to stably maintain the state in which the cells are held in the holding portion.
上記課題を解決するために、本発明者らは鋭意検討を重ねた結果、本発明に到達した。 In order to solve the above problems, the present inventors have made extensive studies and have arrived at the present invention.
すなわち本発明の一態様は、
細胞を保持可能な保持部と、導入口と、排出口と、を設けた基板を備えた細胞保持装置を用いた、細胞の保持方法であって、前記細胞を含む試料を、前記導入口より前記保持部に導入する工程と、前記細胞を前記保持部に保持させる工程と、前記導入口から置換溶媒を導入し、前記試料の溶媒を排出口から排出させる工程と、を含むことを特徴とする。
That is, one aspect of the present invention is
A method for holding cells using a cell holding device equipped with a substrate having a holding part capable of holding cells, an inlet, and an outlet, the sample containing the cells being passed through the inlet. The method includes the steps of introducing the sample into the holding part, holding the cells in the holding part, and introducing a replacement solvent from the inlet and discharging the sample solvent from the outlet. do.
以下、本発明を詳細に説明する。 The present invention will be explained in detail below.
保持部に保持する細胞の一例として、ヒト由来の人工多能性幹細胞(ヒトiPS細胞)もしくは胚性幹細胞(ヒトES細胞)またはこれら細胞由来の分化細胞、ヒト由来の間葉系幹細胞(ヒトMSC細胞)、キメラ抗原受容体T細胞(CAR-T細胞)が挙げられる。 Examples of cells retained in the holding unit include human-derived induced pluripotent stem cells (human iPS cells) or embryonic stem cells (human ES cells), differentiated cells derived from these cells, and human-derived mesenchymal stem cells (human MSCs). CAR-T cells) and chimeric antigen receptor T cells (CAR-T cells).
細胞保持装置は、細胞を保持可能な保持部と、導入口と、排出口と、を設けた基板を備える。保持部は、凹部または貫通孔を有した形状であり、凹部を保持部とした場合、一端が保持させる細胞の全て、またはその一部を保持可能な大きさの開口部を有しており他端が底部を形成している態様とし、貫通孔を保持部とした場合、一端が保持させる細胞の全て、またはその一部を保持可能な大きさの開口部を有し他端が当該開口部よりも狭い開口部とするか、両端が保持させる細胞の一部を保持可能な大きさの開口部である態様とする。なお保持部の大きさ(径)を保持させる細胞を一つだけ保持可能な大きさとすると、検出した細胞の一細胞単位での回収および解析(形態学的分析、組織型分析、遺伝子分析など)が容易に行なえる点で好ましい。
導入口と排出口は、その形状や大きさに特に制限はないが、基板上で対極に位置するように配置されていることが好ましい。
The cell holding device includes a substrate provided with a holding part capable of holding cells, an inlet, and an outlet. The holding part has a shape with a recess or a through hole, and when the recess is used as the holding part, one end has an opening large enough to hold all or a part of the cells to be held, and the other end has an opening large enough to hold all or a part of the cells to be held. When the end forms the bottom and the through hole is used as the holding part, one end has an opening large enough to hold all or a part of the cells to be held, and the other end has the opening. The opening may be narrower than the opening, or the opening may be large enough to hold a portion of the cells to be held at both ends. If the size (diameter) of the holding part is set to a size that can hold only one cell, the detected cells can be collected and analyzed in single cell units (morphological analysis, tissue type analysis, genetic analysis, etc.) This is preferable because it can be easily performed.
Although there is no particular restriction on the shape or size of the inlet and the outlet, it is preferable that the inlet and the outlet are arranged so as to be opposite to each other on the substrate.
細胞を含む試料に用いられる溶媒は、細胞に対して等張かつ当該細胞の生存能力(viability)を維持可能な溶媒であれば特に限定はなく、一例として生理食塩水や、マンニトール、グルコース、スクロース等の糖類を含んだ水溶液や、当該水溶液に塩化カルシウム、塩化マグネシウムなどの電解質、および/またはBSA(ウシ血清アルブミン)等のタンパク質をさらに含んだ水溶液や、前記細胞の培養に用いた培養液が挙げられる。特に細胞を含む液体として、スクロースを含む水溶液に細胞を含んだ試料を懸濁させた液体を用いると、細胞へのダメージが少なくなる点で好ましい。添加するスクロースの濃度は等張液となる濃度とすればよく、具体的には200mM以上350mM以下とするとよい。さらに前記細胞がCTC(血中循環腫瘍細胞)といった血液中に含まれ得る細胞である場合、血液(全血)、希釈血液、血清、血漿、髄液、臍帯血、成分採血液などの血液試料や、尿、唾液、精液、糞便、痰、羊水、腹水などの血液成分を含み得る試料も、本発明における試料溶媒に含まれる。 The solvent used for samples containing cells is not particularly limited as long as it is isotonic to the cells and can maintain the viability of the cells; examples include physiological saline, mannitol, glucose, and sucrose. an aqueous solution containing sugars such as, an aqueous solution further containing an electrolyte such as calcium chloride or magnesium chloride, and/or a protein such as BSA (bovine serum albumin), or a culture medium used for culturing the cells. Can be mentioned. In particular, it is preferable to use a liquid in which a sample containing cells is suspended in an aqueous solution containing sucrose as the liquid containing cells, since damage to the cells is reduced. The concentration of sucrose to be added may be a concentration that provides an isotonic solution, and specifically, it is preferably 200 mM or more and 350 mM or less. Furthermore, if the cells are cells that can be contained in blood, such as CTCs (circulating tumor cells), blood samples such as blood (whole blood), diluted blood, serum, plasma, cerebrospinal fluid, umbilical cord blood, blood component collection, etc. In addition, samples that may contain blood components such as urine, saliva, semen, feces, sputum, amniotic fluid, and ascites are also included in the sample solvent in the present invention.
導入した試料中に含まれる細胞を保持部に保持させる方法は特に限定がなく、単に導入口から細胞を含む試料を導入するだけでもよいし、細胞を含む試料を導入した後、遠心力を利用して保持部へ強制的に当該細胞を保持させてもよい。中でも細胞を含む試料を導入した後、誘電泳動力を利用して保持部へ当該細胞を保持させると、細胞を保持部へ効率的に保持できる点で好ましい。いずれの方法であっても、細胞は保持部に固定されているわけではなく、保持部内で沈降している状態である。誘電泳動力を用いる場合、具体的には、交流電圧を印加することで誘電泳動力を発生させ、保持部に細胞を保持さればよい。印加する交流電圧は、保持部内の細胞の充放電が周期的に繰り返される波形を有した交流電圧であると好ましく、周波数を100kHz以上3MHz以下の間とし、電界強度を1×105から5×105V/mの間とすると特に好ましい(WO2011/149032号および特開2012-013549号公報参照)。好ましい基板の一態様として、後述する図1および図2に示す基板100や、特開2019-100940号公報に開示の基板が挙げられる。 There are no particular limitations on the method for holding cells contained in the introduced sample in the holding unit; it is possible to simply introduce the sample containing cells through the introduction port, or by using centrifugal force after introducing the sample containing cells. The cells may be forcefully held in the holding section. Among these, it is preferable to introduce a sample containing cells and then use dielectrophoretic force to hold the cells in the holding part, since the cells can be efficiently held in the holding part. In either method, the cells are not fixed to the holding part, but are in a state of sedimentation within the holding part. When using dielectrophoretic force, specifically, dielectrophoretic force may be generated by applying an alternating current voltage, and cells may be held in the holding portion. The applied AC voltage is preferably an AC voltage having a waveform in which charging and discharging of the cells in the holding part are periodically repeated, the frequency is between 100 kHz and 3 MHz, and the electric field strength is between 1 x 10 and 5 x. It is particularly preferable to set it between 10 5 V/m (see WO2011/149032 and JP2012-013549). Examples of preferable substrates include the substrate 100 shown in FIGS. 1 and 2, which will be described later, and the substrate disclosed in Japanese Patent Application Publication No. 2019-100940.
置換溶媒は、前記試料溶媒とは異なり、試料中の細胞よりも密度が小さく、かつ動粘度が高い、具体的には3mm2/s(3cSt)以上の溶媒である。好ましい例としてミネラルオイル、流動パラフィン、パルミチン酸イソプロピルが挙げられる。 The replacement solvent, unlike the sample solvent, is a solvent that has a lower density than the cells in the sample and a higher kinematic viscosity, specifically, 3 mm 2 /s (3 cSt) or more. Preferred examples include mineral oil, liquid paraffin, and isopropyl palmitate.
置換溶媒は、基板の導入口から導入すれば、保持部内にある試料溶媒は押し出され、排出口から排出されるため、保持部内の溶媒が試料溶媒から置換溶媒へと置き換わる。導入する置換溶媒の量は、保持部の容積を考慮して十分な量を導入することが好ましい。 When the replacement solvent is introduced from the inlet of the substrate, the sample solvent in the holding part is pushed out and discharged from the outlet, so that the sample solvent in the holding part is replaced by the replacement solvent. It is preferable to introduce a sufficient amount of replacement solvent in consideration of the volume of the holding section.
保持部に保持された細胞は、当該細胞が有する特性に基づき検出を行うことが可能である。例えば、細胞の大きさや、細胞内に存在する顆粒もしくは小器官の存否などに基づき検出しても良く、抗体、アプタマー、タンパクリガンドなどによる標識や細胞核染色試薬などによる染色後、当該標識や染色像に基づき検出しても良い。細胞核染色試薬の好ましい例として、細胞膜を透過可能なHoechst 33342(商品名)が挙げられる。検出対象細胞か否かの指標は、明確な数値で決定する必要はなく、例えば、検出対象細胞の画像データ(明視野像、標識像、染色試薬像)を教師データとした、人工知能(AI)による判定を行なってもよい。 The cells held in the holding part can be detected based on the characteristics of the cells. For example, detection may be based on the size of the cell or the presence or absence of granules or organelles present within the cell. It may be detected based on. A preferred example of the cell nucleus staining reagent is Hoechst 33342 (trade name), which can permeate cell membranes. The index of whether or not a cell is a detection target does not need to be determined by a clear numerical value. ) may be used.
前述した方法で試料中に含まれる細胞を検出した後、検出した前記細胞をキャピラリによる吸引吐出で回収することで、試料中に含まれる特定細胞を一細胞単位で回収できる。前記キャピラリは、特定細胞の一細胞単位での回収が可能な態様であれば特に限定はなく、一例としてナノピペット、ピコピペット、ガラスキャピラリや特開2016-142616号公報に開示の微小粒子回収装置が挙げられる。 After detecting the cells contained in the sample using the method described above, the detected cells are collected by suction and discharge using a capillary, thereby making it possible to collect specific cells contained in the sample in units of cells. The capillary is not particularly limited as long as it is capable of collecting specific cells in single cell units, and examples thereof include a nanopipette, a picopipette, a glass capillary, and a microparticle collection device disclosed in JP-A No. 2016-142616. Can be mentioned.
本発明は、試料中に含まれる死細胞を選択的に除去でき、かつ保持部への細胞保持状態を維持できるため、保持部に保持された細胞の検出および回収が容易となる。 According to the present invention, dead cells contained in a sample can be selectively removed and the state of cell retention in the holding part can be maintained, so that detection and collection of cells held in the holding part can be facilitated.
以下、図面を用いて本発明をさらに詳細に説明する。
本発明の方法を実施可能な装置を構成する基板の一例を図1、その正面図を図2に示す。
Hereinafter, the present invention will be explained in more detail using the drawings.
An example of a substrate constituting an apparatus capable of carrying out the method of the present invention is shown in FIG. 1, and a front view thereof is shown in FIG.
図1に示す基板100は、
貫通孔11aを有した平板状の遮光部材11と、貫通孔12aを有した平板状の絶縁体12と、導入口13a、排出口13bおよび貫通部13cを有した平板状のスペーサ13とからなる細胞導入保持手段10と、
細胞導入保持手段10を上下方向に密着して挟むよう設けた電極21・22と、
電極21・22同士を接続する導線30と、
電極21・22に信号を印加する信号発生器40と、
を備えている。遮光部材11が有する貫通孔11aと絶縁体12が有する貫通孔12aとは互いに同一の寸法および形状であり、かつそれぞれの貫通孔の位置が一致するよう遮光部材11および絶縁体12を設けている。貫通孔11a、貫通孔12aおよび遮光部材11の下部に密着して設けた電極基板21により保持部50が構成され、導入口13aから細胞を含む液体を導入すると、貫通部13cを通じて保持部50へ細胞200が導入される。電極22はスペーサ13上部に密着して設けており、導入口13aから導入した、細胞200を含む液体の飛散や蒸発を防止している。なお、保持部50に保持した細胞200の回収を容易にするため、電極22はスペーサ13から取り外し可能な構造となっている。
The substrate 100 shown in FIG.
Consists of a flat light shielding member 11 having a through hole 11a, a flat insulator 12 having a through hole 12a, and a flat spacer 13 having an inlet 13a, an outlet 13b, and a through part 13c. Cell introduction and holding means 10;
electrodes 21 and 22 provided to closely sandwich the cell introduction and holding means 10 in the vertical direction;
A conductive wire 30 connecting the electrodes 21 and 22,
a signal generator 40 that applies signals to the electrodes 21 and 22;
It is equipped with The through-hole 11a of the light-shielding member 11 and the through-hole 12a of the insulator 12 have the same size and shape, and the light-shielding member 11 and the insulator 12 are provided so that the positions of the respective through-holes match. . A holding part 50 is constituted by the through hole 11a, the through hole 12a, and the electrode substrate 21 provided in close contact with the lower part of the light shielding member 11. When a liquid containing cells is introduced from the introduction port 13a, it enters the holding part 50 through the through hole 13c. Cells 200 are introduced. The electrode 22 is provided in close contact with the upper part of the spacer 13 to prevent the liquid containing the cells 200 introduced from the introduction port 13a from scattering or evaporating. Note that, in order to facilitate recovery of the cells 200 held in the holding section 50, the electrode 22 has a structure that is removable from the spacer 13.
次に、図1および図2に示す基板を備えた装置を用いた、本発明の細胞の検出および回収方法の一例について図3を用いて説明する。 Next, an example of the cell detection and recovery method of the present invention using an apparatus equipped with the substrate shown in FIGS. 1 and 2 will be described using FIG. 3.
(1-1)細胞を標識する工程
検出対象細胞が有するタンパク質を認識する標識タンパク質を含む溶液を試料に添加することで、当該細胞と前記標識タンパク質との複合体を形成させる。
(1-1) Step of labeling cells A solution containing a labeled protein that recognizes a protein possessed by a cell to be detected is added to a sample to form a complex between the cell and the labeled protein.
(1-2)保持部へ細胞を導入する工程
図1に示す基板100に設けた導入口13aから、標識タンパク質300と結合した検出対象細胞210および夾雑細胞220を含む試料を導入し、誘電泳動力60を利用してこれら細胞を保持部50へ導入させる。具体的には、信号発生器40から電極21・22へ交流電圧を印加することで誘電泳動力60を発生させ、保持部50へ検出対象細胞210および夾雑細胞220を導入する。信号発生器40から電極21・22へ印加する交流電圧は、保持部50に保持された細胞の充放電が周期的に繰り返される波形を有した交流電圧とすると好ましく、周波数を100kHz以上3MHz以下とし、電界強度を1×105以上5×105V/m以下と特に好ましい。
(1-2) Step of introducing cells into the holding part A sample containing detection target cells 210 bound to labeled protein 300 and contaminant cells 220 is introduced from the introduction port 13a provided in the substrate 100 shown in FIG. 1, and subjected to dielectrophoresis. These cells are introduced into the holding part 50 using the force 60. Specifically, the dielectrophoretic force 60 is generated by applying an alternating voltage from the signal generator 40 to the electrodes 21 and 22, and the detection target cells 210 and the contaminant cells 220 are introduced into the holding part 50. The AC voltage applied from the signal generator 40 to the electrodes 21 and 22 is preferably an AC voltage having a waveform in which charging and discharging of the cells held in the holding unit 50 are periodically repeated, and the frequency is set to be 100 kHz or more and 3 MHz or less. It is particularly preferable that the electric field strength is 1×10 5 or more and 5×10 5 V/m or less.
(1-3)溶媒置換工程
導入口13aから、置換溶媒を導入することで、排出口13bより残存試料が排出され、共に保持部50内空間を置換溶媒で置換される。本工程により、検出対象細胞210のうち死細胞を選択的に除去できる。
(1-3) Solvent Replacement Step By introducing the replacement solvent from the inlet 13a, the remaining sample is discharged from the outlet 13b, and the space inside the holding part 50 is replaced with the replacement solvent. Through this step, dead cells among the detection target cells 210 can be selectively removed.
(1-4)検出対象細胞210を検出する工程
基板を移動させる手段(図示せず)で基板をXY軸方向に移動させながら、検出部400および計測部により、検出対象細胞210に結合した標識タンパク質由来の蛍光または発光および位置情報を取得し、検出対象細胞210の位置を検出する。
(1-4) Step of detecting detection target cells 210 While moving the substrate in the XY axis directions using means for moving the substrate (not shown), the label bound to the detection target cells 210 is detected by the detection unit 400 and the measurement unit. Protein-derived fluorescence or luminescence and position information are acquired, and the position of the detection target cell 210 is detected.
(1-5)検出対象細胞210を回収する工程
検出部400および計測部により検出した検出対象細胞210を回収するために、電極22をスペーサ13から取り外した後、キャピラリ500で吸引することで、基板100から検出対象細胞210を回収する。電極22を取り外す際は、スペーサ13を剥がさないよう取り外す必要がある。もしスペーサ13が絶縁体12から剥がれると、装置内に保持されている溶液が系外に流れてしまい、検出対象細胞210が破壊されるからである。
検出対象細胞210の吸引は、基板を移動させる手段およびノズルを移動させる手段(ともに図示せず)を用いて前記(1-3)の工程で検出した検出対象細胞210が保持されている保持部へキャピラリ500の中心を移動させ、キャピラリ500により液を吸引することで検出対象細胞210を回収する。なお、キャピラリ500による検出対象細胞210の吸引位置を、検出対象細胞210を保持した保持部50の中心から水平方向に一定距離ずらした位置とすると、検出対象細胞210の吸引を容易に行なえるため好ましい。具体的には検出対象細胞210の吸引位置を、保持部50の中心から水平方向に保持部50の直径の0.1倍から2倍の長さ分(ただし隣接する保持部50間の距離の2分の1以下)ずらし、かつ保持部50の高さから垂直方向に保持部50の高さの0.01倍から2倍の高さ分高い位置とすると好ましい。
(1-5) Step of collecting the detection target cells 210 In order to collect the detection target cells 210 detected by the detection unit 400 and the measurement unit, after removing the electrode 22 from the spacer 13, by suctioning with the capillary 500, Detection target cells 210 are collected from the substrate 100. When removing the electrode 22, it is necessary to remove the spacer 13 without peeling it off. This is because if the spacer 13 were to peel off from the insulator 12, the solution held within the device would flow out of the system and the detection target cells 210 would be destroyed.
The detection target cells 210 are sucked into the holding part in which the detection target cells 210 detected in the step (1-3) are held using a means for moving the substrate and a means for moving the nozzle (both not shown). The detection target cells 210 are collected by moving the center of the capillary 500 and sucking the liquid through the capillary 500. Note that if the position at which the capillary 500 aspirates the detection target cells 210 is shifted by a certain distance in the horizontal direction from the center of the holding part 50 holding the detection target cells 210, the detection target cells 210 can be easily aspirated. preferable. Specifically, the suction position of the detection target cell 210 is set horizontally from the center of the holding part 50 by a distance of 0.1 to twice the diameter of the holding part 50 (however, the distance between adjacent holding parts 50 is It is preferable that the height of the holding part 50 is shifted by 0.01 times to twice the height of the holding part 50 in the vertical direction from the height of the holding part 50.
以下、実施例を用いて本発明をさらに詳細に説明するが、本発明は当該例に限定されるものではない。 Hereinafter, the present invention will be explained in more detail using examples, but the present invention is not limited to the examples.
実施例1 溶媒置換による死細胞除去効果
(1)iPS細胞の調製
(1-1)ヒトiPS細胞株である201B7株(iPSアカデミアジャパン社製)を、iMatrix(ニッピ社製)をコートしたポリスチレンプレート(培養基材)上で、StemFit AK02N培地(タカラバイオ社製)を用いて培養した。
(1-2)PBS(リン酸緩衝生理食塩水)で洗浄後、TrypLE select(Thermo Fisher社製)とVersene Solution(Thermo Fisher社製)との混合液を添加し、10分間37℃で静置することによって、培養基材よりiPS細胞を剥離した。
(1-3)剥離したiPS細胞を、StemFit AK02N培地にY-27632(富士フイルム和光純薬社製:濃度10μM)を添加した培地(以下、StemF+Y培地)で回収した(iPS細胞サンプル)。
Example 1 Effect of dead cell removal by solvent replacement (1) Preparation of iPS cells (1-1) Human iPS cell line 201B7 strain (manufactured by iPS Academia Japan) on a polystyrene plate coated with iMatrix (manufactured by Nippi) (culture substrate) using StemFit AK02N medium (manufactured by Takara Bio Inc.).
(1-2) After washing with PBS (phosphate buffered saline), a mixed solution of TrypLE select (manufactured by Thermo Fisher) and Versene Solution (manufactured by Thermo Fisher) was added and left at 37°C for 10 minutes. By doing so, the iPS cells were detached from the culture substrate.
(1-3) The detached iPS cells were collected in a StemFit AK02N medium to which Y-27632 (manufactured by Fujifilm Wako Pure Chemical Industries, Ltd.: concentration 10 μM) was added (hereinafter referred to as StemF+Y medium) (iPS cell sample).
(2)蛍光標識による染色
(2-1)(1)で作製した「iPS細胞サンプル」を、細胞核染色試薬であるHoechst 33342(富士フイルム和光純薬社製、1:500)と死細胞染色試薬であるSYTOX Green(Thermo Fisher社製、1:2000)とを含むStemF+Y培地に30分間室温で浸し染色した。
(2-2)染色後、250mM Sucrose(富士フイルム和光純薬社製)、10mM HEPES(4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid、同仁化学研究所社製)および1%(w/v)BSA(Merck社製)を含む電気伝導度の低い等張液(以下、DEPB1)を添加し、遠心分離(160×g、5分間)した。
(2-3)上清を除去し、新たにDEPB1を添加して遠心分離する操作を2回繰り返することで洗浄作業を行ない、得られたペレットをDEPB1に懸濁させた。
(2) Staining with fluorescent labeling (2-1) The “iPS cell sample” prepared in (1) was treated with cell nucleus staining reagent Hoechst 33342 (manufactured by Fujifilm Wako Pure Chemical Industries, Ltd., 1:500) and dead cell staining reagent. The cells were stained by soaking in StemF+Y medium containing SYTOX Green (Thermo Fisher, 1:2000) for 30 minutes at room temperature.
(2-2) After staining, 250mM Sucrose (manufactured by Fujifilm Wako Pure Chemical Industries, Ltd.), 10mM HEPES (4-(2-hydroxyethyl)-1-piperazinethanesulfonic acid, manufactured by Dojindo Laboratories) and 1% (w/v ) An isotonic solution with low electrical conductivity (hereinafter referred to as DEPB1) containing BSA (manufactured by Merck) was added and centrifuged (160×g, 5 minutes).
(2-3) Washing was performed by removing the supernatant, adding DEPB1 again, and centrifuging twice, and the resulting pellet was suspended in DEPB1.
(3)誘電泳動の実施と溶媒置換前の細胞画像取得
(3-1)図1および図2に示す構成からなる基板100に(2-3)で得られた懸濁液を導入した。本実施例で用いた基板100が有する細胞保持手段10には、直径30μm、深さ40μmの保持部50を中心間距離50μmの間隔で格子状に設けている。
(3-2)信号発生器40より周波数1KHzの交流電圧を、電極21・22に10分間印加することで、誘電泳動力を用いた保持部50への細胞保持を行なった。
(3-3)保持部50に保持された細胞の有する蛍光(Hoechst 33342およびSYTOX Green)を、蛍光顕微鏡(IX83、オリンパス社製)を用いてCMOSカメラで撮影した。
(3) Implementation of dielectrophoresis and acquisition of cell images before solvent replacement (3-1) The suspension obtained in (2-3) was introduced into the substrate 100 having the configuration shown in FIGS. 1 and 2. In the cell holding means 10 of the substrate 100 used in this example, holding portions 50 each having a diameter of 30 μm and a depth of 40 μm are provided in a lattice shape with a center-to-center distance of 50 μm.
(3-2) Cells were retained in the holding part 50 using dielectrophoretic force by applying an AC voltage with a frequency of 1 KHz from the signal generator 40 to the electrodes 21 and 22 for 10 minutes.
(3-3) Fluorescence (Hoechst 33342 and SYTOX Green) possessed by the cells held in the holding part 50 was photographed with a CMOS camera using a fluorescence microscope (IX83, manufactured by Olympus).
(4)溶媒置換
基板100が有する導入口13aからDEPB1またはミネラルオイル(Merck社製)を導入し、排出口13bから基板内余剰溶液を排出することで、保持部50を含む基板内空間の溶液全てを置換した。
(4) Solvent Replacement By introducing DEPB1 or mineral oil (manufactured by Merck) from the inlet 13a of the substrate 100 and discharging the surplus solution inside the substrate from the outlet 13b, the solution in the space inside the substrate including the holding part 50 is removed. Replaced everything.
(5)溶媒置換後の細胞画像取得
保持部50に保持された細胞の有する蛍光(Hoechst 33342およびSYTOX Green)を、蛍光顕微鏡(IX83、オリンパス社製)を用いてCMOSカメラで撮影した。
(5) Cell image acquisition after solvent replacement Fluorescence (Hoechst 33342 and SYTOX Green) possessed by cells held in the holding unit 50 was photographed with a CMOS camera using a fluorescence microscope (IX83, manufactured by Olympus).
(6)溶媒置換前後での細胞の移動率算出
(3)および(5)で取得した細胞数を比較し、以下に示す除去率を算出した。また、Hoechst 33342陽性の細胞は生細胞を示し、SYTOX Green陽性の細胞は、死細胞を示す。
除去率[%]=100×[1-{(5)溶媒置換後の細胞数/(3)溶媒置換前の細胞数}]
結果を図5に示す。(4)の溶媒置換をミネラルオイルで行なうことで、死細胞を選択的に除去できた(死細胞除去率:27.61%、生細胞除去率:3.13%)。一方、DEPB1で溶媒置換したときは、死細胞、生細胞ともほとんど除去されなかった(死細胞除去率:0.59%、生細胞除去率:0.24%)。
(6) Calculation of cell migration rate before and after solvent replacement The cell numbers obtained in (3) and (5) were compared, and the removal rate shown below was calculated. Furthermore, Hoechst 33342-positive cells indicate live cells, and SYTOX Green-positive cells indicate dead cells.
Removal rate [%] = 100 × [1-{(5) Number of cells after solvent replacement/(3) Number of cells before solvent replacement}]
The results are shown in Figure 5. By replacing the solvent in (4) with mineral oil, dead cells could be selectively removed (dead cell removal rate: 27.61%, live cell removal rate: 3.13%). On the other hand, when the solvent was replaced with DEPB1, almost no dead cells or live cells were removed (dead cell removal rate: 0.59%, live cell removal rate: 0.24%).
実施例2 溶媒置換による保持部への細胞保持効果
(1)MCR-5細胞の調製
iPS細胞よりサイズの大きな細胞としてMRC-5細胞(ヒト胎児肺由来線維芽細胞:JCRB細胞バンクより分譲)(直径30μm程度)を選択した。MRC-5細胞を、10%(w/v)FBS(Fetal Bovine Serum)を含むDMEM(Dulbecco’s Modified Eagle Medium)(富士フイルム和光純薬社製)を用いて培養後、実施例1(1-2)と同様な方法で剥離し、実施例1(1-3)と同様な方法で回収した(MRC-5細胞サンプル)。
Example 2 Cell retention effect in the holding part by solvent replacement (1) Preparation of MCR-5 cells MRC-5 cells (human fetal lung-derived fibroblasts: provided by JCRB cell bank) as cells larger in size than iPS cells ( 30 μm in diameter) was selected. After culturing MRC-5 cells in DMEM (Dulbecco's Modified Eagle Medium) (manufactured by Fujifilm Wako Pure Chemical Industries, Ltd.) containing 10% (w/v) FBS (Fetal Bovine Serum), they were cultured in Example 1 (1). It was detached in the same manner as in Example 1 (1-3) and collected in the same manner as in Example 1 (1-3) (MRC-5 cell sample).
(2)細胞核染色試薬による染色
(2-1)実施例1(1)で作製した「iPS細胞サンプル」または本実施例(1)で作製した「MRC-5細胞サンプル」を、細胞核染色試薬Hoechst 33342(商品名)を含むStemF+Y培地に30分間室温で浸すことで染色した。
(2-2)染色操作後、DEPB1を添加し、遠心分離(160×g、5分間)した。
(2-3)上清を除去し、新たにDEPB1を添加して遠心分離する操作を2回繰り返することで洗浄作業を行ない、得られたペレットをDEPB1に懸濁させた。
(2) Staining with a cell nucleus staining reagent (2-1) The “iPS cell sample” prepared in Example 1 (1) or the “MRC-5 cell sample” prepared in this Example (1) was stained with a cell nucleus staining reagent Hoechst. The cells were stained by immersion in StemF+Y medium containing 33342 (trade name) for 30 minutes at room temperature.
(2-2) After the staining operation, DEPB1 was added and centrifuged (160×g, 5 minutes).
(2-3) Washing was performed by removing the supernatant, adding DEPB1 again, and centrifuging twice, and the resulting pellet was suspended in DEPB1.
(3)保持部への細胞保持
(3-1)図1および図2に示す基板100に(3)で得られた懸濁液を導入した。前記基板100は、直径30μm、深さ40μmの保持部50を中心間距離50μmの間隔で格子状に設けている。
(3-2)信号発生器40より周波数1KHzの交流電圧を、電極21・22に10分間印加することで、誘電泳動力を用いた保持部50への細胞保持を行なった。
(3) Cell retention in the holding part (3-1) The suspension obtained in (3) was introduced into the substrate 100 shown in FIGS. 1 and 2. The substrate 100 has holding portions 50 each having a diameter of 30 μm and a depth of 40 μm arranged in a lattice shape with a center-to-center distance of 50 μm.
(3-2) Cells were retained in the holding part 50 using dielectrophoretic force by applying an AC voltage with a frequency of 1 KHz from the signal generator 40 to the electrodes 21 and 22 for 10 minutes.
(4)溶媒置換
基板100に設けた導入口13aから、DEPB1またはミネラルオイル(Merck社製)を導入し、排出口13bから余剰溶液を排出することによって保持部50内空間の溶媒を置換した。
(4) Solvent Replacement DEPB1 or mineral oil (manufactured by Merck) was introduced from the inlet 13a provided on the substrate 100, and the excess solution was discharged from the outlet 13b to replace the solvent in the space inside the holding part 50.
(5)細胞画像取得
保持部50に保持された細胞が有する、細胞核染色試薬(Hoechst 33342)由来の蛍光を、蛍光顕微鏡を用いてCMOSカメラで撮影した。当該撮影後、電極22を除去し、再び蛍光顕微鏡を用いて蛍光染色画像を撮影した。電極22除去前後における細胞の位置を比較し、以下に示す式で移動率を算出した。
移動率[%]=100×[1-{電極除去前後で位置が移動しなかった細胞数/電極除去前で検出した細胞数}]
結果を図6に示す。(4)の溶媒置換をDEPB1で行なったとき、電極22除去操作により、iPS細胞では3.79%が、MRC-5細胞では23.65%が、それぞれ移動した。一方、(4)の溶媒置換をミネラルオイルで行なったときは、電極22除去操作を行なっても、iPS細胞で0.1%、MRC-5細胞で1.67%と、ほとんど移動しなかった。
(5) Cell image acquisition The fluorescence derived from the cell nucleus staining reagent (Hoechst 33342) possessed by the cells held in the holding unit 50 was photographed with a CMOS camera using a fluorescence microscope. After the photographing, the electrode 22 was removed, and a fluorescence-stained image was photographed again using a fluorescence microscope. The positions of the cells before and after the electrode 22 was removed were compared, and the migration rate was calculated using the formula shown below.
Migration rate [%] = 100 × [1 - {number of cells whose position did not move before and after electrode removal/number of cells detected before electrode removal}]
The results are shown in FIG. When the solvent replacement in (4) was performed with DEPB1, 3.79% of iPS cells and 23.65% of MRC-5 cells migrated due to the electrode 22 removal operation. On the other hand, when the solvent replacement in (4) was performed with mineral oil, there was almost no migration, 0.1% for iPS cells and 1.67% for MRC-5 cells, even after the electrode 22 removal operation. .
実施例3 天板除去に伴う細胞の移動率に対する流動パラフィン置換の効果
(1)実施例1(1)で作製した「iPS細胞サンプル」を、実施例2(2)に記載の方法で細胞核染色試薬で染色し、実施例2(3)に記載の方法で保持部へ細胞を保持させた。
(2)置換溶媒を、250mM Sucrose、2.5mM HEPES、0.5 mM MgCl2(ナカライテスク社製)および1%(v/v)Pluronic F―68(Thermo Fisher社製)を含む電気伝導度の低い等張液(以下、DEPB2)、または流動パラフィン(低粘度タイプ:ナカライテスク社製)とした他は、実施例2(4)と同様な方法で溶媒置換した。
(3)実施例2(5)と同様な方法で、電極22除去前後の蛍光画像を取得し、移動率を算出した。
結果を図7に示す。(2)の溶媒置換をDEPB2で行なったとき、電極22除去操作により6.65%が移動していたが、(2)の溶媒置換を流動パラフィンで行なったときは、電極22除去操作を行なっても移動率は0.15%であった。
Example 3 Effect of liquid paraffin replacement on cell migration rate due to top plate removal (1) Cell nucleus staining of the “iPS cell sample” prepared in Example 1 (1) by the method described in Example 2 (2) The cells were stained with a reagent and held in the holding part by the method described in Example 2 (3).
(2) The substitution solvent was an electrical conductivity solution containing 250mM Sucrose, 2.5mM HEPES, 0.5mM MgCl 2 (manufactured by Nacalai Tesque) and 1% (v/v) Pluronic F-68 (manufactured by Thermo Fisher). The solvent was replaced in the same manner as in Example 2 (4), except that a low isotonic solution (hereinafter referred to as DEPB2) or liquid paraffin (low viscosity type: manufactured by Nacalai Tesque) was used.
(3) In the same manner as in Example 2 (5), fluorescence images before and after removal of the electrode 22 were obtained, and the migration rate was calculated.
The results are shown in FIG. When the solvent replacement in (2) was performed with DEPB2, 6.65% was transferred due to the electrode 22 removal operation, but when the solvent replacement in (2) was performed with liquid paraffin, the electrode 22 removal operation was performed. However, the migration rate was 0.15%.
100:基板
10:細胞導入保持手段
11:遮光部材
12:絶縁体
11a、12a:貫通孔
13:スペーサ
13a:導入口
13b:排出口
13c:貫通部
21・22:電極基板
30:導線
40:信号発生器(交流電圧)
50:保持部
60:誘電泳動力
200:細胞
210:検出対象細胞
220:夾雑細胞
300:標識タンパク質
400:検出部(蛍光顕微鏡)
500:キャピラリ
100: Substrate 10: Cell introduction and holding means 11: Light shielding member 12: Insulator 11a, 12a: Through hole 13: Spacer 13a: Inlet port 13b: Outlet port 13c: Penetration portion 21 and 22: Electrode substrate 30: Conductive wire 40: Signal Generator (AC voltage)
50: Holding part 60: Dielectrophoretic force 200: Cell 210: Detection target cell 220: Contaminant cell 300: Labeled protein 400: Detection part (fluorescence microscope)
500: Capillary
Claims (3)
前記細胞を含む試料を、前記導入口より前記保持部に導入する工程と、
前記細胞を前記保持部に誘電泳動力を用いて保持させる工程と、
前記導入口からミネラルオイルまたは流動パラフィンである置換溶媒を導入し、前記試料の溶媒を排出口から排出させる工程と、
を含む前記方法。 A method for holding cells using a cell holding device equipped with a substrate having a holding part capable of holding cells, an inlet, and an outlet, the method comprising:
introducing the sample containing the cells into the holding part from the introduction port;
holding the cells in the holding section using dielectrophoretic force ;
Introducing a displacement solvent such as mineral oil or liquid paraffin from the inlet and discharging the sample solvent from the outlet;
The method comprising:
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2019234568A JP7443762B2 (en) | 2019-12-25 | 2019-12-25 | How to retain cells contained in the sample |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2019234568A JP7443762B2 (en) | 2019-12-25 | 2019-12-25 | How to retain cells contained in the sample |
Publications (2)
Publication Number | Publication Date |
---|---|
JP2021101659A JP2021101659A (en) | 2021-07-15 |
JP7443762B2 true JP7443762B2 (en) | 2024-03-06 |
Family
ID=76754518
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
JP2019234568A Active JP7443762B2 (en) | 2019-12-25 | 2019-12-25 | How to retain cells contained in the sample |
Country Status (1)
Country | Link |
---|---|
JP (1) | JP7443762B2 (en) |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2011149032A1 (en) | 2010-05-26 | 2011-12-01 | 東ソー株式会社 | Biological-sample affixing device |
-
2019
- 2019-12-25 JP JP2019234568A patent/JP7443762B2/en active Active
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2011149032A1 (en) | 2010-05-26 | 2011-12-01 | 東ソー株式会社 | Biological-sample affixing device |
Non-Patent Citations (4)
Title |
---|
化学工学,2017年,Vol. 81, No. 3,p. 126-128 |
東ソー研究・技術報告,2015年,Vol. 59,p. 3-9 |
東ソー研究・技術報告,2018年,Vol. 62,p. 89-92 |
東ソー研究・技術報告,Vol. 58,2014年,p. 3-12 |
Also Published As
Publication number | Publication date |
---|---|
JP2021101659A (en) | 2021-07-15 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US9109197B2 (en) | Device for concentrating and separating cells | |
JP2019141081A (en) | System and method for sorting sperm | |
CN107304414B (en) | Device for purifying and separating blood circulation tumor cells by using non-contact and automatic identification | |
Davies | Cell separations by flow cytometry | |
US20110123979A1 (en) | Detection of microorganisms | |
JP7443762B2 (en) | How to retain cells contained in the sample | |
Kiryo et al. | Purification of pluripotent embryonic stem cells using dielectrophoresis and a flow control system | |
JP6686361B2 (en) | Specimen preparation method for cell recovery | |
JP6421589B2 (en) | Fine particle capture device | |
JP6572547B2 (en) | Fine particle recovery device | |
Feng et al. | Sorting Technology for Mesenchymal Stem Cells from a Single Tissue Source | |
US20200110054A1 (en) | Microfluidic devices and methods using the same | |
JP2018028457A (en) | Method and device for collecting target particle | |
JP6198186B2 (en) | Method for separating target cells | |
JP7062901B2 (en) | How to detect target cells | |
JP7413692B2 (en) | Pretreatment method for samples containing cells | |
JP6685387B2 (en) | Cell detection method | |
JP2018157812A (en) | Detection method of cells included in sample | |
JP2021019513A (en) | Method for analyzing target particles | |
Segeren et al. | Collection of cells for single-cell RNA sequencing using high-resolution fluorescence microscopy | |
Mukherjee et al. | Isolation and Purification of Various Mammalian Cells: Single Cell Isolation | |
JP2016106622A (en) | Separation and recovery method of cells | |
JP2018021895A (en) | Method of detecting and recovering undifferentiated cell | |
JP2018134026A (en) | Cell holding method | |
JP6686357B2 (en) | Cell recovery method |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
A621 | Written request for application examination |
Free format text: JAPANESE INTERMEDIATE CODE: A621 Effective date: 20221116 |
|
A131 | Notification of reasons for refusal |
Free format text: JAPANESE INTERMEDIATE CODE: A131 Effective date: 20231003 |
|
A521 | Request for written amendment filed |
Free format text: JAPANESE INTERMEDIATE CODE: A523 Effective date: 20231129 |
|
TRDD | Decision of grant or rejection written | ||
A01 | Written decision to grant a patent or to grant a registration (utility model) |
Free format text: JAPANESE INTERMEDIATE CODE: A01 Effective date: 20240123 |
|
A61 | First payment of annual fees (during grant procedure) |
Free format text: JAPANESE INTERMEDIATE CODE: A61 Effective date: 20240205 |
|
R151 | Written notification of patent or utility model registration |
Ref document number: 7443762 Country of ref document: JP Free format text: JAPANESE INTERMEDIATE CODE: R151 |