WO2022244846A1 - 細胞増殖の調整のための組成物及び細胞増殖の調整方法 - Google Patents

細胞増殖の調整のための組成物及び細胞増殖の調整方法 Download PDF

Info

Publication number
WO2022244846A1
WO2022244846A1 PCT/JP2022/020851 JP2022020851W WO2022244846A1 WO 2022244846 A1 WO2022244846 A1 WO 2022244846A1 JP 2022020851 W JP2022020851 W JP 2022020851W WO 2022244846 A1 WO2022244846 A1 WO 2022244846A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition according
amino acids
serine
amount
cell proliferation
Prior art date
Application number
PCT/JP2022/020851
Other languages
English (en)
French (fr)
Japanese (ja)
Inventor
友則 木村
篤 部坂
悠介 塚本
真史 三田
Original Assignee
Kagami株式会社
国立研究開発法人医薬基盤・健康・栄養研究所
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 Kagami株式会社, 国立研究開発法人医薬基盤・健康・栄養研究所 filed Critical Kagami株式会社
Priority to JP2023522721A priority Critical patent/JPWO2022244846A1/ja
Publication of WO2022244846A1 publication Critical patent/WO2022244846A1/ja

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/90Isomerases (5.)

Definitions

  • the present invention relates to a composition for modulating cell proliferation and a method for modulating cell proliferation.
  • Cells are generally recognized as the most basic building blocks of living organisms. They have organs responsible for metabolism such as the glycolytic system and the citric acid cycle for constant life activities, and contain genetic information for self-renewal and replication. It has the ability to express it. Humans are composed of more than 30 trillion somatic and germ cells.
  • somatic cells There are about 200 types of somatic cells that account for most of them, and a proliferating dividing cell group that constantly divides and proliferates (e.g., myeloblasts, basal cells, etc.), a differentiated dividing cell group that divides and proliferates while differentiating (e.g., myeloid cells, neuroblasts, myoblasts, etc.), reversible postmitotic cell groups that do not normally proliferate (e.g., stem cells, smooth muscle cells, lymphocytes, etc.), fixed postmitotic cell groups that have lost their ability to divide (nerve cells, myocardial cells, etc.) cells, red blood cells, etc.).
  • a proliferating dividing cell group that constantly divides and proliferates
  • a differentiated dividing cell group that divides and proliferates while differentiating
  • reversible postmitotic cell groups that do not normally proliferate
  • stem cells smooth muscle cells, lymphocytes, etc.
  • cells that repeat regeneration, function, and death under normal conditions e.g., epithelial cells, blood cells, etc.
  • Cells that proliferate and regenerate under special circumstances e.g., organ parenchymal cells, vascular endothelial cells, connective tissue, etc.
  • highly organized and differentiated cells that do not have the ability to spontaneously generate and proliferate in non-regenerative tissues.
  • cell types such as (eg, nerve cells, skeletal muscle, cardiomyocytes, etc.).
  • a tumor is a cell that autonomously proliferates against normal control due to some abnormality in the gene of the cell, and a malignant tumor (cancer) is a tumor that invasively proliferates and metastasizes.
  • Non-Patent Document 1 describes that the metabolism and regulation of amino acids regulates the proliferative potential of human pluripotent stem cells.
  • D-amino acids are affected by ingestion, symbiotic bacteria, metabolism (decomposition, synthesis), transportation, excretion, etc.
  • Non-Patent Documents 2 to 6 and the amount in living organisms, tissues, cells, and body fluids changes, and kidney disease , showing a chiral amino acid profile characteristic of diseases such as heart disease and diabetes (Patent Document 1), furthermore, the involvement of D-amino acids in intestinal immunity (Non-Patent Document 7) and protecting kidney-derived cells A phenomenon has been reported (Non-Patent Document 3). There is also a report that carbohydrate metabolism is involved in D-serine biosynthesis in nerve cells (Non-Patent Document 8).
  • Patent Document 1 It has been disclosed that the amounts of D-serine and D-alanine in the blood vary in renal cancer, and the amounts of D-alanine, D-proline and D-aspartic acid in the blood vary in diabetes (Patent Document 1. , Patent Document 2). It has also been reported that D-alanine is localized in cells containing insulin in pancreatic islets of Langerhans and cells containing adrenocorticotropic hormone in the anterior pituitary gland (Non-Patent Documents 9 and 10). However, the relationship between the presence of D-amino acids and cell proliferation has not yet been elucidated.
  • Nagamori D-Serine, an emerging biomarker of kidney diseases , is a hidden substrate of sodium-coupled monocarboxylate transporters, bioRxiv preprint. DOI: 10.1101/2020.08.10.244822 A. Hesaka, S. Sakai, K. Hamase, T. Ikeda, R. Matsui, M. Mita, M. Horio, Y. Isaka and T. Kimura, D-Serine reflects kidney function and diseases, Scientific Reports, 9( 2019). DOI: 10.1038/s41598-019-41608-0 J. Sasabe, Y. Miyoshi, S. Rakoff-Nahoum, T. Zhang, M. Mita, B.M.
  • the inventors adjusted the amount of D-amino acid and L-amino acid in the medium in primary or subcultures of renal tubular cells and fibroblasts, and found that D-amino acids promoted cell proliferation more than L-amino acids. I discovered that it works. As a result of intensive research on its effect and mechanism, a method for adjusting the growth and regeneration of cells, tissues and organs inside and outside the body, especially conditionally regenerative tissues, was found by increasing or decreasing the amount of D-amino acids. Arrived. That is, the present invention includes the following inventions.
  • a composition for regulating cell growth comprising an agent for controlling the amount of D-amino acids.
  • the control agent is an agent for controlling the amount of D-amino acids in the subject's living body.
  • the cell proliferation is cell proliferation in a living tissue and/or organ, thereby adjusting the size of the living tissue and/or organ.
  • the composition according to item 2 wherein the subject is a subject with kidney disease.
  • the composition according to item 2, wherein the subject is a kidney transplant donor and/or recipient.
  • the composition according to item 2 which improves renal function.
  • the composition according to item 6, wherein the renal function is glomerular filtration rate.
  • a composition according to item 1 for modulating the growth of isolated cells [9] The composition according to any one of items 1 to 8, wherein the control agent is a D-amino acid or a derivative thereof. [10] The composition according to item 9, wherein the D-amino acid is selected from the group consisting of D-serine, D-asparagine and D-glutamine. [11] The composition according to any one of items 1 to 8, wherein the control agent is an agent that modulates protein activity related to absorption, transport, distribution, metabolism or excretion of D-amino acids. [12] The composition according to item 11, wherein the metabolism is degradation or synthesis.
  • composition according to item 11 or 12, wherein the agent that modulates the activity of the protein is an agent that regulates gene expression of the protein.
  • the protein is selected from the group consisting of D-aspartate oxidase and serine isomerase.
  • the protein is a D-amino acid transporter protein.
  • the D-amino acid transporter protein is one or more selected from the group consisting of SMCT family, GLUT5, CAT1, THTR2, SNAT2, ASCT family, Asc1, PAT1 and ATB 0,+ Composition.
  • composition according to any one of items 1 to 16, which is a pharmaceutical is a pharmaceutical.
  • composition according to any one of Items 1 to 16, which is a food is a food with health claims or a dietary supplement.
  • the control agent is an agent for controlling the amount of D-amino acid in the subject's living body.
  • the cell proliferation is cell proliferation in a living tissue and/or organ, thereby adjusting the size of the living tissue and/or organ. thing.
  • the composition of item 22-2, wherein the subject is a subject with renal disease.
  • the composition according to item 22-2, wherein the subject is a kidney transplant donor and/or recipient.
  • composition according to item 22-6 wherein the renal function is glomerular filtration rate.
  • the composition according to item 22-1 for modulating growth of isolated cells.
  • the control agent is a D-amino acid or a derivative thereof.
  • the D-amino acid is selected from the group consisting of D-serine, D-asparagine and D-glutamine.
  • the control agent is an agent that modulates protein activity related to D-amino acid absorption, transport, distribution, metabolism or excretion; The described composition.
  • composition according to item 22-11 wherein the metabolism is degradation or synthesis.
  • the agent that modulates the activity of the protein is an agent that regulates gene expression of the protein.
  • the protein is selected from the group consisting of D-aspartate oxidase and serine isomerase.
  • the protein is a D-amino acid transporter protein.
  • a method of modulating cell proliferation in a subject comprising: A method comprising administering to a subject in need thereof an agent for controlling the amount of D-amino acids in vivo.
  • a method of modulating cell proliferation in vitro or ex vivo comprising: A method comprising applying a D-amino acid amount controlling agent to a cell, biological tissue or organ and culturing.
  • the present invention by controlling the amount of D-amino acids, it is possible to regulate cell proliferation. It becomes possible to treat or evaluate.
  • FIG. 1 shows the effect of D, L-amino acids (10 ⁇ M) on cell growth in culture.
  • FIG. 2 shows the effect of D, L-amino acids (1 ⁇ M) on cell growth in culture.
  • FIG. 3 shows the effects of glycylserine isomers (10 ⁇ M) on cell growth in culture.
  • FIG. 4 shows administration of D, L-amino acids and uptake into each organ. Ditto.
  • Statistical analysis was performed by two-way ANOVA. *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001. Data are shown as mean ⁇ SEM.
  • FIG. 6 shows the results of pathway analysis on the effects of D-serine administration. Ditto.
  • FIG. 7 shows the results of D-serine administration and cell cycle-related gene expression analysis.
  • FIG. 8 shows Ki67 positive nuclei analysis by D-serine administration.
  • A Representative images of renal cortex stained with anti-Ki67 antibody. The lower left figure in each figure is an enlarged view of the area enclosed by the rectangle in the figure. Scale bar: 50 ⁇ m.
  • FIG. 9 shows the effect of D-serine administration on proliferation of proximal tubule cells.
  • Lectin-positive proximal tubule green, Ki67: red, DAPI (nucleus): blue.
  • Scale bar 50 ⁇ m.
  • FIG. 10 shows the effect of D-serine administration on glomerular cell proliferation.
  • FIG. 11 shows the proliferative effect of D-serine administration on renal tubular cells and HK-2 cells.
  • A Normal human primary renal proximal tubule cells (RPTEC) or
  • B HK-2 cells cultured with 10 ⁇ M D-serine relative proliferation rate (%) over time.
  • C Normal human primary renal proximal tubule cells (RPTEC) or
  • D HK-2 cells cultured with various concentrations of D-serine relative proliferation rate (%).
  • FIG. 12 shows the results of phosphorylation analysis of HK-2 cells by D-serine administration.
  • A Western blot results of phosphorylation of S6K at Thr389 (phospho-S6K (p-S6K)) of HK-2 cells treated with 5 ⁇ M D- or L-serine for 10 min in serine-free medium. show.
  • FIG. 13 shows the results of phosphorylation analysis of mouse kidney cells by administration of D-serine.
  • A Fed on a serine-free diet and water with or without 0.1% D-serine for 1 week and sacrificed 2 days after unilateral nephrectomy (UNX) or not. Immunoblot of phospho-S6 ribosomal protein (p-S6RP) at Ser235/236 from renal cortex of week-old C57 BL/6 male mice.
  • FIG. 14 shows the effect of D-serine administration on cell proliferation in p18-deficient strains.
  • Statistical analysis was performed by two-way ANOVA. ***p ⁇ 0.001.
  • FIG. 15 shows the effect of D-serine administration on cell proliferation in Rheb-deficient strains.
  • FIG. 16 shows cell proliferation effects by administration of D-serine and rapamycin or PI3 inhibitors.
  • Statistical analysis was performed by two-way ANOVA. *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001.
  • (B) Relative growth rates of HK-2 cells treated simultaneously with or without the indicated concentrations of D-serine and Ly294002 for 6 hours using serine-free medium and D-serine untreated. relative growth rate of the corresponding cell lines. N 21-24.
  • FIG. 17 shows the results of AKT phosphorylation analysis by D-serine administration.
  • A Fed on a serine-free diet and water with or without 0.1% D-serine for 1 week and sacrificed 2 days after unilateral nephrectomy (UNX) or not. Immunoblot results for phospho-AKT (p-AKT) at Ser473 from renal cortex of week-old C57 BL/6 male mice are shown (left).
  • B Plotted the relative index (RI) of three independent experiments. Statistical analysis was performed by one-way ANOVA using Dunnett's post-hoc test.
  • FIG. 18 shows localization analysis of mTOR lysosomes by D-serine administration.
  • A High-content microscopy quantification of co-localization of mTOR and LAMP2 in HK-2 cells that were incubated in culture medium followed by amino acid starvation in the presence of 5 ⁇ M D- or L-serine for 10 minutes.
  • Statistical analysis was performed by one-way ANOVA using Bonferroni's post-hoc test.
  • NS not significantly different, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001.
  • B Quantification by high-content microscopy of co-localization of mTOR and LAMP1 in HK-2 cells that were incubated in culture medium followed by amino acid starvation in the presence of 5 ⁇ M D- or L-serine for 10 minutes.
  • Mask overlay primary object (algorithm-defined cell boundaries based on CellMask (red pseudocolor)), and internal secondary object (computationally defined co-localization between mTOR (green) and LAMP1 (yellow)).
  • Statistical analysis was performed by one-way ANOVA using Bonferroni's post-hoc test.
  • NS not significantly different, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001.
  • FIG. 19 shows a schematic representation of the activation scheme of mTOR-related pathways by D-serine.
  • A Schematic showing that D-serine activates cell proliferation through the mTOR-related pathway (mTOR/p18/Rheb). D-serine can mediate renal remodeling after unilateral nephrectomy.
  • B Schematic representation of activation of mTORC1 signaling by D-serine. D-serine enhances the signal from L-amino acids to mTORC1 activation. D-serine also activates mTROC1 through the phosphoinositide 3-kinase (PI3K)/Rheb pathway.
  • FIG. 20 shows the effect of administration of D-serine on increasing glomerular filtration rate (GFR).
  • FIG. 21 shows the results of examining the cell proliferation ability of D-amino acids in B cells.
  • One embodiment of the present invention provides a composition for modulating cell growth, comprising an agent for controlling the amount of D-amino acids.
  • one embodiment of the present invention provides a composition that activates the mTOR-related pathway, comprising a regulator of the amount of D-amino acids.
  • one embodiment of the present invention provides a method for regulating cell proliferation, a method for regulating tissue/organ size, or a tissue/organ function, comprising regulating the amount of D-amino acids in a subject. Provide a way to improve.
  • controlling the amount of D-amino acids refers to intentionally increasing or decreasing the amount of D-amino acids inside or outside cells, tissues and/or organs, and in body fluids. If there is a target amount and concentration, the D-amino acid in the sample and D-amino acid clearance may be evaluated by monitoring as appropriate.
  • cell proliferation may be cell proliferation in a living tissue and/or organ, and may thereby adjust the size of the living tissue and/or organ.
  • the term "adjustment of size of biological tissue and/or organ” means that the amount, morphology, abnormalities, excesses, deficiencies, etc. of cells in a biological tissue and/or organ are brought into a balanced state or in a correct state. It means to Size can be expressed in units of measurement such as physical size, weight, and function, and may be corrected for weight and the like. When there is a desired effect, for example, when the purpose is to grow cultured cells, the effect may be evaluated by performing metabolome analysis of the cell count or medium.
  • markers in urine tests (urine protein, hematuria, creatinine amount, etc.) and blood tests (creatinine, cystatin C, urea nitrogen: BUN, etc.) for the purpose of adjusting kidney cell proliferation, size, and function
  • renal function test renal blood flow: RPF, glomerular filtration rate: GFR
  • X-ray examination angiography, ultrasonography, CT, MRI
  • nuclear medicine examination endoscopy
  • renal biopsy The effect may be evaluated by performing (pathological examination) or the like.
  • D-amino acid amount control agent means that it is applied (eg, administered) to a subject in vivo. (eg, in a cell, tissue, organ, or body fluid), or an agent capable of increasing or decreasing the amount of D-amino acids in an isolated cell.
  • regulating the amount of D-amino acids in cells means increasing or decreasing the amount of D-amino acids in cells by applying a D-amino acid amount controlling agent, It means adjusting the amount of amino acid to an arbitrary range.
  • regulating the amount of D-amino acids in tissues means that the amount of D-amino acids in tissues (e.g., renal tubules, glomeruli, etc.) is It means adjusting the amount of D-amino acids to any range by increasing or decreasing the amount.
  • regulating the amount of D-amino acids in an organ means that the amount of D-amino acids in an organ (for example, kidney, heart, etc.) is controlled by applying a D-amino acid amount controlling agent. It means adjusting the amount of D-amino acids to any range by increasing or decreasing the amount.
  • regulating the amount of D-amino acids in body fluids means that the amount of D-amino acids in body fluids (e.g., blood, urine, etc.) is reduced by applying a D-amino acid amount controlling agent. It means adjusting the amount of D-amino acids to any range by increasing or decreasing the amount.
  • D-amino acid is meant to include "D-form" proteinogenic amino acids, which are stereoisomers of "L-form” proteinogenic amino acids, and glycine without stereoisomers. Specifically, glycine, D-alanine, D-histidine, D-isoleucine, D-allo-isoleucine, D-leucine, D-lysine, D-methionine, D-phenylalanine, D-threonine, D- allo-threonine, D-tryptophan, D-valine, D-arginine, D-cysteine, D-glutamine, D-proline, D-tyrosine, D-aspartic acid, D-asparagine, D-glutamic acid, and D-serine What it contains.
  • D-cysteine contained in a biological sample is oxidized and changed to D-cystine in vitro
  • D-cysteine can be measured instead of D-cysteine.
  • the amount of D-cysteine contained in the biological sample can be calculated.
  • the amount of D-amino acids and/or the amount of L-amino acids can be measured by any method, such as chiral column chromatography, measurement using an enzymatic method, and monoclonal antibodies that distinguish optical isomers of amino acids. can be quantified by immunological techniques using Measurement of the amount of D-amino acid and/or the amount of L-amino acid in the sample in the present invention may be carried out using any method known to those skilled in the art. For example, chromatographic methods and enzymatic methods (Y. Nagata et al., Clinical Science, 73 (1987), 105. Analytical Biochemistry, 150 (1985), 238., A.
  • the separation and analysis system for optical isomers in the present invention may combine multiple separation analyses. More specifically, a step of passing a sample containing components having optical isomers through a first column packing material as a stationary phase together with a first liquid as a mobile phase to separate said components of said sample; holding each of said components of said sample individually in a multi-loop unit, each of said components of said sample held individually in said multi-loop unit as a stationary phase, with a second liquid as a mobile phase; and resolving the optical isomers contained in each of the components of the sample by feeding through a channel to a second column packing material having an optically active center of
  • the amount of D-amino acids and/or the amount of L-amino acids in a sample can be measured by using a method for analyzing optical isomers, which is characterized by including a step of detecting isomers (Japanese Patent No.
  • D- and L-amino acids were previously derivatized with fluorescent reagents such as o-phthalaldehyde (OPA) and 4-fluoro-7-nitro-2,1,3-benzoxadiazole (NBD-F). or diastereomerization using N-tert-butyloxycarbonyl-L-cysteine (Boc-L-Cys) (Kenji Hamase and Kiyoshi Zaitsu, Analytical Chemistry, Vol. 53, 677-690 ( 2004)).
  • fluorescent reagents such as o-phthalaldehyde (OPA) and 4-fluoro-7-nitro-2,1,3-benzoxadiazole (NBD-F).
  • Boc-L-Cys N-tert-butyloxycarbonyl-L-cysteine
  • D-amino acids or L-amino acids are measured by an immunological technique using monoclonal antibodies that distinguish optical isomers of amino acids, such as monoclonal antibodies that specifically bind to D-amino acids or L-amino acids. can do.
  • monoclonal antibodies that distinguish optical isomers of amino acids such as monoclonal antibodies that specifically bind to D-amino acids or L-amino acids.
  • the amino acids can be analyzed without distinguishing between the D- and L-isomers. Also in that case, it can be separated and quantified by an enzymatic method, an antibody method, GC, CE, or HPLC.
  • mTOR-related pathway refers to a signal pathway involving mTOR (mechanistic target of rapamycin), which is a type of protein kinase (serine-threonine kinase) involved in intracellular signal transduction. It may be a signal pathway containing factors involved in the activation of complex 1 (mTORC1) or mTOR complex 2 (mTORC2), such as Akt, Rheb, and the like.
  • mTORC1 complex 1
  • mTORC2 mTOR complex 2
  • the present invention integrates intracellular and extracellular environmental information such as insulin, growth factors, nutritional/energy status, redox status, etc., by activating mTOR-related pathways, and through transcription, translation, etc., cells respond accordingly. Regulates size, division, survival, etc. Their physiology may regulate cell proliferation.
  • the amount of biomolecules such as D-amino acids, creatinine, proteins, and drugs is not limited to mere mass, weight, and amount of substance (mol), but also mass per unit of tissue, cell, organ, molecule, volume, or weight. , weight, amount of substance (mol), mass in a liquid such as blood or urine, weight, amount of substance (mol), concentration, specific gravity, density, or any other physical quantity that can be measured.
  • D-amino acids administration of D-amino acids from the outside, addition or removal of D-amino acids in foods, addition or removal of D-amino acids in culture media, and , and drugs or foods that can increase or decrease the amount of D-amino acids in body fluids.
  • aqueous solution containing D-amino acids it is possible to increase the concentration of D-amino acids in blood and cells/tissues (Non-Patent Document 2).
  • D-serine may be directed to the kidney by oral or intravenous administration to control D-serine levels in the kidney (see, eg, Example 2).
  • the D-amino acid used here may contain modifications or derivatives of D-amino acids, or pharmaceutically acceptable salts thereof, as long as the amount of D-amino acid can be increased or decreased, or a pharmacologically acceptable may contain carriers, diluents or excipients that may be used, and may be in the form of a prodrug. In addition, it may contain a target organ function improving agent and the like.
  • the drug in the present invention can be formulated by selecting a dosage form suitable for its administration route. For oral administration, dosage forms such as tablets, capsules, liquids, powders, granules, chewing agents, etc., and for parenteral administration, injections, powders, infusion preparations, etc. can be designed.
  • these formulations contain various adjuvants used for pharmaceutical purposes, namely, carriers and other adjuvants such as stabilizers, preservatives, soothing agents, flavoring agents, corrigents, fragrances, emulsifiers and fillers. , a pH adjuster, etc., and can be blended within a range that does not impair the effects of the agent (composition) of the present invention.
  • the optical purity of D-amino acids used as drugs and raw materials is preferably 50% or higher, more preferably 90% or higher, but any optical purity can be selected within the range in which the effect is exhibited, It is not limited.
  • the D-amino acid amount controlling agent that can be used in the present invention is selected from the group consisting of D-serine, D-asparagine and D-glutamine, and modifications and derivatives thereof. It may be selected from the group consisting of D-serine, D-asparagine and D-glutamine, and modifications and derivatives thereof. It may be selected from the group consisting of D-serine, D-asparagine and D-glutamine, and modifications and derivatives thereof. It may be any suitable amino acid amount controlling agent that can be used in the present invention.
  • the present invention may utilize any physiological mechanism to vary the amount of target D-amino acids.
  • proteins related to absorption, transport, distribution, metabolism (synthesis/degradation), excretion, action, etc. of D-amino acids such as enzymes (D-amino acid oxidase (DAO), D-aspartate oxidase ( DDO), serine isomerase (SRR), DPP-4, etc.), transporters, and receptor expression (stimulation, suppression, etc.) and activity (action, inhibition, stimulation, etc.) regulate the amount of target D-amino acids. It becomes possible.
  • DAO inhibitors e.g., sodium benzoate, chlorpromazine, risperidone, etc.
  • D-amino acid transporter is an inhibitor/activator. increases or decreases the amount of D-amino acids in the transport source/destination.
  • Agents that act on proteins such as enzymes and transporters may not be effective directly. - It may vary in the amount of amino acids. In that case, the effect can be evaluated by measuring the amount of D-amino acid in body fluids, cells, and tissues at the site of action. In addition, such evaluations can be used to screen drug candidates.
  • any physiological mechanism can be used to vary the amount of D-amino acids in the body, and as a result, it is possible to regulate cell proliferation in the subject.
  • the expression (promotion, suppression) of proteins related to D-amino acid absorption, transport, distribution, metabolism (synthesis and / or degradation), excretion, action, etc., or D-amino acid transporters or receptors etc.) and/or activity (action, inhibition, stimulation, etc.) can control the amount of D-amino acids in the body.
  • the D-amino acid amount control agent that can be used in the present invention directly or indirectly promotes the gene expression of proteins involved in the absorption, transport, distribution, metabolism or excretion of D-amino acids.
  • it may be the protein or a vector that expresses it, or it may be a factor that promotes the upstream activity of the cascade that promotes the expression of the protein, or a vector that expresses it.
  • the D-amino acid amount control agent that can be used in the present invention is one that directly or indirectly suppresses gene expression of proteins related to absorption, transport, distribution, metabolism or excretion of D-amino acids.
  • D-amino acids from, for example, small molecules, aptamers, antibodies, antibody fragments, and antisense RNA or DNA molecules, RNAi-inducing nucleic acids, microRNAs (miRNAs), ribozymes, genome-editing nucleic acids and their expression vectors. may be selected.
  • proteins related to absorption, transport, distribution, metabolism (synthesis and/or degradation), excretion, action, etc. of D-amino acids may be enzymes, for example, D-amino acid oxidation It may be an enzyme (DAO), D-aspartate oxidase (DDO), serine isomerase (SRR), DPP-4, and the like.
  • DAO inhibitors can increase the amount of D-amino acids at the site of action by suppressing the oxidation of D-amino acids. obtain.
  • D-amino acid transporter can increase or decrease the amount of D-amino acid in the source/destination, agents that act directly or indirectly on the D-amino acid transporter can also be applied to the present invention.
  • Non-Patent Document 5 describes that as D-amino acid transporter proteins, the SMCT family, the ASCT family, etc. expressed in the brain and kidneys increase the localized amount of D-amino acids by agonists/inhibitors. It is disclosed to change Since these transporters are affected by co-transport substances (e.g., sodium ions) and coordination/competition through scaffolds, D-amino acid transport activity is reduced even by sodium/glucose co-transporter (SGLT2) inhibitors. can be controlled.
  • Patent Document 3 discloses that angiotensin 2 receptor blockers (ARBs) change the amount of D-amino acids in blood.
  • ARBs angiotensin 2 receptor blockers
  • drugs and candidates that can control the amount of D-amino acids in the body can be identified. Screening is possible.
  • aptamers refer to synthetic DNA or RNA molecules and peptidic molecules that have the ability to specifically bind to target substances, and can be chemically synthesized in vitro in a short period of time. Aptamers used in the present invention can bind to, for example, proteins involved in absorption, transport, distribution, metabolism or excretion of D-amino acids and inhibit their activity.
  • Aptamers used in the present invention can be obtained, for example, by repeatedly selecting bindings to various molecular targets such as small molecules, proteins, and nucleic acids in vitro using the SELEX method (Tuerk C., Gold L., Science, 1990, 249(4968), 505-510; Ellington AD, Szostak JW., Nature, 1990, 346(6287):818-822; No. 5,567,588; U.S. Pat. No. 6,699,843).
  • antibody fragment refers to a portion of a full-length antibody that maintains antigen-binding activity, generally including its antigen-binding domain or variable domain.
  • antibody fragments include F(ab')2, Fab', Fab or Fv antibody fragments (including scFv antibody fragments), and the like.
  • Antibody fragments also include fragments obtained by treating an antibody with a protease enzyme and optionally reducing it.
  • Antibodies or antibody fragments used in the present invention may be any of human-derived antibodies, mouse-derived antibodies, rat-derived antibodies, rabbit-derived antibodies, camelid-derived antibodies such as llamas, or goat-derived antibodies.
  • Antibodies may be monoclonal, complete or truncated (eg, F(ab')2, Fab', Fab or Fv fragments), chimerized, humanized or fully human.
  • antisense RNA or DNA molecule means a base sequence complementary to RNA (sense RNA) having a certain function, such as messenger RNA (mRNA), and forms a double strand with the sense RNA. In other words, it refers to a molecule that has the function of inhibiting protein synthesis that the sense RNA should be responsible for.
  • antisense oligonucleotides including antisense RNA or DNA molecules, inhibit translation into proteins by binding to mRNAs of proteins involved in absorption, transport, distribution, metabolism or excretion of D-amino acids. do.
  • RNAi-inducing nucleic acid refers to a polynucleotide capable of inducing RNA interference (RNAi) when introduced into a cell, usually 19-30 nucleotides, preferably 19-25 nucleotides. , more preferably RNA, DNA, or chimeric molecules of RNA and DNA containing 19-23 nucleotides, optionally modified.
  • RNAi may occur on mRNA or on post-transcriptional RNA before processing, i.e. RNA of nucleotide sequences comprising exons, introns, 3' untranslated regions and 5' untranslated regions.
  • the RNAi method that can be used in the present invention includes (1) direct introduction of short double-stranded RNA (siRNA) into cells, or (2) incorporation of small hairpin RNA (shRNA) into various expression vectors, or (3) constructing a vector that expresses siRNA by inserting a short double-stranded DNA corresponding to the siRNA into a vector having two promoters arranged in opposite directions between the promoters, and RNAi may be induced by techniques such as introduction into
  • the RNAi-inducing nucleic acid may include siRNA, shRNA, or miRNA that allows cleavage of the RNA of the D-serine transporter protein or suppression of its function, and these RNAi nucleic acids may be directly introduced using liposomes or the like. Alternatively, they may be introduced using an expression vector that directs these RNAi nucleic acids.
  • the RNAi-inducing nucleic acid for a protein associated with D-amino acid absorption, transport, distribution, metabolism or excretion used in the present invention is Any nucleic acid that exhibits a biological effect of inhibiting or significantly inhibiting protein expression can be synthesized by those skilled in the art with reference to the base sequence of the protein. For example, it is chemically synthesized using a DNA (/RNA) automatic synthesizer that utilizes DNA synthesis technology such as the solid-phase phosphoramidite method, or by an siRNA-related contract synthesis company (such as Life Technologies). It is also possible to consign and synthesize.
  • the siRNA used in the present invention is derived from its precursor, short-hairpin double-stranded RNA (shRNA), through processing by the intracellular RNase Dicer. There may be.
  • miRNA is a single-stranded RNA molecule with a length of 21 to 25 bases, and refers to a molecule involved in post-transcriptional regulation of gene expression in eukaryotes. miRNAs generally recognize the 3'UTR of mRNAs and suppress translation of target mRNAs to suppress protein production. Therefore, miRNAs that can directly and/or indirectly reduce the expression level of the D-serine transporter protein are also included in the scope of the present invention.
  • ribozyme is a generic term for enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. Some ribozymes have a size of 400 nucleotides or more, such as group I intron type and M1 RNA contained in RNase P, but there are also hammerhead-type and hairpin-type ribozymes that have an active domain of about 40 nucleotides. (See, for example, Makoto Koizumi and Eiko Otsuka, Protein Nucleic Acid Enzyme, 1990, 35, 2191).
  • the self-cleaving domain of the hammerhead ribozyme cleaves the sequence G13U14C15 at the 3' side of C15, but base pairing between U14 and A9 is important for its activity.
  • base pairing between U14 and A9 is important for its activity.
  • Hairpin-type ribozymes can also be used in the present invention.
  • This ribozyme is found, for example, in the minus strand of satellite RNA of tobacco ringspot virus (Buzayan, JM., Nature, 1986, 323, 349.). It has been shown that target-specific RNA-cleaving ribozymes can also be produced from hairpin ribozymes (for example, Kikuchi, Y. & Sasaki, N., Nucl. Acids. Res., 1991, 19, 6751; Kikuchi Hiroshi, Chemistry and Biology, 1992, 30, 112.). Expression of the D-serine transporter protein can be inhibited by specifically cleaving the transcript of the gene encoding the D-serine transporter protein using a ribozyme.
  • a genome-editing nucleic acid refers to a nucleic acid used to edit a desired gene in a nuclease-based system used for gene targeting.
  • Nucleases used for gene targeting include known nucleases as well as new nucleases that will be used for gene targeting in the future.
  • known nucleases include CRISPR/Cas9 (Ran, FA, et al., Cell, 2013, 154, 1380-1389), TALEN (Mahfouz, M., et al., PNAS, 2011, 108 , 2623-2628), ZFN (Urnov, F., et al., Nature, 2005, 435, 646-651) and the like.
  • symbiotic bacteria such as intestinal bacteria are one of the resources of D-amino acids in the body
  • administration of antibiotics, intestinal regulators, oligosaccharides, probiotics, microbial transplantation, fecal transplantation It is possible to increase or decrease the amount of D-amino acids in the living body by changing the microflora or the growth environment by means such as improvement of dysbiosis.
  • a probiotic ingestion of yogurt containing 1073R-1 lactobacillus is known to increase fecal D-serine and decrease D-lysine.
  • D-amino acids such as black vinegar, yogurt, microbial fermentation products such as cheese, and bacterial cells or bacterial cell extracts
  • microbial fermentation products such as cheese
  • bacterial cells or bacterial cell extracts have many active ingredient candidates in addition to D-amino acids. Therefore, when used as the D-amino acid control agent of the present invention, it is essential to use an amount that allows confirmation of an increase or decrease in the amount of D-amino acid at the site of action upon ingestion.
  • L-amino acids have various physiological activities different from those of D-amino acids
  • foods for which the standards (optical purity, amount), etc. of active ingredients that increase or decrease the amount of D-amino acids have not been set are not suitable for the composition of the present invention. not included in the range of
  • drugs or foods that can increase or decrease the amount of D-amino acids in cells, tissues, organs, body fluids, or media for culturing cells in a subject, regardless of the mechanism, are applicable to the present invention. It can optionally be used as a means of controlling the amount of D-amino acids obtained.
  • food means food in general, but in addition to general food including so-called health food, it also includes food with health claims such as food for specified health use and food with nutrient function claims. Supplements (supplements, dietary supplements), feeds, food additives and the like are also included in the food of the present invention.
  • Methods of administering drugs include local administration (cutaneous, inhalation, enema, eye drops, ear drops, nasal, intravaginal, etc.), enteral administration, parenteral administration (intravenous, transarterial, transdermal, intramuscular, etc.). enteral administration is preferred.
  • Enteral administration includes oral administration, tube administration, and enema administration.
  • Tube administration includes administration through a nasogastric tube, gastrostomy, or duodenal fistula.
  • Enema administration includes administration using suppositories and enemas.
  • the dosage form of the drug is not particularly limited, and may be liquid or solid, and can be produced according to the common technical knowledge of those skilled in the art.
  • the specific administration method is also not particularly limited, and administration can be suitably performed according to the common technical knowledge of those skilled in the art.
  • Targets to which the present invention can be applied include cultured cells, tissues, and organoids in any environment different from in vivo, and patients with cell proliferation, abnormal organ function, or organ atrophy due to disease or injury, or abnormal include patients with suspected Organ transplantation can be performed with the liver, pancreas, kidney, digestive tract, heart, eyeball, etc. In many cases, both the donor from whom the organ is removed and the recipient who receives the organ are transplanted before and after surgery. The present invention can be applied to such subjects because organ damage and functional deterioration accompanied by abnormal cell proliferation appear.
  • organ injury for example, the kidney is classified as type I (subcapsular injury), type II (superficial injury), type III (deep injury), etc.
  • Non-Patent Document 11 The present invention can be applied to any subject.
  • the donor loses one of two kidneys, i.e., total kidney size is approximately halved and kidney function is halved, but the size of the remaining kidney increases to compensate for the loss of function.
  • D- By increasing the amount of amino acids, regeneration of cells, tissues, and organs can be assisted, and glomerular filtration rate, which is one of renal functions, can be improved.
  • subjects to which the present invention can be applied may be subjects with kidney disease, kidney transplant donors and/or recipients, and/or dialysis patients, or patients undergoing renal replacement therapy.
  • animals in which abnormal cell proliferation is induced by genetic modification or drugs e.g., uninephrectomized mice, etc.
  • cultured cells/tissues e.g., cancer model cells/tissues, stem cells, differentiated cells/tissues, etc.
  • One embodiment of the present invention is that the amount of D-amino acids in a medium for culturing cells, and the increase or decrease in the amount of D-amino acids in target cells, tissues and/or body fluids affect cell proliferation. , by measuring the amount of D-amino acids thereof, it can be used as an index of the state of abnormal cell proliferation and the effect of treatment. For example, in the kidney, by monitoring the amount of D-amino acids in the body and the amount of glomerular filtration, it is possible to diagnose and evaluate cell proliferation and functional abnormalities, analyze the mechanism of action of drugs, screen effects and toxicity, and select treatment methods and drugs. , it is possible to assist in determining the dosage, period, etc. Since the amount of D-amino acids in body fluids is affected by other diseases, for the purpose of distinguishing them, the values corrected by renal function markers such as creatinine and other markers are analyzed. may be used for
  • One embodiment of the present invention is a method of modulating cell proliferation in a subject, comprising: A method comprising administering to a subject in need thereof an agent for controlling the amount of D-amino acids in vivo is provided.
  • one embodiment of the present invention is a method for modulating cell proliferation in vitro or ex vivo, comprising applying a D-amino acid amount controlling agent to a cell, biological tissue or organ and culturing it.
  • a D-amino acid amount controlling agent for example, in an in vitro or ex vivo environment, by culturing a cell, biological tissue, or organ using a buffer solution (e.g., medium) or physiological saline containing the above-mentioned D-amino acid amount regulator, Cell proliferation can be regulated.
  • the buffer that can be used in the present invention may be any known buffer that can be used for culturing, protecting or preserving cells, living tissue or organs.
  • one embodiment of the present invention provides the use of an agent for controlling the amount of D-amino acids for the manufacture of a pharmaceutical composition for modulating cell proliferation.
  • the method of regulating cell proliferation by controlling the amount of D-amino acids is extremely useful not only for efficient cell culture, but also for the prevention, treatment, and diagnosis of biological tissue and organ abnormalities.
  • TIG-1 cells isolated from fetal tissue were plated on a 96-well plate so that the number of viable cells per well was 1.5 ⁇ 10 4 cells.
  • the cells were cultured in a DMEM high glucose + 10% FBS medium overnight at 37°C in 5% CO 2 , the medium was removed, and the cells were washed with 150 ⁇ l of PBS. It was cultured in DMEM amino acid free medium + 0.5% dialyzed FBS at 37°C under 5% CO 2 for 18 hours.
  • L-serine L-Ser
  • D-serine D-Ser
  • Alanine (Ala), asparagine (Asn), glutamine (Gln), aspartic acid (Asp), glutamic acid (Glu), valine (Val), and lysine (Lys) were found to have a similar significant proliferative effect on DL bodies. rice field. D-methionine (D-Met) had no cell proliferation effect, but L-methionine (L-Met) had a significant cell proliferation effect (Fig. 1).
  • the level of D-serine in plasma was higher than in the vehicle group, and the kidney size (kidney weight per body weight) was increased (Fig. 5). This will serve as a model for cell proliferation/tissue regeneration and functional recovery of the remnant kidney of a donor in renal transplantation.
  • Sequencing was performed on an Illumina HiSeq2500 platform in 75 base single-ended mode and base calling using Illumina Casava 1.8.2 software.
  • the sequence read is Top Hat Ver. 2.0.13 to Bowtie2 Ver. 2.2.3 and SAMtools Ver. 0.1.19, and mapped to the mouse reference genome sequence (mm10), fragments per exon kilobase per million mapped fragments (FPKM) were obtained from Cufflinks Ver. 2.2.1 was used for calculation.
  • RNAseq data uses TargetMine (YA Chen, et al, PLoS One 6, e17844 (2011)), an integrated warehouse of human and mouse biological data from data sources such as Reactome and KEGG. Then, pathway analysis and gene ontology analysis were performed. Genes that were upregulated 2-fold change over the mean in D-serine treated mice compared to Vehicle treated mice were uploaded to TargetMine. Enrichment of Reactome pathways and gene ontology (GO) (M. Ashburner, et al, Nat Genet 25, 25-29 (2000)) was assessed by hypergeometric distributions and estimated p-values, and further by Benajmini and Hochberg Multiple-test correction was performed to suppress false positives using the method (W. S. Noble, et al., Nat Biotechnol 27, 1135-1137 (2009).). Heatmaps were drawn using R.
  • mice were perfused with saline, excised kidneys were sectioned, fixed with 4% paraformaldehyde, embedded in paraffin, and sections were stained with periodic acid Schiff (PAS). , or immunostained with anti-Ki67 antibody and Histofine Simple Stain Rat MAX PO (R) (414181F, Nichirei Biosciences) (T. Kimura, et al, Y. J Am Soc Nephrol 22, 902-913 (2011)). To count Ki67-positive cells per image, at least 10 fields (x200) were reviewed by a nephrologist blinded to the experimental conditions. For immunofluorescence, paraffin sections were incubated with Ki67 antibody and Alexa594-labeled secondary antibody.
  • PAPS periodic acid Schiff
  • FITC-binding lectin from Triticum vulgaris was used to stain proximal tubules and nuclei were counterstained with DAPI. Images were captured using a fluorescence microscope (Axio Observer) and a digital camera (AxioCam506 mono and AxioCamMRc, ZEISS, Oberkochen, Germany). All images were processed using ZEN 2 pro software (ZEISS) and Image J (NIH).
  • HK-2 CRL-2190, ATCC, Manassas, USA
  • RPTEC human primary renal proximal tubular cells
  • HeLa cells JCRB9004, JCRB Cell Bank, Japan, Japan
  • DMEM Dulbecco's Modified Eagle Medium
  • FCS 10% FCS (10270-106, Gibco, Carlsbad, USA).
  • p18-deficient MEFs p18 KO
  • Rev revertant
  • Rheb and p18 double-deficient MEFs Rheb and p18 double-deficient MEFs
  • cells were seeded in 96-well plates using medium. The next day, the medium was changed to 0.5% dialyzed FCS (26400-036, Gibco) and serine-free medium (amino acid-free DMEM [048-33575] supplemented with MEM essential amino acids [132-15641], glycine [073-00732], and GlutaMax ( L-alanyl-L-glutamine) [016-21841, Fujifilm Wako]), was added). After overnight incubation, D-serine and L-serine were added, respectively, and relative cell numbers were measured over time using a WST-8 kit (CK04, Dojindo Laboratories, Kumamoto, Japan). Experiments using MEFs used type I collagen-coated microplates (4860-010, AGC Techno Glass, Haibara, Japan).
  • Immunoblots were performed by lysing cells for 1 hour on ice with RIPA buffer (89901, Thermo Fisher) supplemented with protease inhibitor cocktail (4693132001, Roshe) with or without phosphatase inhibitors (4906837001, Roshe, Basel, Switzerland). and centrifuged at 15000G for 10 minutes. The supernatant was boiled in SDS-PAGE gel loading buffer for 3 minutes, separated by SDS-PAGE, transferred to a PVDF membrane, and subjected to Western blot analysis (T. Kimura, et al EMBO J 36, 42-60 (2017)). .
  • the group supplemented with D-serine promoted cell proliferation at a low concentration in human renal tubular cell lines and primary cells (Fig. 11).
  • Addition of D-serine enhanced Thr389 phosphorylation of S6K in HK-2 cells (FIG. 12).
  • D-serine addition also induced Ser235/236-phosphorylation of S6RP in the kidney of UNX mice (FIG. 13).
  • D-serine was shown to enhance the signal for activation of mTORC1.
  • the effect of D-serine on the mTOR pathway was also confirmed by the suppression of D-serine-induced cell proliferation in p18-deficient strains (Fig. 14) (R.
  • Example 7 D-serine administration and glomerular filtration rate
  • IL-3-dependent mouse pro-B-cell line Ba/F3 cells were seeded in 96-well plates using culture medium.
  • the medium is alanine-free medium (amino acid-free RPMI, MEM essential amino acids, GlutaMax (l-Alanyl-l-Glutamine), and 5% (v/v) dialyzed FCS (26400-036, Gibco) and 100 ng/10 ⁇ M D - Cells were cultured in the presence or absence of alanine for 48 hours, relative cell numbers were determined using WST-8 kit (CK04, Dojindo Laboratories, Kumamoto, Japan) and T-test was performed.
  • WST-8 kit CK04, Dojindo Laboratories, Kumamoto, Japan

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biomedical Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Urology & Nephrology (AREA)
  • Cell Biology (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
PCT/JP2022/020851 2021-05-19 2022-05-19 細胞増殖の調整のための組成物及び細胞増殖の調整方法 WO2022244846A1 (ja)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2023522721A JPWO2022244846A1 (de) 2021-05-19 2022-05-19

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2021084974 2021-05-19
JP2021-084974 2021-05-19

Publications (1)

Publication Number Publication Date
WO2022244846A1 true WO2022244846A1 (ja) 2022-11-24

Family

ID=84141625

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2022/020851 WO2022244846A1 (ja) 2021-05-19 2022-05-19 細胞増殖の調整のための組成物及び細胞増殖の調整方法

Country Status (2)

Country Link
JP (1) JPWO2022244846A1 (de)
WO (1) WO2022244846A1 (de)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2005514069A (ja) * 2002-01-17 2005-05-19 ビーエーエスエフ プラント サイエンス ゲーエムベーハー Dアミノ酸を用いる選択的植物成長
WO2011087091A1 (ja) * 2010-01-15 2011-07-21 協和発酵キリン株式会社 抗システムascアミノ酸トランスポーター2(asct2)抗体
JP2015027265A (ja) * 2013-07-30 2015-02-12 ダイソー株式会社 細胞培養用培地
JP2015516456A (ja) * 2012-05-14 2015-06-11 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 糸球体上皮細胞関連障害及び/又はネフローゼ症候群の治療に用いるdpp−4阻害薬としてのキサンチン誘導体
JP2016510795A (ja) * 2013-03-15 2016-04-11 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 心臓保護及び腎臓保護の抗糖尿病治療におけるリナグリプチンの使用
WO2019235592A1 (ja) * 2018-06-07 2019-12-12 国立大学法人金沢大学 腎障害の予防又は治療用の医薬組成物
JP2019214558A (ja) * 2018-06-07 2019-12-19 国立大学法人金沢大学 腎障害の予防又は治療用の医薬組成物
WO2020218737A1 (ko) * 2019-04-22 2020-10-29 씨제이제일제당 (주) L-쓰레오닌 생산능이 강화된 미생물 및 이를 이용한 쓰레오닌 생산방법
WO2021132691A1 (ja) * 2019-12-27 2021-07-01 Kagami株式会社 D-セリン輸送調節剤及びそのスクリーニング方法、並びにd-セリン輸送体タンパク質のスクリーニング方法

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2005514069A (ja) * 2002-01-17 2005-05-19 ビーエーエスエフ プラント サイエンス ゲーエムベーハー Dアミノ酸を用いる選択的植物成長
WO2011087091A1 (ja) * 2010-01-15 2011-07-21 協和発酵キリン株式会社 抗システムascアミノ酸トランスポーター2(asct2)抗体
JP2015516456A (ja) * 2012-05-14 2015-06-11 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 糸球体上皮細胞関連障害及び/又はネフローゼ症候群の治療に用いるdpp−4阻害薬としてのキサンチン誘導体
JP2016510795A (ja) * 2013-03-15 2016-04-11 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 心臓保護及び腎臓保護の抗糖尿病治療におけるリナグリプチンの使用
JP2015027265A (ja) * 2013-07-30 2015-02-12 ダイソー株式会社 細胞培養用培地
WO2019235592A1 (ja) * 2018-06-07 2019-12-12 国立大学法人金沢大学 腎障害の予防又は治療用の医薬組成物
JP2019214558A (ja) * 2018-06-07 2019-12-19 国立大学法人金沢大学 腎障害の予防又は治療用の医薬組成物
WO2020218737A1 (ko) * 2019-04-22 2020-10-29 씨제이제일제당 (주) L-쓰레오닌 생산능이 강화된 미생물 및 이를 이용한 쓰레오닌 생산방법
WO2021132691A1 (ja) * 2019-12-27 2021-07-01 Kagami株式会社 D-セリン輸送調節剤及びそのスクリーニング方法、並びにd-セリン輸送体タンパク質のスクリーニング方法

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
SATO TSUTOMU, YAMOCHI TADANORI, YAMOCHI TOSHIKO, AYTAC UGUR, OHNUMA KEI, MCKEE KATHRYN S., MORIMOTO CHIKAO, DANG NAM H.: "CD26 Regulates p38 Mitogen-Activated Protein Kinase–Dependent Phosphorylation of Integrin β 1 , Adhesion to Extracellular Matrix, and Tumorigenicity of T-Anaplastic Large Cell Lymphoma Karpas 299", CANCER RESEARCH, vol. 65, no. 15, 1 August 2005 (2005-08-01), US, pages 6950 - 6956, XP055873919, ISSN: 0008-5472, DOI: 10.1158/0008-5472.CAN-05-0647 *
THIELITZ ANJA, ANSORGE SIEGFRIED, BANK UTE, TAGER MICHAEL, WRENGER SABINE, GOLLNICK HARALD, REINHOLD DIRK: "The ectopeptidases dipeptidyl peptidase IV (DP IV) and aminopeptidase N (APN) and their related enzymes as possible targets in the treatment of skin diseases", FRONTIERS IN BIOSCIENCE, vol. 13, no. 13, 1 January 2008 (2008-01-01), US , pages 2364 - 2375, XP093005632, ISSN: 1093-9946, DOI: 10.2741/2850 *

Also Published As

Publication number Publication date
JPWO2022244846A1 (de) 2022-11-24

Similar Documents

Publication Publication Date Title
Hou et al. Ectosomal PKM2 promotes HCC by inducing macrophage differentiation and remodeling the tumor microenvironment
Hua et al. TRIB3 interacts with β-catenin and TCF4 to increase stem cell features of colorectal cancer stem cells and tumorigenesis
Zhang et al. MiR-29b inhibits collagen maturation in hepatic stellate cells through down-regulating the expression of HSP47 and lysyl oxidase
Chao et al. Y-box binding protein-1 promotes hepatocellular carcinoma-initiating cell progression and tumorigenesis via Wnt/β-catenin pathway
US20190231761A1 (en) Compositions and methods for targeting fructose enzymes and transporters for the treatment of cancer
Behmoaras et al. Macrophage epoxygenase determines a profibrotic transcriptome signature
US20210361694A1 (en) Method for preventing or treating cancer using syt11 inhibitor
Pan et al. METTL3 drives NAFLD-related hepatocellular carcinoma and is a therapeutic target for boosting immunotherapy
Ma et al. Phillygenin inhibited M1 macrophage polarization and reduced hepatic stellate cell activation by inhibiting macrophage exosomal miR-125b-5p
Geng et al. Targeting miR-9 in glioma stem cell-derived extracellular vesicles: a novel diagnostic and therapeutic biomarker
Xiao et al. Therapeutic inhibition of phospholipase D1 suppresses hepatocellular carcinoma
EP3597739A1 (de) Zusammensetzung zur regulierung der krebszellenteilung oder -differenzierung mit setdb1 oder einem setdb1-inhibitor
US20180318448A1 (en) Method for determining presence or absence of suffering from malignant lymphoma or leukemia, and agent for treatment and/or prevention of leukemia
Li et al. Novel long non‐coding RNA CYB561‐5 promotes aerobic glycolysis and tumorigenesis by interacting with basigin in non‐small cell lung cancer
WO2022244846A1 (ja) 細胞増殖の調整のための組成物及び細胞増殖の調整方法
Dai et al. miR‑122/SENP1 axis confers stemness and chemoresistance to liver cancer through Wnt/β‑catenin signaling
Chen et al. Damage of uremic myocardium by p-cresyl sulfate and the ameliorative effect of Klotho by regulating SIRT6 ubiquitination
Xia et al. MAT2A inhibits the ferroptosis in osteosarcoma progression regulated by miR-26b-5p
van Zyl et al. Isoginkgetin leads to decreased protein synthesis and activates an ATF4-dependent transcriptional response
CN103800919B (zh) Tuft1在制备肝癌诊断和治疗制剂中的应用
Chen et al. Hsa_circ_0005050 regulated the progression of oral squamous cell carcinoma via miR-487a-3p/CHSY1 axis
Zhu et al. A Zeb1/MtCK1 metabolic axis controls osteoclast activation and skeletal remodeling
EP3406253A1 (de) Inhibitoren und antagonisten von humanem pycr1
WO2022244842A1 (ja) 糖質代謝の調整のための組成物及び、対象の糖質代謝の状態を評価する方法
US20240238236A1 (en) Composition for regulating carbohydrate metabolism, and method for assessing carbohydrate metabolism conditions of subject

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: 22804755

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2023522721

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22804755

Country of ref document: EP

Kind code of ref document: A1