CN111920955A - Application of Grb10 as key negative regulator of beta cell dysfunction - Google Patents

Application of Grb10 as key negative regulator of beta cell dysfunction Download PDF

Info

Publication number
CN111920955A
CN111920955A CN202011025653.7A CN202011025653A CN111920955A CN 111920955 A CN111920955 A CN 111920955A CN 202011025653 A CN202011025653 A CN 202011025653A CN 111920955 A CN111920955 A CN 111920955A
Authority
CN
China
Prior art keywords
grb10
beta cell
leu
mice
ser
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202011025653.7A
Other languages
Chinese (zh)
Inventor
张晶晶
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Second Xiangya Hospital of Central South University
Original Assignee
Second Xiangya Hospital of Central South University
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 Second Xiangya Hospital of Central South University filed Critical Second Xiangya Hospital of Central South University
Publication of CN111920955A publication Critical patent/CN111920955A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/04Endocrine or metabolic disorders
    • G01N2800/042Disorders of carbohydrate metabolism, e.g. diabetes, glucose metabolism

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Diabetes (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Genetics & Genomics (AREA)
  • Physics & Mathematics (AREA)
  • Urology & Nephrology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Obesity (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)

Abstract

The invention relates to the field of biological medicines, in particular to application of Grb10 as a key negative regulator of pancreatic islet beta cell dysfunction. The invention studies the physiological function of Grb10 in beta cells. The experimental results show that Grb10 is highly expressed in human and mouse pancreas and islets, and is highly expressed in islets of diabetic mouse model and in the islets of the elderly. In addition, β cell-specific knockout of Grb10 increased β cell mass and β cell function in High Fat Diet (HFD) mice by promoting β cell maturation and inhibiting β cell dedifferentiation. The invention highlights the important role of the interaction of Grb10-mTORC1 in regulating the dedifferentiation of beta cells and provides new insights for the regulation mechanism of the characteristics and functions of the beta cells.

Description

Application of Grb10 as key negative regulator of beta cell dysfunction
The present application claims priority from chinese patent application filed on 09/06/2020, having application number 202010517856.1 and entitled "use of Grb10 as a key negative regulator of beta cell dysfunction", the entire contents of which are incorporated herein by reference.
Technical Field
The invention relates to the field of biological medicines, in particular to application of Grb10 as a key negative regulator of beta cell dysfunction.
Background
A reduction in the number of beta cells and/or an impaired function of beta cells is one of the mechanisms by which type 1 diabetes (T1D) and type 2 diabetes (T2D) occur. However, the underlying mechanism of diabetes, beta cell depletion, remains to be fully established.
Studies have shown that mature and differentiated beta cells are not in an immortalized state, which can change with environmental changes. There is increasing evidence that islet beta cell dedifferentiation plays a key role in the loss of diabetic beta cells. Dedifferentiated beta cells lose their properties and insulin secretion function, with a concomitant down-regulation of beta cell-specific transcription factors and up-regulation of endocrine precursor surface markers. The de-differentiation of beta cells into neuronal 3(Ngn3) -like progenitor cells and transdifferentiation into other endocrine cells is associated with the development of human T2D. However, the molecular mechanisms that regulate the characteristics of beta cells remain unknown. Studies have shown that glucose transporter 2(Glut2) and the pancreatic duodenal homeobox 1(PDX1) are required for maintaining beta cell properties. However, under normal and abnormal metabolic conditions, it is currently unclear whether other key molecules exist to regulate the identity of beta cells.
Growth factor receptor-bound protein 10(Grb 10) is a protein that negatively regulates insulin and mTORC1 signaling in insulin target cells (PH) and Src homology 2 (SH 2) domain proteins. Grb10 has been identified as a diabetes susceptibility gene, but its tissue-specific and mechanism of action in diabetes remains elusive. Our previous studies showed that pancreatic Grb10 knockout mice increase beta cell mass by promoting beta cell proliferation and protecting beta cells from streptozotocin-induced apoptosis. However, the exact mechanism by which Grb10 regulates islet beta cell homeostasis and function is not clear.
mTORC is a serine/threonine kinase that is critical to cell growth and organ development. Deletion of the beta cell-specific mTORC1 results in beta cell failure due to defects in cell proliferation, apoptosis, and insulin secretion. Consistent with these findings, intraperitoneal injection of rapamycin significantly reduced the number of β cells in mice. On the other hand, activation of the mTORC1 signaling pathway increases beta cell mass. Although these findings suggest that the mTORC1 signaling pathway plays a critical role in regulating beta cell mass, the specific mechanism by which mTORC1 signaling pathway regulates beta cell mass and function remains unclear.
Disclosure of Invention
In view of this, the present invention provides the use of Grb10 as a key negative regulator of beta cell dysfunction.
In order to achieve the above object, the present invention provides the following technical solutions:
the invention provides an application of Grb10 as a key negative regulator of beta cell dysfunction.
In some embodiments of the invention, the beta cell dysfunction comprises beta cell dedifferentiation, loss of beta cell properties, beta cell exhaustion.
The invention also provides application of Grb10 inhibition in preparation of a medicine for preventing and/or treating beta cell dysfunction.
The invention also provides application of inhibition of Grb10 in preparation of drugs for improving glucose tolerance of animals.
The invention also provides application of a blocking agent of Grb10 inhibition or Grb10 regulation signal pathway in preparation of a drug for improving beta cell function and preventing and/or treating diabetes.
In some particular embodiments of the invention, said inhibition of Grb10 comprises a function of reducing the expression of Grb10, knockout and/or down-regulation of the Grb10 gene.
In some embodiments of the invention, said inhibition of Grb10 promotes beta cell proliferation.
In some embodiments of the invention, said inhibition of Grb10 inhibits beta cell dedifferentiation.
In some embodiments of the invention, the signaling pathway regulated by Grb10 enhances beta cell mass and improves beta cell insulin secretion function.
In some embodiments of the invention, the signaling pathway regulated by Grb10 is the mTORC1 signaling pathway.
The invention studies the physiological function of Grb10 in beta cells. The experimental results show that Grb10 is highly expressed in human and mouse pancreas and islets, and is highly expressed in islets of diabetic mouse model and in the islets of the elderly. In addition, β cell-specific knockout of Grb10 increased β cell mass and β cell function in High Fat Diet (HFD) mice by promoting β cell maturation and inhibiting β cell dedifferentiation. Mechanistically, beta cell-specific gene knockout of Grb10 upregulates the mTORC1 signaling pathway, resulting in increased expression of transcription factors that promote beta cell maturation and decreased expression of transcription factors that promote endocrine pancreatic development. The invention highlights the important role of the interaction of Grb10-mTORC1 in regulating the dedifferentiation of beta cells and provides new insights for the regulation mechanism of the characteristics and functions of the beta cells.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the description of the embodiments or the prior art will be briefly described below.
FIG. 1 shows the experimental results of Effect example 1; wherein, (A) qRT-PCR detects the gene expression level of Grb10 in human muscle, brain and pancreatic islets; (B) WB assay Grb10 protein expression levels in human brain, liver, muscle, fat, heart, kidney, spleen and pancreas; tublin is used as an internal reference; (C) human pancreatic tissue sections were examined for Grb10 and immunofluorescence of insulin; (D) WB was used to detect Grb10 protein expression levels in islets of C57BL/6J mice fed Normally (ND) and High Fat (HFD) for 16 weeks; (E) WB detecting the protein expression level of Grb10 in the islets of 3-month-old control mice and ob/ob mice; (F) WB detecting protein expression level of Grb10 in islets of male db/+ mice and diabetic db/db mice of 3 months old; D. the beta-Actin in the F picture is used as an internal reference; (G) detecting the gene expression levels of Grb10, Sox9, Ngn3 and PDX1 in MIN6 cells cultured in medium containing 5.5mM Normal Glucose (NG) or 40mM High Glucose (HG); (H) detecting Grb10 and Ngn3 protein levels in MIN6 cells cultured for 2 days under 5.5mM (NG) or 40mM (HG) glucose conditions; (I) protein expression levels of Grb10 in human islets younger than 18 years of age and human islets older than 18 years of age; beta-Actin is used as an internal reference; data are expressed as mean ± SEM; p < 0.05;
FIG. 2 shows the experimental results of Effect example 2; wherein (a) immunofluorescence detects fluorescence of 8-week old male loxp (wt) mice and Grb10KO (KO) mouse pancreas sections Grb10 (red) and insulin (as a beta cell marker) (green); marking and quantifying the pancreatic area of each slice by a brightness extraction method; (B, C) high-fat fed 16-week male control mice (WT) and Grb10KO (KO) mice tested for beta cell to islet ratio (B) and alpha cell to islet ratio (C); (D, E) high-fat feeding of 16-week male control mice (WT) and Grb10KO (KO) mice to measure the average beta cell mass (D) and the average alpha cell mass (E); (F) HFD was fed to 16 weeks of loxP control mice (WT; n-8), GTT of control mice (Ins 2-cre; n-6) and Grb10KO (KO; n-8) to measure glucose tolerance; blood glucose was measured from the tail vein at 0, 15, 30, 60 and 120 minutes after intraperitoneal injection of glucose (2g/kg body weight); p compared to loxP control group (WT).)<0.05,**P<0.01,***P<0.001; compared with the cre mouse, the mouse has the advantages that,#P<0.05; (G) the ITT of control mice (Ins 2-cre; n-6) and Grb10KO (KO; n-8) from the control group, which were fed with HFD for 16 weeks (WT; n-8), examined insulin tolerance; (H) the hyperglycosylation clamp measures the glucose infusion rate of normally fed control and Grb10KO (KO) mice (n-5/group); (I) control mice and Grb10KO (KO) mice fed normally during the hyperglycosylation phase (n-5/group) were tested for serum insulin levels; experimental data represent mean ± SEM; p<0.05;**P<0.01;***P<0.001 (one-way repeated measures analysis of variance);
FIG. 3 shows the experimental results of Effect example 3; wherein, (A) qRT-PCR analyzes the gene expression of islets Grb10, Ngn3, Glut2, PDX1 and Ins1 of control group mice fed with high fat for 16 weeks and Grb10KO (KO) mice; (B) WB analysis protein expression levels of islets Glut2, PDX1, Ngn3 of control mice and Grb10KO (KO) mice fed with high fat for 16 weeks, with β -Actin as an internal control; (C) immunofluorescence assay control mice and Grb10KO (KO) mice insulin and Glut2 fluorescence of pancreatic sections under ND and HFD feeding conditions; (D) GFPMIN6 cells and Grb10 OE MIN6 cells constructed on MIN6 cells using adenovirus Ad-GFP (GFP) or Ad-Grb10(Grb10 OE) and detecting the gene expression levels of Glut2, PDX1, NKX6-1, Ins1 and Ins 2; (E) WB detected protein expression levels of Glut2 and PDX1 in GFP MIN6 cells and Grb10 OEMIN 6 cells; beta-Actin is used as an internal reference; the relative expression of proteins by Glut2 and PDX1 in control GFPMIN6 cells and Grb10 OE MIN6 cells is shown in the histogram; data represent mean ± SEM; p < 0.05; p < 0.01; p < 0.001;
FIG. 4 shows the experimental results of effect example 4; wherein, (A) qRT-PCR analysis of Grb10, Ngn3 and Sox9 gene expression in islets of Grb10KO (KO) mice and control mice fed with HFD for 16 weeks; (B) WB analysis of protein expression of Ngn3 and Sox9 in islets of control mice fed with HFD for 16 weeks and Grb10KO (KO) mice; beta-Actin is used as an internal reference; (C, D) after Grb10 is over-expressed in MIN6 cells, (C) the gene expression of Grb10, Ngn3, Sox9 in GFP MIN6 cells and Grb10 OEMIN 6 cells is detected by qRT-PCR; (D) WB detects the protein expression of Ngn3 and Sox9 in GFPMIN6 cells and Grb10 OEMIN 6 cells; beta-Actin is used as an internal reference; (E) immunofluorescence insulin and Sox9 fluorescence were detected from pancreas sections of control mice and Grb10KO (KO) mice under HFD feeding conditions, and data are presented as mean ± SEM; p < 0.05; p < 0.01; p < 0.001;
FIG. 5 shows the experimental results of effect example 5; wherein (A) after the control group mice Grb10KO (KO) fed by HFD respectively treat a control solvent and rapamycin, the WB is used for checking the protein expression of Grb10, Glut2, PDX1, Ngn3, S6, 4EBP1 and S6 phosphorylation at Ser235 site and 4EBP1 phosphorylation at Thr37/46 site in the islets of four groups of mice, and beta-Actin is used as an internal reference; (B, C) INS-1 cells were treated with siRNA (NC) and Grb10 specific siRNA (siGrb10), respectively, for 48 hours; (B) qRT-PCR detection of gene expression conditions of Grb10, Ngn3 and Sox9 is carried out 24 hours after the cells are infected with a control shRNA or sh-raptor lentivirus; (C) cells are treated by rapamycin for 24 hours and then are cracked, and WB detects the phosphorylation of Glut2, PDX1, S6 and S6 kinase at Thr389 site, 4EBP1 and 4EBP1 at Thr37/46 site and the expression of Grb10 protein; beta-Actin is used as an internal reference; (D) the glucose tolerance of mice is detected by using GTT after HFD-fed control group mice and Grb10KO (KO) mice respectively treat a control solvent and rapamycin; (WT; n-10), Grb10KO (KO; n-6), (WT + Rap; n-10) and (KO + Rap; n-7); (E) beta cell mass was measured in four groups of mice after HFD-fed control mice and Grb10KO mice treated with control solvent and rapamycin, respectively; (WT; n-7), Grb10KO (KO; n-6), (WT + Rap; n-8), and (KO + Rap; n-6); (D, E) data represent mean ± SEM; (D) p <0.05, P <0.01, P < 0.001; (F) immunofluorescence was measured for fluorescence expression of insulin and Glut2 in pancreas sections of four groups of mice treated with control solvent and rapamycin, respectively, in HFD-fed control mice and Grb10KO (KO) mice; (G) mode diagram: the mechanism by which Grb10 regulates beta cell dedifferentiation suggests: the long-term HFD feeding can induce the dedifferentiation of beta cells, is accompanied by the down-regulation of the expression of beta cell-specific genes and proteins and up-regulates the expression of genes and proteins related to secretion precursors, Grb10 negatively regulates mTORC1 signal pathways, and after the beta cell-specific knockout of Grb10, mTORC1 activates the signal pathways, inhibits the dedifferentiation of the beta cells and promotes the proliferation of the beta cells.
Detailed Description
The invention discloses application of Grb10 as a key negative regulator of beta cell dysfunction, and a person skilled in the art can appropriately modify process parameters for realization by referring to the content. It is expressly intended that all such similar substitutes and modifications which would be obvious to one skilled in the art are deemed to be included in the invention. While the methods and applications of this invention have been described in terms of preferred embodiments, it will be apparent to those of ordinary skill in the art that variations and modifications in the methods and applications described herein, as well as other suitable variations and combinations, may be made to implement and use the techniques of this invention without departing from the spirit and scope of the invention.
In this study, we explored the mechanism by which Grb10 regulates beta cell mass. We found that β cell Grb10 gene knock-out enhances the ability of pancreatic islet β cells to stimulate insulin secretion (GSIS) by glucose, which is associated with a sustained state of cell maturation under long-term HFD feeding conditions. Mechanistically, knockout of Grb10 results in activation of the mTORC1 signaling pathway, inhibition of beta cell dedifferentiation, maintenance of beta cell identity and increase of beta cell mass. Our studies indicate a key role of the Grb10-mTORC1 axis in regulating beta cell dedifferentiation and provide clues for the development of potential new targets for the effective treatment of beta cell failure, the major cause of diabetes.
Maintaining the mature nature of islet beta cells is critical to maintaining beta cell mass and glucose homeostasis in the body. Unlike other terminally differentiated cells, beta cells may lose their properties and dedifferentiate under certain pathophysiological conditions, leading to beta cell failure and consequent diabetes. However, the mechanisms that regulate the properties of beta cells remain to be further elucidated. In this study, we found that specific deletion of mouse Grb10 in beta cells inhibited HFD-induced expression of precursor genes such as Ngn3 and Sox9 (fig. 4). The lack of Grb10 in β cells also increased the expression of genes characteristic of the mature differentiation of β cells, such as insulin, PDX1, Glut2, etc. (fig. 3). These results indicate that inhibition of beta cell dedifferentiation contributes to an increase in beta cell mass and insulin content in HFD fed Grb10ko (ko) mice.
Several signaling pathways, such as the Hedgehog, Wnt, Notch and NF-. kappa.b signaling pathways, have been found to regulate beta cell dedifferentiation. We found that the beta cell-specific knockout of Grb10 had no significant effect on activation of Wnt, Notch and NF- κ B signaling pathways. On the other hand, Grb10 gene knockout greatly increased phosphorylation of islet beta cell S6 at Ser235 site and 4EBP1 at thr37/46 site, suggesting activation of mTORC1 signaling pathway. Inhibition of the mTORC1 signaling pathway by rapamycin or Raptor knockdown increased the expression of the endocrine precursor marker Ngn3 (fig. 5A-B). Consistent with this result, intraperitoneal injection of rapamycin also significantly reduced β -cell mass (fig. 5E) and glucose intolerance in Grb10ko (ko) mice. These results indicate that activation of the mTORC1 signaling pathway plays an important role in inhibiting the differentiation of beta cells to a precursor cell-like state.
Maintenance of beta cell mass is regulated by many complex processes, and insufficient beta cell mass can lead to type 1 diabetes and type 2 diabetes. While islet transplantation can complement beta cell mass, this approach is limited by the lack of donors. Therefore, the development of effective therapeutic methods for promoting beta cell proliferation is crucial for beta cell transplantation. Unfortunately, although human cells can be induced to proliferate in vitro, most proliferating beta cells lose the property of expressing and secreting insulin. Lineage tracking indicates that beta cells undergo rapid loss of beta cell phenotype when they promote human cell proliferation in vitro. Activation of the Hedgehog signaling pathway promotes proliferation of beta cells, but also results in de-differentiation of beta cells. The research shows that the knockout of the Grb10 gene in the beta cell can not only promote the proliferation of the beta cell, but also obviously inhibit the dedifferentiation of the beta cell, and the fact that the targeting Grb 10-regulated beta intracellular signal pathway is probably an effective strategy for enhancing the quality of the beta cell and improving the insulin secretion function of the beta cell is suggested.
Taken together, we determined that Grb10 is a key regulator of beta cell dedifferentiation. Furthermore, we found that inhibition of Grb10 expression in beta cells can inhibit the dedifferentiation of beta cells by activating mTORC1 signaling pathway, thereby maintaining beta cell characteristics and improving cell function. Increasing beta cell dedifferentiation and apoptosis and decreasing cell proliferation are key factors in the pathogenesis of T1D and T2D. Understanding the regulatory mechanisms of cell dedifferentiation and determining key regulatory factors in the dedifferentiation process of beta cells can provide valuable information for the formulation of new strategies for T1D and T2D treatment.
The gene name is: grb10
The species are as follows: mouse
ID:14783
Amino acid sequence: as shown in SEQ ID No. 1.
Figure BDA0002702042560000071
Figure BDA0002702042560000081
The gene name is: grb10
The species are as follows: human being
ID:2887
Amino acid sequence: as shown in SEQ ID No. 2.
Figure BDA0002702042560000082
Figure BDA0002702042560000091
The raw materials and reagents used in the application of the Grb10 provided by the invention as the key negative regulator of the beta cell dysfunction are all available in the market.
The invention is further illustrated by the following examples:
example 1 human study
Islets from 13 human organ donors were used in this study, including 6 minors (under 18 years) and 7 adults (over 18 years). Islets less than 18 years of age were compared to Grb10 expression from adult donors (older than 18 years of age). Human pancreas was obtained from dead non-diabetic donors by the urinary surgery organ/liver transplant department of the Hunan Yabi Hospital, southern China university.
Human islets were isolated by digestion with collagenase P, the adipose tissue surrounding the pancreas, lymph nodes, blood vessels and fascia were carefully cleaned and the human pancreas was trimmed, the pancreatic canula was inflated with a 5mL syringe, and Hanks Balanced Salt Solution (HBSS) containing 1mg/mL of collagenase P was injected through the pancreatic duct in an amount equivalent to twice the weight of the pancreas. The pancreas was removed and placed in collagenase P solution and digested at 37 ℃ for about 20-30 minutes, and the tubes were placed in an ice bath (4 ℃) to inactivate collagenase P and preserve the islets. The digestion was stopped by adding Hanks buffer, then the pancreas was washed 3 times with RPMI-1640 medium, human islet medium and dithizone dye were added and islets were selected under a microscope for isolation. The ethical committee of xiang yadi hospital, central university, approved the use of isolated human islets (protocol MSRC2016 LF).
For WB and qRT-PCR experiments, we used human brain, liver, muscle, fat, heart, kidney, spleen and pancreas from urological organ/liver transplantations in yaja, xiang, university, central southern china. The samples obtained were snap frozen and stored at-80 ℃ until use. The ethical committee of xiang yadi hospital, central university, approved the use of isolated human islets (protocol MSRC2016 LF).
Example 2 animal models
Beta cell specific Grb10 knockout mice Grb10KO (KO) were generated by breeding homozygous Grb10 female (loxP) mice on a C57BL/6J background with female homozygous Ins2-Cre mice on a C57BL/6J background. Ins2-Cre is expressed in pancreatic beta cells, but not in hypothalamic neurons. loxP littermates and Ins2-Cre littermates were used as controls (WT). The Ins2-Cre mice were provided by octopine (university of Tianjin medical science). All mice were housed in specific pathogen free facilities (SPF) at the animal care center of yaja, xiang, university, central south, with a 12 hour bright-dark cycle. All procedures for animal use were performed according to the Hunan Yay second Hospital animal Care and the Committee for animal Care and use in the Central and south university.
Example 3 body weight, food intake and body composition
Mice were fed either a plain diet (ND) containing 19% protein, 5% fat and 5% fiber (Hunan silake jingda laboratory animal Co Ltd) or a High Fat Drink (HFD) containing 20% protein; 60% fat; 20% carbohydrate (D12492, research Diets Inc.) for 16 weeks. Mice body weight and food intake were monitored weekly at the same time point. Mice were analyzed for lean mass and fat mass by MQ Minispec 7.5HZ live mouse Analyzer (MinispecLF 50; BRUKER Optik GmbH; Germany).
EXAMPLE 4 high sugar tongs
Awake, normally-fed 4-month-old loxP wild-type littermates (WT) and male Grb10KO (KO) mice were fasted overnight and anesthetized with ketamine (IP) (100mg/kg body weight) and xylazine (10mg/kg body weight) by intraperitoneal injection, 4-5 days prior to the clamp experiment, an indwelling catheter was inserted into the right internal jugular vein of the mice, the mice were housed in individual cages, and post-operative recovery and weight gain were monitored. After an overnight fast, a2 hour high glucose clamp experiment was performed in conscious mice with 10% glucose infusion at a variable rate to raise and maintain plasma glucose concentrations at 400mg/dL at 350-. Blood samples (20 μ L) were collected every 5 to 10 minutes to measure plasma glucose and insulin levels.
Example 5 mouse islet isolation and insulin particle count
Mouse islets were isolated by collagenase P digestion. An overnight fasted loxP (WT) control littermate and male Grb10KO (KO) mice were anesthetized by intraperitoneal injection of tribromoethanol (1mL/40g body weight), mice were pancreas-distended with 3mL of a 1mg/mL solution of collagenase P (Sigma Chemical of St. Louis, Mo.; collagenase P was added to Hank's buffered saline (HBSS)), the pancreas was removed and digested at 37 ℃ for about 12 minutes, then 30mL of HBSS was added to stop digestion, and finally washed 3 times with 20mL of RPMI-1640 medium, islet medium and dithizone dye were added and islets were isolated under a microscope.
For insulin particle counting, islets were isolated and fixed using 1% glutaraldehyde, embedded in epon, and tissue blocks (0.1-1.5 mm)3) In the presence of 1% OsO40.1mol/L sodium cocoate buffer (pH 7.4) for 1 hour at room temperature, washed in a cocoate buffer, then washed in distilled water, and then subjected to a 2% uranyl acetate double distilled water treatment for 1 hour. The sections were dehydrated in gradient alcohol and then placed in 100% propylene oxide. The blocks were incubated overnight in propylene oxide-propylene oxide (1: 1) at room temperature, then changed to 100% ethylene oxide and polymerized overnight. Sections (90nm) were cut on a Reichert-Jung UltracutE microtome, collected on a slotted grid, and stained with 2% uranyl acetate and lead citrate. Pictures were taken by Jeol1230 transmission electron microscope (Jeol, peabody, massachusetts). Three different empty particles (EG), immature particles (IG) and mature secretory particles (MSG) were counted accordingly.
Example 6 islet proliferation assay and TUNEL assay
Mouse pancreatic sections were analyzed by double staining with anti-ki 67 (Abcam; 16667) and anti-insulin (Sigma; I2018) antibodies. Insulin positive beta cells were counted and the percentage of total insulin positive nuclei detected for loxp (wt) control mice and Grb10KO (KO) mice ki67 and insulin double positive nuclei. The percentage of ki67 and the ratio of insulin double positive nuclei to total insulin positive nuclei were determined as the proliferation rate of loxp (wt) control mice and Grb10KO (KO) mice. The total ki67 positive cell number was quantified by using an anaerotactic method to ensure that the same ki67 labeled cells were not counted twice on adjacent sections and that the pancreatic section area remained consistent for each animal.
Male loxp (wt) control and Grb10KO (KO) mouse pancreas sections fed with HFD for 16 weeks were labeled with in situ cell death detection kit and insulin antibody and HFD-induced apoptosis was determined by immunofluorescence using an olympus inverted microscope (IX71) and captured with a Sport II digital camera. Six mice per group were used for (TUNEL) analysis (Beyotime C1090). More than 2500 insulin positive cells were counted per mouse. Apoptosis was calculated from TUNEL and insulin double positive nuclei to total insulin positive nuclei.
EXAMPLE 7 administration of rapamycin
Male loxP (WT) control and Grb10KO (KO) mice were fed HFD for 16 weeks every 3 days with an intraperitoneal injection of 5mg/kg rapamycin or control group solvent for 12 days. Rapamycin was dissolved in 100% ethanol at a concentration of 5mg/mL, diluted to 0.5mg/mL in a carrier solution of 5% Tween-80 and 5% PEG-400 in PBS, and then used after filtration.
Example 8 RNA interference in INS-1 cells
Cells were cultured in antibiotic-free medium for 24 hours and then transfected with siRNA targeting Grb10 (Ribobio; siG161129043512) or scrambled siRNA (Ribobio) at a final concentration of 25nM for 24 hours. Lipofectamine was used according to the manufacturer's instructionsTM3000 (ThermoFisher; L3000015) for transfection assay. After overnight transfection, the medium was replaced with conventional medium.
Example 9 Generation of adenovirus and Adenoviral infection
Adenovirus encoding GFP and Grb10 adenovirus were generated by using pAdEasy system. MIN6 cells infected adenovirus at a multiplicity of infection of 30. The efficiency of adenovirus infection was assessed by fluorescent GFP expression using a fluorescence microscope 24 hours post infection.
Example 10 real-time PCR
Total RNA was isolated from cells using Trizol (life technology) according to the manufacturer's instructions. The RNA concentration was determined using a UV-Vis spectrophotometer Q5000. Quantitative PCR reactions were performed using SYBR mixtures (Roche) and quantified using an applied biosystems 7900HT sequence detection system. Duplicate runs of each sample were performed and β -Actin was used as an internal reference to determine relative expression levels.
Example 11 GTT, ITT, Immunofluorescence (IF).
The Glucose Tolerance Test (GTT) was performed by injecting glucose (2g/kg body weight i.p.) into overnight fasted mice. Blood was withdrawn from the tail vein 0, 15, 30, 60 and 120 minutes after the glucose injection, and the blood glucose level of the mice was measured with a glucometer (one touch; Bionime Corp.).
Insulin resistance test (ITT) was performed by injection of human insulin (0.75U/kg body weight i.p.) into 4 hour fasted mice. At 0, 15, 30, 60 and 90 minutes after insulin injection, tail vein bleeds were initiated and serum insulin levels were determined using an insulin hypersensitivity enzyme immunoassay (Alpco Diagnostics, Slemm, NH).
Immunofluorescence (IF) experiments were performed by taking the pancreas of loxp (wt) control mice and Grb10KO (KO) mice, fixing overnight (DING GUO) with 4% paraformaldehyde, then dehydrating with 30% sucrose, embedding with o.c.t., and cutting 8 μm sections of pancreas using frozen sections for use. The sections were fixed, blocked, permeabilized and stained with specific antibodies-Grb 10 (homemade), 1: 50; insulin (Sigma I2018)1: 500; glut2(Millipore company 07-1402-I)1: 1000; ki67(abcam 16667)1: 500; sox9(Milipore AB5535)1:500, cell nuclei were identified using DAPI staining. The LSM 780 laser scanning confocal microscope multi-tracking setup detects the fluorescence signal using the corresponding fluorescent secondary antibody. Each channel uses the same pinhole diameter.
Example 12 statistics
The summary data represent mean ± SEM. Statistical analysis was performed using GraphPadPrism 7(GraphPad software). Statistical analysis of the data was performed by using unpaired t-test or ANOVA (single repeated measures ANOVA or two-way ANOVA with Bonferroni test after the fact). Statistical significance was set at P values P <0.05, P <0.01 and P < 0.001. P values less than 0.05 are considered statistically significant.
Effect example 1 whether expression of Grb10 in beta cells is regulated by metabolic stress
Grb10 was highly expressed in human islets (FIGS. 1A-C) and mouse islets, and in islets of diet-induced obese mice (FIG. 1D), ob/ob mice (FIG. 1E), and db/db mice (FIG. 1F). mRNA (fig. 1G) and protein (fig. 1H) levels of Grb10 were also increased in high concentration glucose-treated MIN6 cells. Considering that aging is a key risk factor for the prevalence of type 2 diabetes, we also examined the expression of Grb10 in islets isolated from non-adults and elderly. The elderly islet Grb10 protein expression was increased compared to minors (fig. 1I). The above data indicate that the expression of Grb10 is dynamically regulated by a variety of metabolic stimuli.
TABLE 1 FIG. 1D data
Figure BDA0002702042560000141
TABLE 2 FIG. 1E data
Figure BDA0002702042560000151
TABLE 3 FIG. 1F data
Figure BDA0002702042560000152
TABLE 4 FIG. 1G data
Figure BDA0002702042560000153
TABLE 5 FIG. 1H data
Figure BDA0002702042560000154
TABLE 6 FIG. 1I data
Figure BDA0002702042560000155
Effect example 2 knockout of the beta cell-specific Gene Grb10 improves the ability of the mouse beta cell GSIS and HFD-induced glucose intolerance
To determine the functional role of Grb10 in beta cells, we crossed female Grb10 flox (loxp) mice with male transgenic mice expressing Cre recombinase (Ins2-Cre) to generate beta cell-specific Grb10 knockout mice (Grb10 KO). By immunoblotting and immunofluorescence staining, we found that Grb10ko (ko) mice had a significant reduction in Grb10 protein levels in islets, while wild-type control mice had no significant reduction in Grb10 protein levels (fig. 2A and 4B). The Ins2-cre mediated deletion of Grb10 had no effect on Grb10 levels in other tissues including the hypothalamus. The weight, food intake, body composition and tissue weight of the Grb10KO (KO) mice were not significantly different from those of the control group. On the other hand, the β cell to islet ratio (fig. 2B) and the α cell to islet ratio (fig. 2C) of the Grb10ko (ko) mice were significantly increased and decreased, respectively. Consistent with these findings, Grb10ko (ko) mice showed more beta cell mass than control mice (fig. 2D). In contrast, the number of alpha cells was significantly reduced in Grb10KO (KO) mice (fig. 2E). Immunofluorescence experiments with the ki67 antibody showed that high fat diet Grb10ko (ko) mice proliferated significantly increased numbers of islet cells compared to control mice. The TUNEL method shows that Grb10 gene knockout reduces beta cell apoptosis. On the other hand, the over-expression of Grb10 increased the expression of clear-caspase-3, and inhibited the expression of cyclin D2. Suggesting that Grb10 overexpression may reduce the mass of beta cells by promoting apoptosis and inhibiting cell proliferation. The β -cell specific Grb10 knockout had no significant effect on insulin sensitivity (fig. 2G), but significantly increased glucose tolerance (fig. 2F), indicating that Grb10ko (ko) mice have improved glucose tolerance. High glucose clamp experiments showed that Glucose Infusion Rate (GIR) was higher in Grb10ko (ko) mice than in control mice (fig. 2H). Furthermore, the insulin secretion rate of the Grb10KO (KO) mice was higher than that of the control group mice (FIG. 2I). Transmission electron microscope observation results show that the number of insulin particles, especially the number of mature secretory particles in Grb10KO (KO) mouse cells is obviously increased. Taken together, these data indicate that Grb10 knockout cells can improve beta cell function.
TABLE 7 FIG. 2B data
Figure BDA0002702042560000161
Table 8 fig. 2C data
Figure BDA0002702042560000171
TABLE 9 FIG. 2D data
Figure BDA0002702042560000172
TABLE 10 FIG. 2E data
Figure BDA0002702042560000173
TABLE 11 FIG. 2F data
Figure BDA0002702042560000174
TABLE 12 FIG. 2G data
Figure BDA0002702042560000181
TABLE 13 FIG. 2H data
Figure BDA0002702042560000182
Figure BDA0002702042560000191
TABLE 14 FIG. 2I data
Figure BDA0002702042560000192
Effect example 3 inhibition of Grb10 expression in beta cells protects against HFD-induced loss of beta cell properties
Dedifferentiation causes the beta cells to lose their identity and return to a precursor state, resulting in a reduction in cell number and impaired insulin secretion. To determine the mechanism by which Grb10 regulates beta cell mass, we wanted to figure out whether Grb10 gene knock-out helps maintain beta cell properties. One of the features of beta cell function is the expression of Glut2(Slc2a2), PDX1, NK6 homeobox 1(Nkx6-1), and insulin 1/2. We found that HFD fed Grb10ko (ko) mice islet Glut2 and PDX1 mRNA levels were significantly higher than control mice (fig. 3A). The expression of the insulin genes Ins1 and Ins2 was also higher in the islets of the Grb10KO mouse (fig. 3A). Consistent with the above results, protein levels of Grb10ko (ko) islets Glut2 and PDX1 were significantly higher in mice than in control mice (fig. 3B). There was no significant difference between the control group fed normally and the Glut2 immunoreactivity of islets of Grb10KO (KO) mice, but insulin expression of islets of Grb10KO (KO) mice was significantly higher than that of WT mice (FIG. 3C). The expression of Glut2 and insulin was significantly reduced in WT mice islets 16 weeks after high fat feeding (fig. 3C). Interestingly, HFD-fed Grb10ko (ko) mice expressed higher Glut2 and insulin immunoreactivity, similar to ND-fed WT mice (fig. 3C). In contrast, adenovirus-mediated overexpression of Grb10 in MIN6 cells (Grb10 OE MIN6 cells) significantly reduced mRNA expression of Glut2, PDX1, Nkx6-1, Ins1, and Ins2 (fig. 3D). MIN6 cell Glut2 and PDX1 protein levels of Grb10 OE were significantly reduced (fig. 3E). These findings indicate that down-regulation of Grb10 promotes the ability to maintain beta cell characteristics.
Table 15 fig. 3A data
Figure BDA0002702042560000201
TABLE 16 FIG. 3B data
Figure BDA0002702042560000202
TABLE 17 FIG. 3D data
Figure BDA0002702042560000211
Table 18 fig. 3E data
Figure BDA0002702042560000212
Effect example 4 Grb10 cell-specific knockdown prevented HFD-induced beta cells from returning to the precursor state
To further confirm the potential role of Grb10 in beta cell dedifferentiation, we examined the expression of islet endocrine precursor cell markers in Grb10KO mice and normal control mice. The expression levels of endocrine islet progenitor cell markers Ngn3 and Sox9 in HFD-fed Grb10KO mouse islets were significantly lower than in the control group (fig. 4A). Protein levels of Ngn3 and Sox9 were also significantly reduced (fig. 3B and 4B). In contrast, MIN6 cells from Grb10 OE showed a significant increase in gene expression (fig. 4C) and protein expression (fig. 4D) of Ngn3 and Sox 9. Immunofluorescence analysis confirmed that insulin and Sox9 double positive cells were detected in the pancreas of control mice compared to Grb10ko (ko) mouse tissue (fig. 4E, arrow labeled). The above results indicate that Grb10 induces beta cell dedifferentiation by upregulating cellular endocrine precursor markers.
Table 19 fig. 4A data
Figure BDA0002702042560000213
Table 20 fig. 4C data
Figure BDA0002702042560000221
Table 21 fig. 4D data
Figure BDA0002702042560000222
Effect example 5 potential role of Signal pathway in beta cell dedifferentiation
Knockout of Grb10 in beta cells maintained maturation of beta cells in a mTORC1 dependent manner. A number of signaling pathways such as Hedgehog, Wnt, Notch, and NF-. kappa.b have been identified as signaling pathways that regulate cell dedifferentiation. To elucidate whether these signaling pathways are involved in Grb 10-induced cell dedifferentiation, we examined the expression of key components of these signaling pathways in Grb10 OE MIN6 cells and GFPMIN6 cells. Grb10 overexpression had no significant effect on the expression levels of key components in Notch1, Hes1, p-p65, p65 and the beta-catenin signaling pathway.
Based on the study of Grb10 negatively regulating mTORC1 and the insulin signaling pathway in mammalian cells, we examined the potential role of these signaling pathways in beta cell dedifferentiation. Glut2 and PDX1 gene expression were not altered following treatment of MIN6 cells with PI3 kinase inhibitor Ly 294002. Suggesting that the effect of Grb10 on beta cell dedifferentiation is independent of the insulin signaling pathway. On the other hand, we found that phosphorylation of the islets of Langerhans of Grb10KO (KO) mice at Ser235 site of S6 and at thr37/46 site of 4EBP1 was significantly enhanced upon glucose stimulation compared to WT mice. In addition, overexpression of Grb10 in MIN6 cells inhibited phosphorylation of glucose-stimulated S6 at the Ser235 site and 4EBP1 at the thr37/46 site. These results confirm the inhibitory effect of Grb10 on mTORC1 signaling pathway in cells. The increased Glut2 and PDX1 expression induced by the Grb10 knockout was inhibited by rapamycin treatment, indicating that mTORC1 signaling pathway has a promoting effect in modulating the β cell characteristics of Grb10ko (ko) mice (fig. 5A). In addition, siRNA inhibited Raptor from increasing the expression of endocrine precursor genes such as MIN6 cells Ngn3 and Sox9 (fig. 5B). Similarly, INS-1 cells inhibited increased expression of PDX1 and Glut2 following Grb10, while expression of PDX1 and Glut2 was decreased following rapamycin treatment (FIG. 5C). To further demonstrate the role of mTORC1 signaling pathway in modulating beta cell characteristics in vivo, we administered HFD to greb 10ko (ko) mice and control mice intraperitoneally with rapamycin. Rapamycin injection inhibited mTORC1 signaling pathway for 12 days, with significantly reduced phosphorylation of S6 at Ser235 site and 4EBP1 at thr37/46 site (fig. 5A). Rapamycin had no significant effect on insulin resistance and weight gain in mice, but the improvement in glucose tolerance was significantly reversed in Grb10ko (ko) mice compared to the control group (fig. 5D). The β -cell mass of the Grb10ko (ko) mice was significantly higher than the control mice, but the β -cell mass was significantly reduced after rapamycin use (fig. 5E). The Grb10 knockout caused an increase in Glut2 immunoreactivity, while rapamycin treatment significantly reduced Glut2 immunoreactivity (fig. 5F). The above data indicate that knockout of Grb10 promotes maintenance of beta cell properties by activating mTORC1 signaling pathway.
TABLE 22 FIG. 5B data
Figure BDA0002702042560000231
Table 23 fig. 5D data
Figure BDA0002702042560000232
Figure BDA0002702042560000241
Figure BDA0002702042560000251
Table 24 fig. 5E data
Figure BDA0002702042560000252
Figure BDA0002702042560000261
The foregoing is only a preferred embodiment of the present invention, and it should be noted that, for those skilled in the art, various modifications and decorations can be made without departing from the principle of the present invention, and these modifications and decorations should also be regarded as the protection scope of the present invention.
Sequence listing
<110> Xiangya II Hospital of Zhongnan university
Application of <120> Grb10 as key negative regulator of beta cell dysfunction
<130> MP2026653
<160> 2
<170> SIPOSequenceListing 1.0
<210> 1
<211> 594
<212> PRT
<213> Grb10
<400> 1
Met Ala Leu Ala Gly Cys Pro Asp Ser Phe Leu His His Pro Tyr Tyr
1 5 10 15
Gln Asp Lys Val Glu Gln Thr Pro Arg Ser Gln Gln Asp Pro Ala Gly
20 25 30
Pro Gly Leu Pro Ala Gln Ser Asp Arg Leu Ala Asn His Gln Glu Asp
35 40 45
Asp Val Asp Leu Glu Ala Leu Val Asn Asp Met Asn Ala Ser Leu Glu
50 55 60
Ser Leu Tyr Ser Ala Cys Ser Met Gln Ser Asp Thr Val Pro Leu Leu
65 70 75 80
Gln Asn Gly Gln His Ala Arg Ser Gln Pro Arg Ala Ser Gly Pro Pro
85 90 95
Arg Ser Ile Gln Pro Gln Val Ser Pro Arg Gln Arg Val Gln Arg Ser
100 105 110
Gln Pro Val His Ile Leu Ala Val Arg Arg Leu Gln Glu Glu Asp Gln
115 120 125
Gln Phe Arg Thr Ser Ser Leu Pro Ala Ile Pro Asn Pro Phe Pro Glu
130 135 140
Leu Cys Gly Pro Gly Ser Pro Pro Val Leu Thr Pro Gly Ser Leu Pro
145 150 155 160
Pro Ser Gln Ala Ala Ala Lys Gln Asp Val Lys Val Phe Ser Glu Asp
165 170 175
Gly Thr Ser Lys Val Val Glu Ile Leu Ala Asp Met Thr Ala Arg Asp
180 185 190
Leu Cys Gln Leu Leu Val Tyr Lys Ser His Cys Val Asp Asp Asn Ser
195 200 205
Trp Thr Leu Val Glu His His Pro His Leu Gly Leu Glu Arg Cys Leu
210 215 220
Glu Asp His Glu Leu Val Val Gln Val Glu Ser Thr Met Ala Ser Glu
225 230 235 240
Ser Lys Phe Leu Phe Arg Lys Asn Tyr Ala Lys Tyr Glu Phe Phe Lys
245 250 255
Asn Pro Met Asn Phe Phe Pro Glu Gln Met Val Thr Trp Cys Gln Gln
260 265 270
Ser Asn Gly Ser Gln Thr Gln Leu Leu Gln Asn Phe Leu Asn Ser Ser
275 280 285
Ser Cys Pro Glu Ile Gln Gly Phe Leu His Val Lys Glu Leu Gly Lys
290 295 300
Lys Ser Trp Lys Lys Leu Tyr Val Cys Leu Arg Arg Ser Gly Leu Tyr
305 310 315 320
Cys Ser Thr Lys Gly Thr Ser Lys Glu Pro Arg His Leu Gln Leu Leu
325 330 335
Ala Asp Leu Glu Asp Ser Asn Ile Phe Ser Leu Ile Ala Gly Arg Lys
340 345 350
Gln Tyr Asn Ala Pro Thr Asp His Gly Leu Cys Ile Lys Pro Asn Lys
355 360 365
Val Arg Asn Glu Thr Lys Glu Leu Arg Leu Leu Cys Ala Glu Asp Glu
370 375 380
Gln Thr Arg Thr Cys Trp Met Thr Ala Phe Arg Leu Leu Lys Tyr Gly
385 390 395 400
Met Leu Leu Tyr Gln Asn Tyr Arg Ile Pro Gln Gln Arg Lys Ala Leu
405 410 415
Leu Ser Pro Phe Ser Thr Pro Val Arg Ser Val Ser Glu Asn Ser Leu
420 425 430
Val Ala Met Asp Phe Ser Gly Gln Thr Gly Arg Val Ile Glu Asn Pro
435 440 445
Ala Glu Ala Gln Ser Ala Ala Leu Glu Glu Gly His Ala Trp Arg Lys
450 455 460
Arg Ser Thr Arg Met Asn Ile Leu Gly Ser Gln Ser Pro Leu His Pro
465 470 475 480
Ser Thr Leu Ser Thr Val Ile His Arg Thr Gln His Trp Phe His Gly
485 490 495
Arg Ile Ser Arg Glu Glu Ser His Arg Ile Ile Lys Gln Gln Gly Leu
500 505 510
Val Asp Gly Leu Phe Leu Leu Arg Asp Ser Gln Ser Asn Pro Lys Ala
515 520 525
Phe Val Leu Thr Leu Cys His His Gln Lys Ile Lys Asn Phe Gln Ile
530 535 540
Leu Pro Cys Glu Asp Asp Gly Gln Thr Phe Phe Ser Leu Asp Asp Gly
545 550 555 560
Asn Thr Lys Phe Ser Asp Leu Ile Gln Leu Val Asp Phe Tyr Gln Leu
565 570 575
Asn Lys Gly Val Leu Pro Cys Lys Leu Lys His His Cys Ile Arg Val
580 585 590
Ala Leu
<210> 2
<211> 594
<212> PRT
<213> Grb10
<400> 2
Met Ala Leu Ala Gly Cys Pro Asp Ser Phe Leu His His Pro Tyr Tyr
1 5 10 15
Gln Asp Lys Val Glu Gln Thr Pro Arg Ser Gln Gln Asp Pro Ala Gly
20 25 30
Pro Gly Leu Pro Ala Gln Ser Asp Arg Leu Ala Asn His Gln Glu Asp
35 40 45
Asp Val Asp Leu Glu Ala Leu Val Asn Asp Met Asn Ala Ser Leu Glu
50 55 60
Ser Leu Tyr Ser Ala Cys Ser Met Gln Ser Asp Thr Val Pro Leu Leu
65 70 75 80
Gln Asn Gly Gln His Ala Arg Ser Gln Pro Arg Ala Ser Gly Pro Pro
85 90 95
Arg Ser Ile Gln Pro Gln Val Ser Pro Arg Gln Arg Val Gln Arg Ser
100 105 110
Gln Pro Val His Ile Leu Ala Val Arg Arg Leu Gln Glu Glu Asp Gln
115 120 125
Gln Phe Arg Thr Ser Ser Leu Pro Ala Ile Pro Asn Pro Phe Pro Glu
130 135 140
Leu Cys Gly Pro Gly Ser Pro Pro Val Leu Thr Pro Gly Ser Leu Pro
145 150 155 160
Pro Ser Gln Ala Ala Ala Lys Gln Asp Val Lys Val Phe Ser Glu Asp
165 170 175
Gly Thr Ser Lys Val Val Glu Ile Leu Ala Asp Met Thr Ala Arg Asp
180 185 190
Leu Cys Gln Leu Leu Val Tyr Lys Ser His Cys Val Asp Asp Asn Ser
195 200 205
Trp Thr Leu Val Glu His His Pro His Leu Gly Leu Glu Arg Cys Leu
210 215 220
Glu Asp His Glu Leu Val Val Gln Val Glu Ser Thr Met Ala Ser Glu
225 230 235 240
Ser Lys Phe Leu Phe Arg Lys Asn Tyr Ala Lys Tyr Glu Phe Phe Lys
245 250 255
Asn Pro Met Asn Phe Phe Pro Glu Gln Met Val Thr Trp Cys Gln Gln
260 265 270
Ser Asn Gly Ser Gln Thr Gln Leu Leu Gln Asn Phe Leu Asn Ser Ser
275 280 285
Ser Cys Pro Glu Ile Gln Gly Phe Leu His Val Lys Glu Leu Gly Lys
290 295 300
Lys Ser Trp Lys Lys Leu Tyr Val Cys Leu Arg Arg Ser Gly Leu Tyr
305 310 315 320
Cys Ser Thr Lys Gly Thr Ser Lys Glu Pro Arg His Leu Gln Leu Leu
325 330 335
Ala Asp Leu Glu Asp Ser Asn Ile Phe Ser Leu Ile Ala Gly Arg Lys
340 345 350
Gln Tyr Asn Ala Pro Thr Asp His Gly Leu Cys Ile Lys Pro Asn Lys
355 360 365
Val Arg Asn Glu Thr Lys Glu Leu Arg Leu Leu Cys Ala Glu Asp Glu
370 375 380
Gln Thr Arg Thr Cys Trp Met Thr Ala Phe Arg Leu Leu Lys Tyr Gly
385 390 395 400
Met Leu Leu Tyr Gln Asn Tyr Arg Ile Pro Gln Gln Arg Lys Ala Leu
405 410 415
Leu Ser Pro Phe Ser Thr Pro Val Arg Ser Val Ser Glu Asn Ser Leu
420 425 430
Val Ala Met Asp Phe Ser Gly Gln Thr Gly Arg Val Ile Glu Asn Pro
435 440 445
Ala Glu Ala Gln Ser Ala Ala Leu Glu Glu Gly His Ala Trp Arg Lys
450 455 460
Arg Ser Thr Arg Met Asn Ile Leu Gly Ser Gln Ser Pro Leu His Pro
465 470 475 480
Ser Thr Leu Ser Thr Val Ile His Arg Thr Gln His Trp Phe His Gly
485 490 495
Arg Ile Ser Arg Glu Glu Ser His Arg Ile Ile Lys Gln Gln Gly Leu
500 505 510
Val Asp Gly Leu Phe Leu Leu Arg Asp Ser Gln Ser Asn Pro Lys Ala
515 520 525
Phe Val Leu Thr Leu Cys His His Gln Lys Ile Lys Asn Phe Gln Ile
530 535 540
Leu Pro Cys Glu Asp Asp Gly Gln Thr Phe Phe Ser Leu Asp Asp Gly
545 550 555 560
Asn Thr Lys Phe Ser Asp Leu Ile Gln Leu Val Asp Phe Tyr Gln Leu
565 570 575
Asn Lys Gly Val Leu Pro Cys Lys Leu Lys His His Cys Ile Arg Val
580 585 590
Ala Leu

Claims (10)

  1. Use of Grb10 as a key negative regulator of beta cell dysfunction.
  2. 2. The use of claim 1, wherein the beta cell dysfunction comprises beta cell dedifferentiation, loss of beta cell properties, or beta cell exhaustion.
  3. Use of inhibition of Grb10 in the manufacture of a medicament for the prevention and/or treatment of beta cell dysfunction.
  4. Use of inhibition of Grb10 in the manufacture of a medicament for improving glucose tolerance in an animal.
  5. Use of an inhibitor of Grb10 or a blocker of the Grb 10-regulated signalling pathway in the preparation of a medicament for improving beta cell function, preventing and/or treating diabetes.
  6. 6. Use according to any one of claims 3 to 5, wherein said inhibition of Grb10 comprises a function of reducing the expression of Grb10, the knock-out and/or down-regulation of the Grb10 gene.
  7. 7. The use according to any one of claims 3 to 6, wherein said inhibition of Grb10 promotes beta cell proliferation.
  8. 8. The use according to any one of claims 3 to 7, wherein said inhibition of Grb10 inhibits beta cell dedifferentiation.
  9. 9. The use of claim 5, wherein the signaling pathway regulated by Grb10 enhances beta cell mass and improves beta cell insulin secretion function.
  10. 10. The use of claim 9, wherein the signaling pathway regulated by Grb10 is mTORC1 signaling pathway.
CN202011025653.7A 2020-06-09 2020-09-25 Application of Grb10 as key negative regulator of beta cell dysfunction Pending CN111920955A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010517856.1A CN111494635A (en) 2020-06-09 2020-06-09 Application of Grb10 as key negative regulator of β cell dysfunction
CN2020105178561 2020-06-09

Publications (1)

Publication Number Publication Date
CN111920955A true CN111920955A (en) 2020-11-13

Family

ID=71878817

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202010517856.1A Pending CN111494635A (en) 2020-06-09 2020-06-09 Application of Grb10 as key negative regulator of β cell dysfunction
CN202011025653.7A Pending CN111920955A (en) 2020-06-09 2020-09-25 Application of Grb10 as key negative regulator of beta cell dysfunction

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202010517856.1A Pending CN111494635A (en) 2020-06-09 2020-06-09 Application of Grb10 as key negative regulator of β cell dysfunction

Country Status (1)

Country Link
CN (2) CN111494635A (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114832107A (en) * 2022-04-25 2022-08-02 上海交通大学医学院附属瑞金医院 Application of mTORC1 in preparation of beta cell compensation drugs

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
JINGJING ZHANG等: "Disruption of Growth Factor Receptor–Binding Protein 10 in the Pancreas Enhances b-Cell Proliferation and Protects Mice From Streptozotocin-Induced b-Cell Apoptosis", 《DIABETES》 *
LING LI: "Growth receptor binding protein 10 inhibits glucose-stimulated insulin release from pancreatic b-cells associated with suppression of the insulin/insulin-like growth factor-1 signalling pathway", 《CLINICAL AND EXPERIMENTAL PHARMACOLOGY AND PHYSIOLOGY》 *
张晶晶: "Pancreaticβ-Cell Specific Knockout of Grb10 Improvesβ-Cell Function by Enhancing β-Cell Differentiation and Suppressingβ-Cell Dedifferentiation", 《百度学术》 *
张晶晶: "胰腺组织特异性敲除Grb10基因对胰岛beta细胞功能改善的研究", 《万方》 *
张晶晶等: "新星奖获奖者报告3:胰腺组织特异性敲除Grb10基因对胰岛β细胞功能改善的研究", 《中华内分泌代谢杂志》 *

Also Published As

Publication number Publication date
CN111494635A (en) 2020-08-07

Similar Documents

Publication Publication Date Title
Cipriani et al. Change in populations of macrophages promotes development of delayed gastric emptying in mice
Kuwabara et al. Insulin biosynthesis in neuronal progenitors derived from adult hippocampus and the olfactory bulb
JP6195891B2 (en) Method for producing enteroendocrine cells that produce and secrete insulin
Imbernon et al. Tanycytes control hypothalamic liraglutide uptake and its anti-obesity actions
WO2013055834A2 (en) Er stress relievers in beta cell protection
WO2015148980A2 (en) Methods for detecting and reversing beta cell de-differentiation and uses thereof
WO2014145625A1 (en) Compositions and methods for promoting the generation of endocrine cells
EP2963108A1 (en) Methods for inducing insulin production and uses thereof
Dermentzaki et al. Complex effects of the ZSCAN21 transcription factor on transcriptional regulation of α-Synuclein in primary neuronal cultures and in vivo
Han et al. Indispensable role of HIF-1α signaling in post-implantation survival and angio-/vasculogenic properties of SHED
CN111920955A (en) Application of Grb10 as key negative regulator of beta cell dysfunction
Fang et al. Enhanced cell growth and tumorigenicity of rat glioma cells by stable expression of human CD133 through multiple molecular actions
Liu et al. Ectopic brown adipose tissue formation within skeletal muscle after brown adipose progenitor cell transplant augments energy expenditure
Masjkur et al. Hes3 is expressed in the adult pancreatic islet and regulates gene expression, cell growth, and insulin release
Falchetti et al. Inhibition of telomerase in the endothelial cells disrupts tumor angiogenesis in glioblastoma xenografts
US9745578B2 (en) Targeting microRNA miR-409-3P to treat prostate cancer
Cai et al. GRB10 regulates β-cell mass by inhibiting β-cell proliferation and stimulating β-cell dedifferentiation
CN113577282A (en) Application of Rheb1 as key regulator of beta cell dysfunction
CN114606323A (en) Application of marker LGSN for identifying gastric cancer stem cells as gastric cancer diagnosis and treatment target
CN113398249A (en) Application of over-expressed chemokine CCL14 in preparation of medicine for treating tumors under participation of immune system
CN109172559B (en) Application of ethacrynic acid in preparing medicine for treating and preventing obesity and related metabolic diseases
Sobral et al. Cyclosporin A-induced gingival overgrowth is not associated with myofibroblast transdifferentiation
CN110646621B (en) Application of CHRNA4 in preparation of drug for treating non-alcoholic steatohepatitis
CN114767702B (en) Inhibitor for knocking down circXPO1 and application thereof in preparation of glioma treatment drugs
Chen et al. Synergistic Effect of Hyperglycemia and p27kip1 Suppression on Adult Mouse Islet Beta Cell Replication

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
RJ01 Rejection of invention patent application after publication
RJ01 Rejection of invention patent application after publication

Application publication date: 20201113