CN117143351A - Zn-MOF for treating spinal cord injury and preparation method and application thereof - Google Patents

Zn-MOF for treating spinal cord injury and preparation method and application thereof Download PDF

Info

Publication number
CN117143351A
CN117143351A CN202311271851.5A CN202311271851A CN117143351A CN 117143351 A CN117143351 A CN 117143351A CN 202311271851 A CN202311271851 A CN 202311271851A CN 117143351 A CN117143351 A CN 117143351A
Authority
CN
China
Prior art keywords
mof
spinal cord
injury
cord injury
mice
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
CN202311271851.5A
Other languages
Chinese (zh)
Inventor
纪志盛
伍平
陈天俊
林宏生
廖玉辉
郑举敦
阳华
张国威
邹建宇
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.)
First Affiliated Hospital of Jinan University
Original Assignee
First Affiliated Hospital of Jinan 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 First Affiliated Hospital of Jinan University filed Critical First Affiliated Hospital of Jinan University
Priority to CN202311271851.5A priority Critical patent/CN117143351A/en
Publication of CN117143351A publication Critical patent/CN117143351A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G83/00Macromolecular compounds not provided for in groups C08G2/00 - C08G81/00
    • C08G83/008Supramolecular polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/74Synthetic polymeric materials
    • A61K31/80Polymers containing hetero atoms not provided for in groups A61K31/755 - A61K31/795
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Polymers & Plastics (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to Zn-MOF for treating spinal cord injury and a preparation method and application thereof. The Zn-MOF provided by the invention can effectively reduce the intracellular active oxygen level, reduce the IL-6 and IL-1 beta level after spinal cord injury, inhibit the expression of MMP9, realize remarkable antioxidant capacity, repair hippocampal neuron cells, resist injury induced by oxidative stress, and promote neuron branch formation and neurite growth. Meanwhile, the Zn-MOF can obviously promote oxygen free radicals in the body of a spinal cord injury mouse and promote the repair of spinal cord injury and the recovery of motor functions. The Zn-MO prepared by the method has no obvious toxic or side effect on main tissues and organs of an organism, and the safety is obviously improved, so that a new research thought and a medicine source are provided for treating diseases related to oxidative stress including spinal cord injury, and the Zn-MO has obvious clinical application prospect and social value.

Description

Zn-MOF for treating spinal cord injury and preparation method and application thereof
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to Zn-MOF for treating spinal cord injury and a preparation method and application thereof.
Background
Spinal cord injury (Spinal cord injury, SCI) is a serious complication of spinal injury, often secondary to spinal fractures caused by direct or indirect external forces, such as direct violence, exercise, falls, traffic accidents, overhead falls, and the like. Spinal cord injuries can lead to dysfunction of the patient's extremities, intestines, bladder, and sex, etc., causing serious physical and mental injuries to the patient, greatly reducing the quality of life of the patient. It is counted that there are about 2000 or more tens of thousands of spinal cord injured patients worldwide, and the number of about 70 tens of thousands of patients per year is increasing, with a male to female ratio of about 2:1, and the number of adults is greater than that of children. At present, the treatment of spinal cord injury is mainly focused on surgical treatment, drug treatment, stem cell treatment and the like, and no definite and effective treatment method exists. Therefore, there is an urgent need for more intensive studies on spinal cord injury. .
Spinal cord injury has extremely complex pathophysiological mechanisms, such as cell depletion and death, lack of neurotrophic factors, inflammatory response, axon rupture, demyelination, and gliosis, which inhibit regeneration and recovery of damaged nerves following spinal cord injury. After spinal cord injury occurs, a series of complex pathophysiological changes occur in the injured area, and the changes can be classified into primary injury and secondary injury according to time and pathophysiological changes. Primary injury is the initial stage of injury, and occurs immediately after injury, including spinal cord concussion, contusion and fracture at the time of injury, and spinal cord vascular rupture, erythrocyte shedding in the damaged area, axonal rupture, myelin rupture, etc., and the severity of injury is mainly determined by the degree of initial injury and the duration of compression of the spinal cord. Secondary injury occurs after primary injury, which triggers a complex cascade of secondary injuries that produce further chemical and mechanical injury to spinal cord tissue, including a series of inflammatory cascades that result from the aggregation of various activated inflammatory cells to the injured area, lipid peroxidation, free radical formation, apoptosis, waller degeneration, tissue edema, gliosis, and the like. Spinal cord injury is divided into acute, subacute and chronic phases. The spinal cord injury belongs to the acute phase within 48h, the injury 48h to 14d belongs to the subacute phase, the injury 14d to 6 months belongs to the medium phase, and the injury more than 6 months belongs to the chronic phase. In the acute phase of spinal cord injury, severe bleeding due to spinal cord vascular rupture, and significant increases in the levels of many inflammatory factors such as IL-6, IL-1. Beta. And TNF-alpha, can lead to cellular dysfunction and death. In the acute and subacute phases of spinal cord injury, further injury to the spinal cord can be exacerbated by disruption of the blood-spinal cord barrier, severe inflammatory reactions, and many cytotoxic substances released by inflammatory cells, including free radicals (such as superoxide anions and hydrogen peroxide, etc.). With the resolution of the acute inflammatory response, i.e., the mid-chronic phase of spinal cord injury, vascular remodeling, myelination, neural circuit remodeling, etc., may occur during this phase.
Metal-organic framework Materials (MOFs), also known as porous coordination polymers, are a class of porous crystalline materials assembled from metal ions/clusters and functional organic ligands through strong coordination bonds. MOFs have various structures, have the advantages of simple synthesis, easiness in surface functionalization, high porosity, low density, high specific surface area and loading capacity, regular pore channels, adjustable pore diameter, topological structure diversity and tailorability, good biocompatibility and biodegradability and the like, and have been widely applied to the fields of drug delivery, biological imaging and sensing, gas adsorption/separation/storage, sea water desalination, biomedical application and the like. The preparation method has good drug carrying and release controlling capacity in the fields of MOFs drug transportation, release and the like. However, the conventionally carried metals are manganese and chromium, which have great toxicity and are therefore not suitable for biomedical applications. In order to successfully apply MOFs to biological fields, such as intracellular imaging, drug carriers, etc., the non-toxicity of MOFs must be ensured.
Currently, there is still a very limited treatment for spinal cord injury, and thus, intensive studies on the pathogenesis of spinal cord injury are critical for the treatment of spinal cord injury. Studies have shown that oxidative stress mediated inflammatory responses are one of the important pathogenesis of spinal cord injury and that secondary injury during spinal cord injury treatment can be reduced by relieving oxidative stress. In addition, many studies have shown that metal-organic framework Materials (MOFs) can encapsulate drugs stably, non-toxic within their pores, and release drugs slowly. Thus, MOFs can be used as effective drug carriers for drug delivery. Zinc is a low toxicity transition metal cation and has become an ideal candidate for preparing Zn-MOF, which can be used in biological system applications, especially as a carrier for drugs. However, zn-MOF is currently less studied in terms of spinal cord injury, and there is no article reporting the repair effect of Zn-MOF on spinal cord injury. The research aims at discussing whether Zn-MOF has antioxidant capacity and can promote the repair of spinal cord injury, further researching the mechanism of Zn-MOF for promoting the repair of spinal cord injury, and providing a certain reference basis for clinically treating spinal cord injury.
Disclosure of Invention
The invention aims to solve the defects in the prior art and provides a Zn-MOF for treating spinal cord injury, and a preparation method and application thereof. The Zn-MOF compound provided by the invention can effectively reduce the intracellular active oxygen level, realize remarkable antioxidation capability and promote the growth of neurons and the recovery of motor functions after spinal cord injury. Meanwhile, compared with the conventional metal framework compounds such as manganese, chromium and the like, the Zn-MOF compound has better safety, has no obvious toxic or side effect on neuron cells and main organ tissues of organisms, and provides a new medicine source for the auxiliary treatment of spinal cord injury medicines.
In order to solve the technical problems, the invention is realized by the following technical scheme.
The first aspect of the invention provides a Zn-MOF compound, which is prepared by the following preparation method:
(1) ZnCl 2 Placing gallic acid and water into a round bottom flask, refluxing and stirring uniformly;
(2) Adjusting the pH value to 7-9 by using NaOH solution, and heating in a muffle furnace to obtain a mixture;
(3) Centrifuging the mixture obtained in the step (2), and discarding the supernatant to obtain a light gray solid;
(4) And (3) washing the light gray solid obtained in the step (3) with water and drying to obtain the light gray solid.
Preferably, the ZnCl in step (1) 2 And gallic acid in a mass ratio of 1:2-6.
Preferably, the concentration of NaOH in the step (2) is 10mol/L.
Preferably, the heating temperature in the step (2) is 110-170 ℃ and the heating time is 12-36h; most preferably, the heating temperature is 140 ℃ and the heating time is 24 hours.
Preferably, the rotational speed of the centrifugation in the step (3) is 10000-15000rpm, the centrifugation time is 10-20min, and the centrifugation temperature is 2-6 ℃; most preferably, the rotational speed of the centrifugation is 10000-15000rpm, the centrifugation time is 15min, and the centrifugation temperature is 4 ℃.
Preferably, the number of times of washing in step (4) is 1 to 3; most preferably, the number of washes is 2.
The second aspect of the invention provides a method for preparing a Zn-MOF compound, comprising the steps of:
(1) ZnCl 2 Placing gallic acid and water into a round bottom flask, refluxing and stirring uniformly;
(2) Adjusting the pH value to 7-9 by using NaOH solution, and heating in a muffle furnace to obtain a mixture;
(3) Centrifuging the mixture obtained in the step (2), and discarding the supernatant to obtain a light gray solid;
(4) And (3) washing the light gray solid obtained in the step (3) with water and drying to obtain the light gray solid.
Preferably, the ZnCl in step (1) 2 And gallic acid in a mass ratio of 1:2-6.
Preferably, the concentration of NaOH in the step (2) is 10mol/L.
Preferably, the heating temperature in the step (2) is 110-170 ℃ and the heating time is 12-36h; most preferably, the heating temperature is 140 ℃ and the heating time is 24 hours.
Preferably, the rotational speed of the centrifugation in the step (3) is 10000-15000rpm, the centrifugation time is 10-20min, and the centrifugation temperature is 2-6 ℃; most preferably, the rotational speed of the centrifugation is 10000-15000rpm, the centrifugation time is 15min, and the centrifugation temperature is 4 ℃.
Preferably, the number of times of washing in step (4) is 1 to 3; most preferably, the number of washes is 2.
In a third aspect, the invention provides the use of a Zn-MOF compound as described above or a Zn-MOF compound prepared according to the preparation method described above in the preparation of a medicament for the treatment of a disease associated with oxidative stress.
Preferably, the disorder associated with oxidative stress is selected from spinal cord injury.
In a fourth aspect, the invention provides a pharmaceutical composition for the treatment of a disease associated with oxidative stress comprising a Zn-MOF compound as described above or a Zn-MOF compound prepared according to the preparation method described above, and a pharmaceutically acceptable carrier.
Preferably, the disorder associated with oxidative stress is selected from spinal cord injury.
Preferably, the pharmaceutically acceptable carrier is selected from one or more of filler, disintegrant, lubricant, adhesive, antioxidant, antibacterial agent, correctant, aromatic agent and chelating agent.
Spinal cord injury involves a complex inflammatory response process, coordinated by a number of inflammatory factors including IL-1 beta, IL-6. The expression of IL-1 beta, IL-6 is dramatically increased after spinal cord injury. It has been found that overexpression of IL-6 promotes glioblast proliferation and, therefore, lowering IL-6 levels during the acute phase of spinal cord injury can inhibit gliosis. In addition, high levels of IL-6 after spinal cord injury inhibit the growth of neuronal axons. IL-1β is one of the most widely studied pro-inflammatory cytokines, which is produced mainly by microglia, and in addition, neurons, astrocytes, etc. can also produce IL-1β. In the injured central nervous system, the expression of IL-1β is dramatically increased. IL-1β can activate the immune system, promote the production of inflammatory factors and other cytokines, and also activate neurotoxic mediators. IL-1β has a great negative impact on the formation of glial cells after spinal cord injury, plasticity of axons, etc., and is associated with a severe functional prognosis. Studies have shown that IL-1β knockout can reduce astrocyte proliferation and inhibit inflammatory responses following spinal cord injury.
Matrix metalloproteinase 9 (MMP 9), one of the most widely studied members of the family of matrix metalloproteinases (Matrix Metalloproteinases, MMPs), is a gelatinase. MMP9 is involved in degradation of extracellular matrix and can promote degradation of gelatin, type IV, type V and type XI collagen, myelin basic protein, elastin, etc. Death. After spinal cord injury, the expression of MMP9 is increased, which is associated with inflammation after spinal cord injury and disruption of the blood-spinal cord barrier. Neutrophils, macrophages and monocytes involved in the spinal cord injury inflammatory response can all express MMP9.MMP9 is upregulated and reaches maximum activity 12-24h after spinal cord injury, and neutrophils at the site of spinal cord injury are the primary source of MMP9. Inhibiting MMP9 activity can reduce vascular permeability and reduce inflammation in the first 3 days after spinal cord injury. During the development of the entire nervous system, there is expression of MMP9 and it is involved in the growth of axons during development.
The synthesized Zn-MOF can effectively reduce the intracellular active oxygen level, realize remarkable antioxidation capability, reduce the levels of IL-6 and IL-1 beta after spinal cord injury, and inhibit the expression of MMP9. The Zn-MOF treatment can promote branch formation and neurite growth, which indicates that the Zn-MOF can repair neuron cells and resist oxidative stress induced injury. In addition, in vivo studies show that the recovery of motor functions of spinal cord injured mice can be significantly promoted after Zn-MOF treatment, and BMS score improvement is obvious. Meanwhile, the ROS bioluminescence intensity in the spinal cord injury mice treated by Zn-MOF is obviously reduced, and the Zn-MOF has obvious oxygen free radical scavenging and spinal cord injury repairing effects in vivo. In addition, the methods of a foot-to-air experiment, a rod rotating experiment, a foot print experiment, a Catwalk gait analysis, an electrophysiological evaluation and the like show that the hind limb movement function of the mice after Zn-MOF treatment is obviously superior to that of the SCI model group, and further prove that the Zn-MOF can obviously improve the repair of spinal cord injury and promote the recovery of limb functions.
Compared with the prior art, the invention has the following beneficial effects:
(1) The Zn-MOF compound provided by the invention can effectively reduce the intracellular active oxygen level, reduce the IL-6 and IL-1 beta level after spinal cord injury, inhibit the expression of MMP9, realize remarkable antioxidant capacity, repair hippocampal neuron cells, resist injury induced by oxidative stress, and promote neuron branch formation and neurite growth. Meanwhile, the Zn-MOF can obviously promote oxygen free radicals in the body of a spinal cord injury mouse and promote the repair of spinal cord injury and the recovery of motor functions.
(2) The Zn-MOF compound prepared by the invention has no obvious toxic or side effect on HT22 cells, hippocampal neurons and main tissues and organs of organisms, and compared with the common metal framework compounds such as manganese, chromium and the like in the prior art, the safety is obviously improved, thereby providing a new research thought and medicine source for treating oxidative stress related diseases including spinal cord injury, and having obvious clinical application prospect and social value.
Drawings
FIG. 1 is a schematic representation of transmission electron microscopy characterization of Zn-MOF.
FIG. 2 is a schematic diagram showing the distribution of C, O, zn elements in Zn-MOF.
FIG. 3 is a schematic representation of the results of Zn-MOF analysis by a Fourier infrared spectrometer and a nanoparticle size analyzer.
FIG. 4 is a graph showing the result of Zn-MOF radical scavenging ability.
FIG. 5 is a schematic representation of the effect of DCFH-DA staining analysis of Zn-MOF at different concentrations on HT22 cell ROS levels.
FIG. 6 is a graph showing the results of flow cytometry analysis of the effect of different concentrations of Zn-MOF on HT22 cell ROS levels.
FIG. 7 is a graph showing the effect of different Zn-MOF concentrations on HT22 cell viability.
FIG. 8 is a graph showing the effect of different Zn-MOF concentrations on hippocampal neuronal cell viability.
FIG. 9 is a graph showing the effect of Zn-MOF on primary hippocampal neurite extension.
Fig. 10 is a schematic diagram of BMS scoring results for different groups.
Figure 11 is a morphological image of different groups of mice 60 days after spinal cord injury.
FIG. 12 is a graph showing the results of reactive oxygen species levels in different groups of mice.
FIG. 13 is a graph showing the results of foot fault rate for different groups of mice.
Fig. 14 is a graph showing drop latency results for different groups of mice.
Figure 15 is a graph showing the results of step size and step width for different groups of mice,
fig. 16 is a graph showing the results of the average of the maximum contact area of the left hind limb and the maximum contact average intensity of the left hind limb of the mice of different groups.
Fig. 17 is a graph showing the results of the average of the maximum contact area of the right hind limb and the maximum contact average intensity of the right hind limb of the mice of different groups.
FIG. 18 is a graph showing the results of the normal index of the step sequences of mice of different groups.
FIG. 19 is a graph showing the results of exercise-induced potentials of mice of different groups.
FIG. 20 is a graphical representation of HE staining of spinal cord tissue of different groups of mice 60 days after spinal cord injury.
FIG. 21 is a graph showing the results of HE staining of the main organs of different groups of mice.
FIG. 22 is a graph showing the results of serum IL-6 and IL-1. Beta. Content detection in mice of different groups.
FIG. 23 is a graph showing MMP9 content detection results of different groups of mice.
Detailed Description
In order to make the objects, technical solutions and effects of the present invention more clear and clear, the present invention will be described in further detail with reference to examples. It should be understood that the specific embodiments described herein are for purposes of illustration only and are not intended to limit the scope of the invention.
All reagents used in the context of the present invention are commercially available, unless otherwise specified. For animal experiments, the related procedures and methods meet the medical ethics requirements. The experimental methods used in the present invention are all conventional methods and techniques in the art.
Representative results of selection from the biological experimental replicates are presented in the context figures, and data are presented as mean±sd and mean±sem as specified in the figures. All experiments were repeated at least three times. Data were analyzed using GraphPad Prism 5.0 or SPSS 22.0 software. And comparing the average value difference of two or more groups by adopting conventional medical statistical methods such as t-test, chi-square test, analysis of variance and the like.pA difference of < 0.05 was considered significant.
Example 1 preparation and characterization of Zn-MOF
(1) 1.0g ZnCl 2 3.8g of gallic acid and 50g of water are placed in a round bottom flask and are stirred under reflux.
(2) The pH was adjusted to 8 using 10mol/L NaOH solution, and the mixture was heated in a muffle furnace (Sxi-8-10, new Korean instrument Co., ltd.) at 140℃for 24 hours.
(3) Centrifuging the mixture obtained in the step (2) at the temperature of 4 ℃ and the speed of 12000rpm for 15min, and discarding the supernatant to obtain a light gray solid.
(4) And (3) washing the light gray solid obtained in the step (3) with water for 2 times and drying to obtain the Zn-MOF compound.
The Zn-MOF compound prepared above is taken, the morphology and structure of the Zn-MOF compound are observed by a transmission electron microscope, and the morphology and structure information of the sample is obtained by carrying out amplified imaging on an electron beam carrying the internal structure information of the sample, and the result is shown in figure 1. The elements on the surface of the Zn-MOF compound were then characterized using an X-ray photoelectron spectrometer, and the results are shown in FIG. 2. The results show that the Zn-MOF compound prepared exhibits a sphere-like shape with a diameter of about 100nm, and the distribution of Zn, C, O elements therein is shown in FIG. 2. The Zn-MOF compounds were further examined by a nano-particle size analyzer, respectively, and the results are shown in FIG. 3. The test result of the nanometer particle size analyzer shows that: the Zn-MOF diameter was approximately equal to 100nm, consistent with the transmission electron microscope measurements described above. The above test results indicate successful synthesis of Zn-MOF compounds.
Example 2 investigation of the Effect of Zn-MOF on oxidative stress in vitro
The Zn-MOF compound prepared in example 1 was taken and examined for its free radical scavenging ability by the ABTS method, and the specific procedure was as follows:
(1) 20mL of PBS solution was placed in a 50mL centrifuge tube, 54.87mg of ABTS was weighed and added to the centrifuge tube containing the PBS solution, and the ABTS stock solution was prepared by thoroughly mixing the solution.
(2) Preparing a piece of filter paper and placing a sufficient amount of MnO on the filter paper 2 The filter paper is then combined with MnO 2 Transfer to the top of the centrifuge tube, then to MnO 2 And (3) dropwise adding 10mL of the ABTS stock solution prepared in the step (1), filtering for 5min in a dark place, continuing to react for 12h in a dark place to form an ABTS free radical mixed solution containing ABTS free radicals (ABTS+), and storing at the temperature of-20 ℃ for standby.
(3) 100. Mu.L of Zn-MOF (6. Mu.g/mL, 12.5. Mu.g/mL, 25. Mu.g/mL, 50. Mu.g/mL, 100. Mu.g/mL) at various concentrations were mixed with 100. Mu.L of the ABTS free radical mixture solution, respectively; a blank and a control were also provided, wherein only 200. Mu.L of PBS solution was added to the blank, and 100. Mu.L of PBS solution and 100. Mu.L of ABTS free radical mixture solution were added to the control.
(3) Absorbance at 734nm was measured using a cell imaging multimode reader (station 5,BioTek Instruments Inc.) at 10min, 20min, 40min, 60min, 90min and 120min, respectively.
The detection results are shown in FIG. 4. The results show that Zn-MOF can obviously inhibit the formation of ABTS free radicals, has time and dose dependence, can obviously inhibit the ABTS free radicals at the concentration of 6 mug/mL, and can reach the inhibition rate (in x) of the ABTS free radicals close to 100% at the concentration of 12.5 mug/mLp< 0.001, vs 0. Mu.g/mL group (control)). The experiment shows that Zn-MOF has higher activity of eliminating ABTS, can effectively eliminate free radicals outside cells to play an antioxidant role, and can be used as an effective free radical scavenger to prevent ROS from damaging neurons.
Subsequently, HT22 cells were examined by using an active oxygen fluorescent probe (DCFH-DA) and flow cytometry to verify whether Zn-MOF also had significant free radical scavenging activity in the cells, wherein the DCFH-DA experiment was performed as follows:
(1) 9mL of DMEM culture medium without serum is absorbed and placed in a 15mL centrifuge tube wrapped by tinfoil and protected from light, 9 mu L of DCFH-DA stock solution is added, and DCFH-DA solution is prepared after fully and uniformly mixing and placed in the protected from light environment for storage.
(2) The cell damage model was constructed by adding 120mmol/L of sodium L-glutamate to HT22 cells, followed by exposure to 37℃and 5% CO 2 Is cultured in a cell culture incubator for 12 hours.
(3) After the cell damage model is successfully constructed, the cells are treated for 12 hours by adopting Zn-MOFs (12.5 mug/mL, 25 mug/mL, 50 mug/mL and 100 mug/mL) with different concentrations respectively; a blank (Control) was also set up, wherein the blank was normal HT22 cells, and a Control was HT22 cells damaged with sodium L-glutamate and added with Zn-MOF at 0. Mu.g/mL.
(4) Taking HT22 cells (cultured in 24-well plate) out of cell incubator, removing original culture medium, gently adding 200 μl of DCFH-DA solution prepared in step (1) into each well along the wall of the well, wrapping 24-well plate with tinfoil to avoid light transmission, and thenIt was placed at 37℃with 5% CO 2 Is cultured in a cell culture tank for 30min.
(5) Taking out the cells in an incubator to a light-proof room, removing DCFH-DA solution, washing twice with PBS, and then adding 500 mu L of TBST (containing Triton X-100) solution into each well to punch 3 times for 5min each time; and then sealing the film by ICC technology, and taking pictures under a laser confocal microscope.
The results are shown in FIG. 5. The results showed that the fluorescence intensity of the Control group (injured without dosing) was significantly increased compared to the blank group (Control group), indicating that the construction of the sodium L-glutamate injured HT22 cell model was successful. And, the fluorescence intensity of the Zn-MOF group was significantly reduced compared to the control group, indicating that Zn-MOF effectively inhibited intracellular ROS and alleviated oxidative stress.
The specific steps for detecting active oxygen by flow cytometry are as follows:
(1) Taking HT22 cells in logarithmic growth phase, removing original culture medium, adding 120mmol/L sodium L-glutamate solution, and then placing at 37deg.C and 5% CO 2 Culturing for 12h in a cell culture box, and constructing a cell damage model.
(2) After the cell damage model is successfully constructed, the cells are treated for 12 hours by adopting Zn-MOFs (12.5 mug/mL, 25 mug/mL, 50 mug/mL and 100 mug/mL) with different concentrations respectively; a blank (Control) was also set up, wherein the blank was normal HT22 cells, and a Control was HT22 cells damaged with sodium L-glutamate and added with Zn-MOF at 0. Mu.g/mL.
(3) The cell supernatant was removed, the cells were digested with pancreatin and resuspended in culture plates, then the cell resuspension and cell supernatant were mixed and placed in a 5mL centrifuge tube, centrifuged at 2000rpm for 5min, and the supernatant was discarded. Cells were resuspended in 500. Mu.L of PBS and transferred to a 1.5mL centrifuge tube and centrifuged at 2000rpm for 5min.
(7) Adding 300 mu L of active oxygen DCFH-DA dye solution (Biyun day), gently mixing, and incubating for 20min in dark place; centrifuging at 2000rpm for 5min, discarding supernatant, adding 500 μl PBS solution, gently mixing, centrifuging at 2000rpm for 5min again, and discarding supernatant; 300. Mu.L of PBS solution was added, and the mixture was gently mixed and transferred to a flow tube for detection.
The results are shown in FIG. 6. The results are similar to the DCFH-DA experiment, i.e. ROS levels are significantly increased in the Control group (injured without dosing) compared to the blank group (Control group); the ROS levels were significantly reduced in the Zn-MOF group compared to the control group, indicating that Zn-MOF effectively inhibited intracellular ROS and alleviated oxidative stress. The ROS level detection of HT22 cells by the reactive oxygen fluorescent probe and the flow cytometry fully proves that Zn-MOF can effectively inhibit ROS in cells to play a role in relieving oxidative stress.
Further, HT22 cells were selected for toxicity testing of Zn-MOF, as follows:
(1) HT22 cells were seeded in 96-well plates at a cell number of 5000 cells/100. Mu.L/well using a cell counter.
(2) After cell adhesion, zn-MOF was added at various concentrations (1.57,3.13,6.25, 12.5, 25, 50, 100, units: μg/mL) to each well and incubated for an additional 12 hours. A blank and a Control group were additionally provided, wherein the blank was only added with an equal volume of medium without seeding cells, and the Control group (Control group) was seeded with an equal volume of cells without Zn-MOF treatment.
(3) After 12h, 110. Mu.L of the prepared CCK-8 solution (10. Mu.L of MEM solution+10. Mu.L of CCK-8 stock solution) was added, and after mixing, the 96-well plate was wrapped with tin foil to protect from light, and then incubation was continued for 1h.
(4) After incubation, the OD (absorbance) value (absorbance) at 450nm wavelength of each group of cells was measured with a microplate reader, and then the cell viability was calculated according to the formula: cell viability (%) = (dosing group-blank)/(Control group-blank) ×100%.
The results are shown in FIG. 7. The results showed that at each concentration of 1.57-100. Mu.g/mL studied, the survival rate of the hippocampal neurons after Zn-MOF treatment was close to 100%, and the survival rate was not significantly different from that of the Control group (Control group)p> 0.05). From this, it was confirmed that Zn-MOF was excellent in safety and not significantly cytotoxic to HT 22.
Subsequently, rat hippocampal tissue cells were selected to further verify the toxicity of Zn-MOF, specifically as follows:
(1) Brain hippocampus tissue from 1 day old Sprague-Dawley (SD) rats was minced with ophthalmic scissors and added to a 15mL centrifuge tube (Corning), papain (Sigma, cat#P 4762) was added and digested at 37℃for 25 minutes.
(2) Tissue is washed after stopping the protease with bovine serum albumin.
(3) The isolated hippocampal neurons were seeded at 20000 cells/mL in 96-well plates (Corning) at 200. Mu.L per well in DMEM medium (glutamine, gibco, carlsbad, calif., cat # 12430054) containing 10% fetal bovine serum, 10% nutrient mix F-12, at 37℃at 95% O 2 、5% CO 2 Is cultured in an incubator for 6 hours.
(4) Replacing original culture medium with Neurobasal medium containing 5% B-27 (Gibco), placing at 37deg.C and 95% O 2 、5% CO 2 Is cultured in an incubator for 12 hours.
(5) After cell adhesion, zn-MOF was added at various concentrations (0.40,0.79,1.57,3.13,6.25, 12.5, 25, 50, 100, units: μg/mL) to each well and incubated for an additional 24 hours. A blank and a Control group were additionally provided, wherein the blank was only added with an equal volume of medium without seeding cells, and the Control group (Control group) was seeded with an equal volume of cells without Zn-MOF treatment.
(6) After 12h, 110. Mu.L of the prepared CCK-8 solution (10. Mu.L of MEM solution+10. Mu.L of CCK-8 stock solution) was added, and after mixing, the 96-well plate was wrapped with tin foil to protect from light, and then incubation was continued for 1h.
(7) After incubation, the OD (absorbance) value (absorbance) at 450nm wavelength of each group of cells was measured with a microplate reader, and then the cell viability was calculated according to the formula: cell viability (%) = (dosing group-blank)/(Control group-blank) ×100%.
The results are shown in FIG. 8. The results show that at each concentration of 0.40-100 mug/mL, the survival rate of the hippocampal neuron cells after Zn-MOF treatment is close to 100%, and the survival rate is not significantly different from that of a blank control group p> 0.05). Thereby proceeding toThe Zn-MOF has excellent safety and no obvious cytotoxicity to the hippocampal neuron cells.
EXAMPLE 3 repair of neurons by Zn-MOF
The method selects rat hippocampal tissue for Zn-MOF neuron damage repair detection, and comprises the following specific steps:
(1) Rat brain hippocampus tissue of 1 day old Sprague-Dawley (SD) was cut with an ophthalmic scissors and added to a 15mL centrifuge tube (Corning), papain (Sigma, cat#P 4762) was added and digested at 37℃for 25 minutes; tissue is washed after stopping the protease with bovine serum albumin.
(2) The isolated hippocampal neurons were seeded at 20000 cells/mL in 24-well plates (Corning) at 500. Mu.L per well in DMEM medium (glutamine, gibco, carlsbad, calif., cat # 12430054) containing 10% fetal bovine serum, 10% nutritional mix F-12, at 37deg.C, 95% O 2 、5% CO 2 Culturing in an incubator for 24 hours;
(3) Replacing original culture medium with 120 mu M L-sodium glutamate-containing culture medium, placing at 37deg.C and 95% O 2 、5% CO 2 Culturing for 12 hours in an incubator of (2) to form a hippocampal neuron damage model;
(4) After cell adhesion, zn-MOF with different concentrations (12.5, 25, 50, 100, unit: μg/mL) was added to each well, and incubation was continued for 24 hours, wherein Control group was a blank Control group, and hippocampal neuronal cells were not subjected to damage treatment; a blank (Control) and a Control (Injury) were additionally provided, wherein the blank was normal hippocampal neuronal cells and the Control was damage-modelled hippocampal neuronal cells, without Zn-MOF treatment. Hippocampal neuronal cells were stained and mounted using the ICC technique, the mounted slides were photographed using a laser confocal microscope, and finally the number of neuronal primary projections, secondary projections, and total projections and their length were delineated and counted using Image J software.
The effect of Zn-MOF on neuronal damage repair was evaluated by quantifying the length and total number of neuronal projections, and the results are shown in FIG. 9. The results showed that neurons of the Injury group were primary prominent, secondary, compared to the Control groupThe number and length of projections, as well as the total projections, were significantly reduced, indicating that the construction of the primary hippocampal neuron model for sodium L-glutamate injury was successful. In the Zn-MOF treated group, the length of the primary and secondary branches of the neurons and the total length of the projections were significantly greater than in the model group (x)p<0.05,**p<0.01,***p< 0.001, vs Injury group (0. Mu.g/mL group)), and after Zn-MOF treatment, the primary branch length of the neuron and the total length of the protrusion have no significant difference from the blank Control group (Control group). It was also found that the number of primary and secondary branches and the total number of projections were significantly greater than the model group (x) in neurons treated with Zn-MOFp<0.01,***p< 0.001, vs Injury group (0. Mu.g/mL group)), and after Zn-MOF treatment, the number of primary branches of neurons and the total number of projections can be restored to have no significant difference from the blank group. In conclusion, the Zn-MOF compound prepared by the invention can obviously promote the growth of injured neuron protrusion after being added, and has obvious protection and recovery promotion effects on the ROS-induced neuron oxidative stress injury by inhibiting the generation of ROS to promote the axon extension of injured neurons.
Example 4 in vivo functional study of Zn-MOF
(1) 24 female mice of 6 weeks of age were selected and divided into four groups of 6, including a blank Control group (Control group, 6, without any treatment), a sham Control group (sham group, 6, with only the lamina excised, without damaging the spinal cord), a model group (Injury group, 6), a Zn-MOF group (injury+zn-MOF, 6), and the following day after spinal cord Injury, each drug was injected intraperitoneally at a volume of 100 μl/dose of 50 μg, with sham group and Injury group injected with equal volumes of PBS, 1 time daily, once daily, for three consecutive days. The method for constructing the spinal cord injury mouse model is as follows:
a. hair on the back of the mice was removed, and skin on the back of the mice was exposed.
b. Anesthesia: anesthesia was successfully indicated by intraperitoneal injection of 1.25% tribromoethanol at a dose of 200 μg/20g, smooth breathing, significant weakening of limb muscle strength, and disappearance of pain reflex and cornea reflex.
c. Exposing spinal cord: wiping the skin for 2 times by using an iodophor cotton ball to disinfect the skin; the level of thoracic vertebrae at paragraphs 9-11 (T9-T11) was determined and a longitudinal incision of 2.5cm length was made centrally on the back at the level of T9-T11. The paraspinal muscles were blunt stripped, spinous processes and laminae were removed with rongeurs, the spinal cord of the T9-T11 segment was thoroughly exposed, the T10 plane was fixed with a fixator, and the T10 lamina was excised, taking care to avoid additional tissue damage during the procedure. And then distracted with an anchor to fully expose the spinal cord.
d. The mice were fixed to a lewis-wirl-injury system device (Louisville Injury System Apparatus, LISA), spinal cord was adjusted to under the impactor, and the impact position was confirmed by monitoring the laser beam thereof, and the extent of spinal cord injury was determined by the depth of impact, in this example, the impact tip was set to 18psi, the depth of impact was 0.8mm, and the injury time was 0.5s; after the collision is completed, the mice are removed from the injury device and removed from the holder. After sufficient hemostasis was given, the mice were sutured to the muscle and skin using sutures. Mice in the sham group were T10 laminectomy and did not undergo spinal cord impact.
e. The incision was closed, gentamicin 2000U was subcutaneously injected post-operatively, 1 time daily, 3 days in succession, and 1 time of artificial bladder emptying was performed every 8 hours until spontaneous urination.
(2) Monitoring: starting from post-molding, each group of mice was monitored daily for motor status and BMS scored every 3 days, with the results shown in fig. 10. The results show that initially, all SCI mice, except the blank, had complete loss of hind limb locomotion. Subsequently, hindlimb motor function began to gradually recover over time in each group of SCI mice, with the recovery rate of Zn-MOF treated groups at day 6 of Injury being already significantly higher than in the model group (Injury group), with a gradual improvement in BMS scores. The BMS scores of Control and Sham mice continued to be 9 points throughout the duration of the experiment, while the BMS scores of Injury mice tended to increase slowly, but by day 60 the scores did not exceed 1 point, and the rate of score increase was very slow. However, the BMS score increased more rapidly and scored higher for Zn-MOF group mice than for Injury group, and the score was approximately 4 up to day 60, indicating that Zn-MOF can promote recovery of motor function in spinal cord injured mice. The morphology of each group of mice after 60 days post Injury is shown in FIG. 11, wherein the hind limbs of the Control group and the Sham group mice are normal, the hind limbs of the Injury group mice are paralyzed, and the hind limb morphology of the Zn-MOF group mice is improved compared with the Injury group mice. The results show that Zn-MOF treatment significantly enhanced behavioral functional recovery after SCI in mice.
(3) In vivo imaging of small animals: mice were imaged in vivo on day 3 post injury. First, 0.1mL/20g L-012 (chemiluminescent (CHL) probe) was injected intraperitoneally, and the concentration of L-012 was 4mg/mL. After 5 minutes, the anesthesia is injected into the abdominal cavity by using 1.25% tribromoethanol at the dosage of 200 mug/20 g, and after the anesthesia is successful, the living animal imaging is carried out.
The results are shown in FIG. 12. The results show that the bioluminescence of ROS is rarely seen in normal mouse spinal cord, whereas the bioluminescence intensity of ROS in spinal cord of spinal cord injured mice is significantly increased and increases with time. After Zn-MOF treatment, the bioluminescence intensity of ROS was significantly reduced, significantly lower than that of the model group (Injury group), even lower than that of Sham group (x)p<0.05,**p< 0.01, vs Injury group (0. Mu.g/mL group)). The quantitative value of the ROS bioluminescence intensity indicates that the Zn-MOF promotes the repair of spinal cord injury by reducing ROS at the spinal cord of a mouse, and further proves that the Zn-MOF has obvious oxygen free radical scavenging in vivo and can reduce the high active oxygen level in the mouse after spinal cord injury, thereby playing an antioxidant role and promoting the repair after spinal cord injury.
On day 60 after spinal cord injury, mice were subjected to a foot-to-air test and a stick-turning test. The pedal-air experiment comprises the following specific steps: the mice were placed on parallel bars, framed with a square rack, allowed to move freely within a defined range, and then video-imaged for 3-5 min. In the crawling process of the mice, the lower limbs are not stepped on the parallel rods, the sensing device can automatically record the mice as foot errors once, record the total number of foot errors of 60 steps of crawling of the mice on the parallel rods, and then calculate the foot error rate of each mouse. Foot error rate = number of times the parallel bars were dropped from the lower limbs/total number of steps (60 steps).
The experimental results are shown in FIG. 13. The results showed that the foot error rate was significantly higher in the Injury group of mice compared to the Control and Sham groups, indicating that the hindlimb locomotor function was poor in the Injury group of mice. However, the foot error rate was significantly reduced in the Zn-MOF group compared to the Injury group, indicating that Zn-MOF can promote recovery of hindlimb motor function in spinal cord injured mice.
The rotating rod experiment comprises the following specific steps: turning on the rotating rod switch to make it rotate continuously; grabbing 1 mouse and placing the mouse on a rotating rod, enabling the movement direction of the mouse to be opposite to the rotation direction of the rotating rod, starting timing when the mouse is just placed on the rotating rod, and recording the movement time of each mouse on the rotating rod when the mouse falls off the rotating rod; data were collected and analyzed.
The experimental results are shown in FIG. 14. The results show that the drop latency of the Injury group mice is significantly shorter than that of the Control and Sham groups. Whereas the drop latency of the Zn-MOF group mice was significantly longer than that of the Injury group, further demonstrating that Zn-MOF can promote recovery of motor function in spinal cord injured mice.
Right hind limb paralysis of the half-cut injured mice at the early stage of injury (7 days) presents trawling, and joint movement is less and cannot be evaluated. And then, the lower limb functions are gradually recovered after 7 to 14 days, the dragging is reduced, and the knee joint and hip joint activities are obviously increased. Thus, footprint analysis was performed 14 days after injury, and the evaluation parameters in the footprint analysis experiment included step size and step width. The method comprises the following specific steps:
The experimental mice were randomly divided into 4 groups, designated as group 1-group 4, and spinal cord injury model preparation was performed according to the method described above. Wherein group 1 is Control, group 2 is Sham, group 3 is model (Injury), group 4 is Zn-MOF, right spinal cord half-cut in row T10, and Zn-MOF treatment is given simultaneously in the same manner as described above. Before the experiment, a long and narrow channel is prepared, white paper is paved in the channel, the hind feet of each group of mice are dyed by nontoxic red ink, then the mice are put into the channel prepared in advance, the mice walk freely along the channel, the hind paw footprints of the mice are visible on the white paper, and the step size or the step width is analyzed by calculating the average value of continuous 3 footprints.
The experimental results are shown in fig. 15, and the results show that the spinal cord half-cut model can cause partial loss of hindlimb locomotor function of the mice, and compared with Sham group (group 2), the hindlimb locomotor function of the mice in the Injury group (group 3) is dragged (fig. 15A), and the hindlimb locomotor function of the mice after Zn-MOF treatment (group 4) is significantly better than that of the Injury group. The footprint quantitative statistical analysis results show that the stride and the stride width of the mice in the experimental group at 14 days after operation are better than those of the SCI group (fig. 15B and 15C) (n=6 in all groups p<0.05,**p<0.01,***p<0.001, ns stands for insignificant). Thus, it is further demonstrated that Zn-MOF can significantly improve spinal cord injury repair and promote recovery of limb functions.
Subsequently, catWalk gait analysis was performed on different groups of mice, specifically as follows: placing the mice in a darkened room for a moment; then the mouse is grabbed and placed at the tail end of the organic glass floor of the CatWalk less analysis system, a cover is covered, the mouse freely passes through one end of the floor to the other end, meanwhile, the process that the mouse freely passes through the organic glass floor is observed on a computer, the mouse cannot stay in the process that the mouse passes through the organic glass floor, otherwise, the mouse must be repeated once, and after the mouse passes through the organic glass floor very smoothly, the computer automatically saves the motion video of the mouse; repeating the steps until gait related data of all experimental mice are acquired.
The experimental results are shown in FIGS. 16-18. The results showed that the average of the maximum contact area of the double hindlimb and the average of the maximum contact strength of the double hindlimb were very small for the Injury mice compared to the Control group and the Sham group, indicating that the double hindlimb locomotor function was poor for the Injury mice. However, both the mean of the maximum contact area and the mean of the maximum contact intensity of the double hind limbs were significantly increased in the Zn-MOF group mice compared to the Injury group (fig. 16-17). The normative index of the gait in the Injury mice was very small compared to the Control and Sham mice, indicating that the hindlimb locomotor function was poor in the Injury mice. Whereas the step normal index was significantly increased in the Zn-MOF group compared to the Injury group (fig. 18). The CatWalk gait analysis result shows that Zn-MOF can promote the recovery of the motor function of the spinal cord injury mice.
The above mouse behavioural tests demonstrate that Zn-MOF can promote the recovery of motor function in spinal cord injured mice. Electrophysiological testing was then performed to verify whether Zn-MOF could promote recovery of nerve conduction function in spinal cord injured mice. The method comprises the following specific steps: 1.25% 2, 2-tribromoethanol is adopted to anesthetize the mice (the dosage is 0.2mL/10 g), the hairs of the head and the thigh of the mice are removed, and the cerebral cortex movement area and the bilateral thigh gastrocnemius muscle groups of the mice are dissected; turning on the neurophysiological monitor, and setting parameters as follows: the voltage is 0.05mV, and the current is 0.05mA. All signal processing procedures were performed on BL-420N biosignal acquisition and analysis system (Chengdu Union); inserting a stimulating electrode into a cerebral cortex exercise area of a mouse, inserting an electrode for receiving electric signals into a bilateral gastrocnemius muscle group of the mouse, then collecting signals of the gastrocnemius exercise evoked potentials of the mouse, and repeating the collection of the exercise evoked potentials of each mouse for 3 times; after all mice were collected, they were analyzed.
The experimental results are shown in FIG. 19. The results show that the amplitude of the movement-induced potential of the Injury group mice is obviously reduced compared with that of the Control group and the Sham group, and the amplitude of the movement-induced potential of the Zn-MOF group mice is obviously increased compared with that of the Injury group, which proves that the Zn-MOF can effectively promote the recovery of the nerve conduction function of the spinal cord injured mice.
To more intuitively evaluate the repair of Zn-MOF to the damaged spinal cord of mice, HE staining was performed on spinal cord tissue of each group of mice on day 60 after spinal cord injury to observe the morphology of spinal cord tissue. The results show that the spinal cord tissues of the Control group and the Sham group are not obviously damaged; the spinal cord tissue of the Injury group can see obvious structural damage; the significantly reduced extent of spinal tissue Injury in the Zn-MOF group compared to the Injury group, suggesting that Zn-MOF may indeed promote repair of injured spinal cord in the mice (see figure 20).
In the previous experiments, it was verified that Zn-MOF has no obvious toxic or side effects on HT22 cells and hippocampal neurons in vitro, and to further evaluate the safety, HE staining was performed on major organs of each group of mice on day 60 after spinal cord injury to observe pathological changes of alarm, heart, liver, spleen, lung, kidney, and the results are shown in fig. 21. The results show that the brain, heart, liver, spleen, lung and kidney of different groups of mice have no obvious difference, which indicates that Zn-MOF has no obvious toxic and side effects on the brain, heart, liver, spleen, lung and kidney of mice, and further proves that Zn-MOF has higher safety.
Example 5 study of the mechanism of action of Zn-MOF
To clarify the mechanism of action of Zn-MOF and its relationship with inflammatory response after spinal cord injury, zn-MOF was injected in spinal cord injured mice for three consecutive days. The sera of the mice of the different groups were then subjected to enzyme-linked immunosorbent assay (ELISA) to detect the amounts of inflammatory factors IL-6 and IL-1β in the sera of the mice of the different groups, the results are shown in FIG. 22. The results show that the levels of IL-6 and IL-1β in the mice of the Injury group were significantly increased compared to the Control and Sham groups, while the levels of IL-6 and IL-1β in the mice of the Zn-MOF group were significantly decreased compared to the Injury group, indicating that Zn-MOF can inhibit IL-6 and IL-1β and thereby reduce the inflammatory response after spinal cord Injury.
Since the level of expression of MMP9 is generally increased after spinal cord injury, MMP9 acts as an extracellular enzyme that degrades a range of extracellular matrix proteins and participates in the spinal cord injury process. In this regard, protein extraction and immunoblotting (Western blot) were further performed on cortical neurons and spinal cord tissues of different groups of spinal cord injured mice, and then detection was performed using MMP9 antibodies, and the results are shown in fig. 23. The results show that the content of MMP9 in different groups of cortical neurons is compared: MMP9 expression was increased in the Injury group compared to the Control group, while MMP9 expression was decreased in the Zn-MOF group compared to the Injury group (FIG. 23A). Comparison of the content of MMP9 in spinal cord tissue of mice of different groups: MMP9 expression was increased in the Injury group compared to the Control and Sham groups, while MMP9 expression was significantly decreased in the Zn-MOF group compared to the Injury group (FIG. 23B). It was demonstrated that Zn-MOF could reduce MMP9 expression following spinal cord injury.
Spinal cord injury involves very complex pathophysiological mechanisms, one of which is the oxidative stress-mediated inflammatory response. The invention starts from the relation between oxidative stress and spinal cord injury, discusses the relation between Zn-MOF and oxidative stress, researches the influence of Zn-MOF on the axon extension of injured neurons at the cellular level and the influence of Zn-MOF on the spinal cord injury repair of mice at the animal level, and researches the action mechanism of Zn-MOF in the spinal cord injury repair.
Firstly, the invention verifies that Zn-MOF has antioxidant effect outside cells through ABTS+ free radical scavenging experiments, and verifies that Zn-MOF can inhibit ROS in cells at the cellular level to play an antioxidant role and promote the axon extension of injured neurons. Then, by constructing a mouse spinal cord injury model and researching the action of Zn-MOF after the mouse spinal cord injury, the Zn-MOF is found to have an antioxidant action in a spinal cord injury mouse body, the level of IL-6 and IL-1 beta after the spinal cord injury and the expression of MMP9 can be reduced, and the restoration of the motor function and the nerve conduction function of the spinal cord injury mouse is promoted, so that the restoration of the injured spinal cord of the mouse is promoted.
Studies have shown that the pathogenesis of neurodegenerative diseases may be related to oxidative stress and excitatory toxins. Glutamate, an excitatory neurotransmitter, may cause certain neurological diseases that may mediate oxidative stress damage and excitotoxicity, a phenomenon known as glutamate neurotoxicity. It can cause damage to many cellular components, including mitochondria, ultimately leading to cell death. During cell death, large amounts of Reactive Oxygen Species (ROS) such as superoxide anions (O2. Cndot.) are produced - ) Hydrogen peroxide (H) 2 O 2 ) Hydroxyl radicals (OH.) and the like, the overproduction of ROS mediates the excitotoxicity of glutamate. The invention damages HT22 cells by using L-sodium glutamate, so that excessive ROS are generated, and then DCFH-DA staining and flow cytometry are used for detecting the ROS level in HT22 cells, so that the Zn-MOF can inhibit the ROS in the cells to play a role in relieving oxidative stress.
Neurons are the most susceptible cell types in the central nervous system to oxidative stress, and are particularly susceptible to ROS. ROS can damage nucleic acids, proteins, lipids, etc. of cells, leading to neuronal dysfunction and even neuronal death. ROS are involved in the development of many neurodegenerative diseases such as alzheimer's disease, amyotrophic lateral sclerosis, parkinson's disease, etc. It has been thought that endogenously produced ROS can act as signaling molecules to regulate a range of neurological physiological processes, such as neuronal development, neuronal polarity, synaptic plasticity, growth cone routing, and regulation of neural circuits. However, excessive ROS production can lead to an imbalance in the oxidation and antioxidant mechanisms, leading to oxidative stress. The present invention verifies whether Zn-MOF promotes repair of damaged neurons by inhibiting ROS by counting the number and length of neuronal projections. The results show that the number and length of the primary and secondary projections, as well as the total projections, of neurons of the Zn-MOF group are significantly increased compared to the Injury group. It is demonstrated that Zn-MOF can promote axonal extension of injured neurons by inhibiting ROS. Further, it was confirmed whether Zn-MOF can exert its antioxidant effect in spinal cord injured mice by detecting the level of active oxygen in mice using L-012 and in vivo imaging of mice. The results show that Zn-MOF can reduce the high active oxygen level in mice after spinal cord injury, thereby playing a role in relieving oxidative stress.
To investigate whether Zn-MOF could promote repair of spinal cord injury in mice, mice of different groups were tested by BMS scoring, foot-to-foot experiments, rod rotation experiments, foot print experiments, catWalk gait analysis, electrophysiological evaluation, etc. The result shows that the hindlimb motor function of the Zn-MOF group mice is recovered faster 60 days after spinal cord Injury, the BMS score is close to 4 minutes, and the foot error rate is obviously reduced compared with the Injury group; the results of the rod rotating experiment show that the drop latency of the Zn-MOF group mice is obviously prolonged compared with that of the Injury group; the footprint experiment result shows that the step size and the step width of the Zn-MOF group mice are obviously increased compared with those of the Injury group; the CatWalk gait analysis result shows that the average value of the maximum contact area of the double hind limbs, the average strength of the maximum contact of the double hind limbs and the normal index of the walking sequence of the Zn-MOF group mice are improved by the harness compared with the Injury group. In addition, at day 60 after spinal cord injury, the morphology of spinal cord tissue was observed by HE staining of spinal cord tissue of different groups of mice. The results show a significant reduction in the extent of spinal tissue damage in the Zn-MOF group compared to the Injury group. Together, the above results demonstrate that Zn-MOF can promote recovery of motor function in spinal cord injured mice.
In addition, the invention has also conducted intensive research on the safety of Zn-MOF. As a result, zn-MOF was found to be non-significantly toxic to both HT22 cells and primary hippocampal neurons. And after 60 days of spinal cord injury, the main organs of different mice are detected and analyzed, and the brain, heart, liver, spleen, lung and kidney of the mice in different groups have no obvious difference, so that the Zn-MOF has no obvious toxic and side effects on the brain, heart, liver, spleen, lung and kidney of the mice, and the Zn-MOF has higher safety.
The above detailed description describes the analysis method according to the present invention. It should be noted that the above description is only intended to help those skilled in the art to better understand the method and idea of the present invention, and is not intended to limit the related content. Those skilled in the art may make appropriate adjustments or modifications to the present invention without departing from the principle of the present invention, and such adjustments and modifications should also fall within the scope of the present invention.

Claims (10)

1. A Zn-MOF compound characterized in that it is prepared by the following preparation method:
(1) ZnCl 2 Placing gallic acid and water into a round bottom flask, refluxing and stirring uniformly;
(2) Adjusting the pH value to 7-9 by using NaOH solution, and heating in a muffle furnace to obtain a mixture;
(3) Centrifuging the mixture obtained in the step (2), and discarding the supernatant to obtain a light gray solid;
(4) And (3) washing the light gray solid obtained in the step (3) with water and drying to obtain the light gray solid.
2. The Zn-MOF compound as defined in claim 1 wherein in step (1) the ZnCl 2 And gallic acid in a mass ratio of 1:2-6.
3. The Zn-MOF compound of claim 1, wherein the concentration of NaOH in step (2) is 10mol/L.
4. The Zn-MOF compound of claim 1, wherein the heating in step (2) is at a temperature of 110 to 170 ℃ for a time of 12 to 36 hours.
5. The Zn-MOF compound as defined in claim 1 wherein the rotational speed of centrifugation in step (3) is 10000-15000rpm, the time of centrifugation is 10-20min and the temperature of centrifugation is 2-6 ℃.
6. The Zn-MOF compound as defined in claim 1 wherein the number of times of washing in step (4) is 1 to 3.
7. Use of a Zn-MOF compound as defined in any one of claims 1 to 6 in the manufacture of a medicament for the treatment of a disease associated with oxidative stress.
8. The use according to claim 8, wherein the disorder associated with oxidative stress is selected from spinal cord injury.
9. A pharmaceutical composition for treating a disease associated with oxidative stress, comprising a Zn-MOF compound as described above or a Zn-MOF compound prepared according to the above preparation method, and a pharmaceutically acceptable carrier.
10. The pharmaceutical composition of claim 9, wherein the disorder associated with oxidative stress is selected from spinal cord injury.
CN202311271851.5A 2023-09-28 2023-09-28 Zn-MOF for treating spinal cord injury and preparation method and application thereof Pending CN117143351A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311271851.5A CN117143351A (en) 2023-09-28 2023-09-28 Zn-MOF for treating spinal cord injury and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202311271851.5A CN117143351A (en) 2023-09-28 2023-09-28 Zn-MOF for treating spinal cord injury and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN117143351A true CN117143351A (en) 2023-12-01

Family

ID=88912190

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311271851.5A Pending CN117143351A (en) 2023-09-28 2023-09-28 Zn-MOF for treating spinal cord injury and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN117143351A (en)

Similar Documents

Publication Publication Date Title
Deng et al. Collagen scaffold combined with human umbilical cord-mesenchymal stem cells transplantation for acute complete spinal cord injury
Gao et al. An anti-inflammatory and neuroprotective biomimetic nanoplatform for repairing spinal cord injury
AU2008292407B2 (en) Prophylactic or alleviating agent for peripheral nerve disorder induced by anti-cancer agent
Parikh et al. Curcumin-loaded self-nanomicellizing solid dispersion system: Part II: In vivo safety and efficacy assessment against behavior deficit in Alzheimer disease
Tian et al. Exogenous CGRP regulates apoptosis and autophagy to alleviate traumatic brain injury through Akt/mTOR signalling pathway
Azizi et al. Improvement of functional recovery of transected peripheral nerve by means of chitosan grafts filled with vitamin E, pyrroloquinoline quinone and their combination
KR101329936B1 (en) External preparation for skin containing flavanone derivative
WO2023202477A1 (en) Dihydromyricetin tetrahydropyrrole complex and preparation method therefor, and application
Zhang et al. Berberine enhances survival and axonal regeneration of motoneurons following spinal root avulsion and re-implantation in rats
Li et al. Chitosan-modified hollow manganese dioxide nanoparticles loaded with resveratrol for the treatment of spinal cord injury
Huang et al. Localised delivery of quercetin by thermo-sensitive PLGA-PEG-PLGA hydrogels for the treatment of brachial plexus avulsion
CN117653739B (en) Preparation method of Ce@UCNP-BCH and application thereof in spinal cord injury treatment
Puniak et al. Comparison of a serotonin antagonist, opioid antagonist, and TRH analog for the acute treatment of experimental spinal trauma
Ladol et al. The effects of Hippophae rhamnoides in neuroprotection and behavioral alterations against iron-induced epilepsy
Zhu et al. Schwann cell-derived exosomes and methylprednisolone composite patch for spinal cord injury repair
RU2545734C1 (en) Drug preparation for parkinson disease
RU2275924C2 (en) Method for preparing complex of biologically active polypeptides for normalization of brain function and pharmaceutical agent based on thereof
CN117143351A (en) Zn-MOF for treating spinal cord injury and preparation method and application thereof
CN111281927A (en) Traditional Chinese medicine composition for treating Parkinson&#39;s disease and preparation method and application thereof
CN115671147B (en) Auricularia auricula extract and application thereof
KR101289220B1 (en) Peptide substance stimulating regeneration of central nervous system neurons, pharmaceutical composition on its base, and the method of its application
CN116904571A (en) Use of CPNE1 gene or CPNE1 protein in the preparation of a medicament for preventing or treating neurological disorders
Chen et al. Bexarotene enhances astrocyte phagocytosis via ABCA1-mediated pathways in a mouse model of subarachnoid hemorrhage
RU2766083C1 (en) Biologically active substance in the form of a bioregulatory composition, exhibiting a supportive and restorative effect on tissues and organs, and pharmacological composition
CN117304501A (en) AIE-N for the treatment of spinal cord injury 3 -Zn-MOF and preparation method and application thereof

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