WO2025044677A1 - Use of prrt2 acting on forebrain excitatory neuron and up-regulator thereof - Google Patents
Use of prrt2 acting on forebrain excitatory neuron and up-regulator thereof Download PDFInfo
- Publication number
- WO2025044677A1 WO2025044677A1 PCT/CN2024/109426 CN2024109426W WO2025044677A1 WO 2025044677 A1 WO2025044677 A1 WO 2025044677A1 CN 2024109426 W CN2024109426 W CN 2024109426W WO 2025044677 A1 WO2025044677 A1 WO 2025044677A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- prrt2
- expression
- vector
- forebrain
- promoter
- 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
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/1703—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- A61K38/1706—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from fish
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/1703—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- A61K38/1709—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/08—Antiepileptics; Anticonvulsants
Definitions
- the present invention belongs to the field of biomedicine, and more specifically, the present invention relates to the application of PRRT2 and its up-regulator acting on forebrain excitatory neurons.
- Epilepsy is a typical central nervous system disease caused by excitation-inhibition imbalance.
- the main clinical manifestations include convulsions, muscle spasms, tonic convulsions, atonia and absence, accompanied by a brief loss of consciousness.
- the patient's EEG shows epileptic discharge characteristics.
- Epileptic seizures are characterized by suddenness and recurrence, which has a serious impact on the patient's physical and mental health and normal social activities.
- the causes of epilepsy are relatively diverse.
- pathogenic factors include developmental abnormalities, genetic mutations, craniocerebral injury, cerebrovascular disease and inflammation. Except for a small number of self-limited epilepsy, most epilepsy patients need to achieve control of epilepsy through treatment.
- the treatment methods currently used in clinical practice mainly include drug therapy, surgical resection of local lesions, ketogenic diet, vagus nerve stimulation and deep brain stimulation.
- more than two-thirds of epilepsy patients mainly use anti-epileptic drugs for epilepsy control.
- anti-epileptic drugs targeting different targets have been launched since the 1950s, forming a relatively effective clinical treatment plan, about 30% of patients with refractory epilepsy still cannot benefit from these drugs or drug combinations.
- some epilepsy patients who respond well to existing drugs still face the problem of adverse drug reactions during long-term medication and the inability to fully control epilepsy.
- the above unmet medical needs are an important driving force for the research of new mechanisms and the development of new therapies.
- the homeostatic balance of neural networks depends on the cooperation between excitatory and inhibitory neurons, and epilepsy is mainly manifested as abnormal synchronous excitement of the brain caused by imbalance of neural network homeostasis. Therefore, the ideal epilepsy intervention strategy is to selectively reduce the activity of excitatory neurons or increase the activity of inhibitory neurons through drugs or other treatments, so as to correct the abnormal excitement of the neural network and return it to normal homeostasis; at the same time, the intervention plan should try to avoid or reduce interference with non-epilepsy-related neural networks, such as movement, arousal, cognition and emotional circuits, so as to reduce adverse reactions during long-term medication.
- non-epilepsy-related neural networks such as movement, arousal, cognition and emotional circuits
- Voltage-gated sodium channels (hereinafter referred to as sodium channels or Nav) are important molecules that the generation of action potentials and impulse conduction of neurons in the brain rely on, and are the basis of the excitability of neural tissue.
- sodium channels are important targets for anti-epileptic drugs. More than a quarter of the anti-epileptic drugs currently approved for clinical use target such channels, including sodium phenytoin, carbamazepine, oxcarbazepine, and lacosamide. The mechanism of action of these drugs is to reduce the excitability of neural networks by regulating the activity of sodium channels, thereby reducing or preventing related types of epileptic seizures.
- anti-epileptic drugs targeting sodium channels have been widely used in the treatment of epilepsy, their central adverse reactions (such as dizziness, drowsiness, ataxia, and irritability) have not been effectively resolved.
- Nav1.2 and Nav1.6 are mainly distributed in excitatory neurons with glutamate as the main neurotransmitter, while Nav1.1 is mainly distributed in inhibitory interneurons with ⁇ -aminobutyric acid (GABA) as the main neurotransmitter; in the cerebellar cortex related to motor regulation, Nav1.1, Nav1.2 and Nav1.6 are expressed separately or in combination in GABAergic interneurons, granule cells and Purkinje neurons.
- GABA ⁇ -aminobutyric acid
- Antiepileptic drugs that target sodium channels generally lack selectivity among sodium channel members. After entering the central nervous system, they will act on non-epilepsy-related neural cell groups and cause adverse central reactions.
- Cell state selectivity refers to the ability of anti-epileptic drugs or treatments to act on cells that are abnormally excited during epileptic seizures without interfering with the normal functions of similar cells in the non-epileptic state.
- drugs with poor selectivity for cell epileptic states often cause patients to feel uncomfortable during daily medication (non-epileptic state), affecting their daily life and work, and even causing patients to interrupt medication and cause recurrence of epilepsy.
- the purpose of the present invention is to provide a use of an upregulator of proline-rich transmembrane protein 2 (PRRT2) that selectively acts on forebrain excitatory neurons, for the preparation of drugs for treating or preventing epilepsy.
- PRRT2 proline-rich transmembrane protein 2
- an upregulator of PRRT2 that selectively acts on forebrain excitatory neurons for preparing a drug for treating or preventing epilepsy.
- the upregulation is a significant upregulation, promotion, increase or enhancement; for example, an upregulation, promotion, increase or enhancement of 10%, 20%, 30% or more.
- the selective action on forebrain excitatory neurons includes: selectively acting on the slow inactivation of sodium ion channels (preferably Nav1.2 and Nav1.6), and/or selectively acting on abnormal cell excitability.
- the treating or preventing epilepsy comprises reducing the number, frequency, level and/or duration of epileptic seizures.
- the treating or preventing epilepsy does not affect the subject's daily activities.
- the number, frequency, level and/or duration of epileptic seizures are assessed by behavioral experiments in mice.
- the behavioral experiments include EEG and behavioral observations.
- daily behavior is assessed by a mouse behavioral experiment, wherein the behavioral experiment includes but Not limited to: open field test, wheel test, social behavior test.
- the subjects of the use include: humans, non-human primates and rodents.
- the upregulator of PRRT2 that selectively acts on forebrain excitatory neurons is a construct, or an expression system formed by the construct (e.g., a viral (infection) system); the construct includes: an expression drive system, and a PRRT2 encoding gene whose expression is driven by the drive system.
- the expression driving system comprises a single or combined forebrain excitatory neuron-specific expression driving system.
- the forebrain excitatory neuron-specific expression driving system includes a single form of forebrain glutamatergic neuron-specific promoter, including (but not limited to): CaMKIIa promoter; a single form of promoter is used to directly drive the expression of PRRT2.
- the forebrain excitatory neuron-specific expression driving system includes a combination of forebrain glutamatergic neuron-specific promoters, including (but not limited to): a forebrain excitatory neuron-specific promoter for driving Cre recombinase expression (such as Emx1 promoter or CaMKIIa promoter) and a strong promoter for driving PRRT2 expression (such as but not limited to beta-actin promoter (CAG) promoter); the combined expression driving system is used to take into account both forebrain excitatory neuron specificity and high-efficiency expression characteristics.
- a forebrain excitatory neuron-specific promoter for driving Cre recombinase expression such as Emx1 promoter or CaMKIIa promoter
- PRRT2 expression such as but not limited to beta-actin promoter (CAG) promoter
- the construct includes an operational gene expression regulatory element that depends on Cre recombinase, such as (but not limited to) Double-floxed inverse orientation, DIO); preferably, the element for DIO is LoxP/Lox2272, and the Cre recombinase-dependent expression of PRRT2 is performed using specific recognition sequences based on LoxP and Lox2272.
- Cre recombinase such as (but not limited to) Double-floxed inverse orientation, DIO
- the element for DIO is LoxP/Lox2272
- the Cre recombinase-dependent expression of PRRT2 is performed using specific recognition sequences based on LoxP and Lox2272.
- the construct includes: construct 1, including sequentially connected: a promoter (such as but not limited to beta-actin promoter), LoxP/Lox2272, a PRRT2 encoding gene, LoxP/Lox2272; the PRRT2 encoding gene is reversely connected between two pairs of LoxP/Lox2272 sequences; construct 2: including sequentially connected: a forebrain excitatory neuron-specific expression promoter, a Cre recombinase encoding gene (such as an Emx1 promoter driving the expression of the Cre recombinase encoding gene).
- a promoter such as but not limited to beta-actin promoter
- LoxP/Lox2272 such as but not limited to beta-actin promoter
- PRRT2 encoding gene is reversely connected between two pairs of LoxP/Lox2272 sequences
- construct 2 including sequentially connected: a forebrain excitatory neuron-specific expression promoter, a Cre recomb
- the expression of the gene encoding Cre recombinase is driven by a forebrain excitatory neuron-specific expression promoter; the Cre recombinase acts on LoxP/Lox2272 to convert the reverse PRRT2 encoding gene into a forward connection, and its promoter drives the expression.
- an inducible expression system (such as but not limited to Tet-on/off) may also be provided to achieve the on/off of regulatable PRRT2 expression.
- the construct is contained in an expression vector or is directly inserted into the genome of the intervention object through gene editing;
- the expression vector includes: viral vector, non-viral vector; preferably, the viral vector includes: adeno-associated virus (AAV) vector, lentiviral vector, adenoviral vector, retroviral vector; more preferably, the adeno-associated virus vector includes: PHP.eB serotype AAV vector, Cap-B10 serotype AAV vector; or the gene editing includes (but is not limited to) gene editing based on CRISPR-Cas (such as Cas9) technology.
- AAV adeno-associated virus
- the gene editing includes (but is not limited to) gene editing based on CRISPR-Cas (such as Cas9) technology.
- the PRRT2 is: (a) a protein having an amino acid sequence as shown in SEQ ID NO: 2, 4 or 6; (b) a protein derived from (a) which is obtained by replacing, deleting or adding one or more (e.g., 1-20; preferably 1-15; more preferably 1-10, such as 5, 3) amino acid residues in the amino acid sequence of the protein (a), and has the function of the protein (a); (c) a protein derived from (a) which has more than 80% (preferably more than 85%; more preferably more than 90%; more preferably more than 95%, such as 98%, 99%) homology with the amino acid sequence of the protein (a) and has the function of the protein (a); or (d) a protein formed by adding a tag sequence to the N or C terminus of any of the polypeptides of (a) to (c), or adding a signal peptide sequence or a secretion signal sequence to its N terminus.
- a construct for treating or preventing epilepsy comprising: an expression drive system, and a PRRT2 encoding gene whose expression is driven by the drive system.
- the expression driving system comprises a single or combined forebrain excitatory neuron-specific expression driving system.
- the forebrain excitatory neuron-specific expression driving system includes a single form of forebrain glutamatergic neuron-specific promoter, including (but not limited to): CaMKIIa promoter; a single form of promoter is used to directly drive the expression of PRRT2.
- the forebrain excitatory neuron-specific expression driving system includes a combination of forebrain glutamatergic neuron-specific promoters, including (but not limited to): a forebrain excitatory neuron-specific promoter for driving Cre recombinase expression (such as Emx1 promoter or CaMKIIa promoter) and a strong promoter for driving PRRT2 expression (such as but not limited to beta-actin promoter (CAG) promoter); the combined expression driving system is used to take into account both forebrain excitatory neuron specificity and high-efficiency expression characteristics.
- a forebrain excitatory neuron-specific promoter for driving Cre recombinase expression such as Emx1 promoter or CaMKIIa promoter
- PRRT2 expression such as but not limited to beta-actin promoter (CAG) promoter
- the construct includes: an operably linked operative gene expression regulatory element dependent on Cre recombinase, such as (but not limited to) Double-floxed inverse orientation, DIO); preferably, the element for DIO is LoxP/Lox2272, and the Cre recombinase-dependent PRRT2 expression is performed using specific recognition sequences based on LoxP and Lox2272.
- an operably linked operative gene expression regulatory element dependent on Cre recombinase such as (but not limited to) Double-floxed inverse orientation, DIO
- the element for DIO is LoxP/Lox2272
- the Cre recombinase-dependent PRRT2 expression is performed using specific recognition sequences based on LoxP and Lox2272.
- the construct is contained in an expression vector or is directly inserted into the genome of the intervention object through gene editing, and the expression vector includes: a viral vector, a non-viral vector; preferably, the viral vector includes: an adeno-associated virus (AAV) vector, a lentiviral vector, an adenoviral vector, a retroviral vector; more preferably, the adeno-associated virus vector includes: a PHP.eB serotype AAV vector, a Cap-B10 serotype AAV vector.
- AAV adeno-associated virus
- the gene editing includes (but is not limited to) gene editing based on CRISPR-Cas (such as Cas9) technology.
- an expression system for treating or preventing epilepsy is provided, which is a viral system obtained by packaging the viral vector.
- a drug for treating or preventing epilepsy or a drug kit containing the drug is provided, wherein the drug contains the expression system.
- a method for selectively delivering an effective amount of PRRT2, or the PRRT2 upregulator that selectively acts on forebrain excitatory neurons, to forebrain excitatory neurons comprising contacting the forebrain excitatory neurons with an expression vector comprising an effective amount of PRRT2, or its upregulator;
- the expression vector comprises: a viral vector, a non-viral vector; more preferably, the viral vector comprises: an adeno-associated virus (AAV) vector, a lentiviral vector, an adenoviral vector, a retroviral vector; more preferably, the adeno-associated virus vector comprises: a PHP.eB serotype AAV vector, a Cap-B10 serotype AAV vector;
- the expression vector is delivered by intraorbital injection (preferably orbital vein injection), intracranial injection, intrathecal (spinal cord) injection, intrathecal (cerebral cisterna magna) injection,
- a method for selectively acting on slow inactivation of sodium ion channels, selectively acting on abnormal cell excitability, and/or selectively acting on forebrain excitatory neurons comprising administering to a subject an effective amount of PRRT2, or an upregulator thereof (preferably an expression vector that overexpresses the PRRT2).
- FIG. 1 The protein coding region (CDS) sequence of the mouse Prrt2 gene.
- Fig. 2 Amino acid sequence of mouse PRRT2 protein.
- FIG. 1 Schematic diagram of EGFP control plasmid pCAG-empty-IRES-EGFP.
- Figure 4 Schematic diagram of mouse PRRT2 overexpression plasmid pCAG-mouse PRRT2(HA)-IRES-EGFP.
- Fig. 6 Amino acid sequence of human PRRT2 protein.
- Figure 7 Schematic diagram of human PRRT2 overexpression plasmid pCAG-human PRRT2(HA)-IRES-EGFP.
- Fig. 8 The protein coding region (CDS) sequence of the zebrafish Prrt2 gene.
- Fig. 9 Amino acid sequence of zebrafish PRRT2 protein.
- Figure 10 Schematic diagram of zebrafish PRRT2 overexpression plasmid pCAG-zebra PRRT2(HA)-IRES-EGFP.
- Fig. 11 Effect of PRRT2 overexpression on the fast inactivation process of Nav1.2 sodium channel.
- the upper part is a schematic diagram of the voltage clamp scheme for activating sodium ion channels.
- the lower part is a representative sodium channel current curve.
- Fig. 13 Effect of PRRT2 overexpression on the slow inactivation process of Nav1.2 sodium channel.
- the top is a schematic diagram of the voltage clamp protocol for detecting the slow inactivation of sodium channels.
- the bottom is an example of the sodium current measured during the first depolarization of control cells and the sodium current measured after the sodium channels experienced depolarization of different lengths followed by a brief hyperpolarization.
- Fig. 14 Effect of PRRT2 overexpression on the recovery of Nav1.2 sodium channel from slow inactivation.
- Fig. 15 Effect of zebrafish PRRT2 overexpression on slow inactivation of Nav1.2 sodium channel.
- Figure 16 Schematic diagram of adeno-associated virus overexpressing mCherry plasmid pAAV-CAG-DIO-mCherry.
- Figure 17 Schematic diagram of adeno-associated virus overexpressing mouse PRRT2 plasmid pAAV-CAG-DIO-mouse PRRT2-HA.
- PRRT2-OE PRRT2 over-expression.
- FIG. 19 Overexpression of PRRT2 in forebrain excitatory neurons inhibits convulsant-induced epileptic seizures.
- FIG. 20 Overexpression of PRRT2 in forebrain excitatory neurons inhibits epileptic EEG induced by convulsants.
- Pentylenetetrazol can induce epileptiform EEG signals in the mouse cerebral cortex; overexpression of PRRT2 in forebrain excitatory neurons can effectively inhibit PTZ-induced epileptiform discharge events in the mouse cerebral cortex.
- FIG. 22 Overexpression of PRRT2 in forebrain inhibitory neurons facilitates convulsant-induced epileptic seizures.
- the inventors discovered for the first time that the protein encoded by the PRRT2 (proline-rich transmembrane protein 2) gene can selectively act on sodium channels in a slow inactivation state, and reduce the effective supply of activatable sodium channels by enhancing the slow inactivation of sodium channels. Furthermore, the inventors selectively overexpressed the PRRT2 protein in forebrain excitatory neurons to feedback reduce the excitability of neurons in abnormally excited states, and achieved the goal of effectively suppressing epileptic seizures without interfering with daily behavior in a mouse epilepsy model.
- the specific expression of PRRT2 in forebrain excitatory neurons is the key to successfully intervening in epilepsy and reducing side effects.
- the expression of PRRT2 in forebrain excitatory neurons has both cell type selectivity and cell state selectivity, and has good application prospects in the field of epilepsy treatment.
- PRRT2 includes proteins having SEQ ID NO: 2 (mouse), SEQ ID NO: 4 (human), or SEQ ID NO: 6 (zebrafish), and also includes variant forms of sequences having the same function as PRRT2.
- PRRT2 also includes proteins having a homology of 80% or higher with the polypeptide sequence defined by the above SEQ ID NO: 2, 4 or 6; preferably a homology of 85% or higher, such as 90%, 95%, 98% or 99% homology) and having the same function as the PRRT2 involved in the embodiments of the present invention are also included in the present invention.
- Methods and tools for aligning sequence identity are also well known in the art, such as BLAST.
- Homology refers to the level of similarity (i.e., sequence similarity or identity) between two or more nucleic acids or polypeptides based on the percentage of identical positions.
- a polynucleotide sequence encoding PRRT2 or its variant protein can also be applied to the present invention.
- the term "coding gene" can be a polynucleotide that includes the protein, or a polynucleotide that also includes additional coding and/or non-coding sequences.
- the polynucleotide sequence encoding the PRRT2 protein is a polynucleotide sequence shown in SEQ ID NO: 1, 3 or 5.
- PRRT2 protein-rich transmembrane protein 2
- PRRT2 protein-rich transmembrane protein 2
- PRRT2 overexpression can promote the slow inactivation of sodium ion channels, but has no significant effect on the fast inactivation of sodium ion channels.
- PRRT2 has the characteristics of sodium ion channel state-dependent regulation. By enhancing the slow inactivation of sodium ion channels, it reduces the effective supply of cellular sodium ion channels under the state of continuous depolarization. This feature of PRRT2 is conducive to reducing the abnormal excitability of cells without affecting the normal excitatory activity of cells, so that it has good cell state selectivity.
- the inventors used adeno-associated virus as a gene delivery vector, selectively overexpressed PRRT2 protein in the excitatory neurons of the forebrain, and feedback-reduced the excitability of abnormally excited neurons.
- the trial found that selectively targeting PRRT2 therapy can effectively suppress epileptic seizures without interfering with daily behavior.
- the present invention provides a use of an up-regulator of PRRT2 that selectively acts on forebrain excitatory neurons, for preparing a drug for treating or preventing epilepsy.
- the "subject” described in the present invention includes but is not limited to: humans, non-human primates, and rodents (such as mice).
- epileptic seizures can be divided into different levels, including but not limited to: tonic-clonic, tonic, clonic, myoclonic, absence or atonic seizures.
- epileptic seizures can be divided into six levels, namely: Level 1: The mouse lies motionless on the ground, crawling on the belly; Level 2: sudden convulsions, sudden cessation of behavior, and the tail is raised; Level 3: muscle clonus, head twisting, hand convulsions; Level 4: tonic-clonic, falling to the ground and convulsions, jumping and running wildly; Level 5: falling to the ground and developing to rigid extension of limbs; Level 6: death.
- upregulators of PRRT2 acting selectively on forebrain excitatory neurons, can reduce the number, frequency, level and/or duration of epileptic seizures experienced by a subject by about 5%, about 10%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 99% or 100%, including all ranges and subranges therebetween.
- the "selective effect" is relative, for example, the effect on forebrain excitatory neurons is increased by about 5%, about 10%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 99% or 100%, relative to non-forebrain excitatory neurons, including all ranges and subranges therebetween.
- the "excitatory neuron” is a neuron that can release neurotransmitters such as glutamate, which can cause the connected postsynaptic neuron to become more likely to be excited.
- the “inhibitory neuron” is a neuron that can release neurotransmitters such as gamma-aminobutyric acid, which can cause the connected postsynaptic neuron to become less likely to be excited.
- the PRRT2 upregulator includes agonists, promoters, stimulants, etc., and these terms can be used interchangeably.
- the PRRT2 upregulator refers to any substance that can increase the activity of PRRT2, enhance the stability of PRRT2, upregulate the expression of PRRT2, increase the effective action time of PRRT2, or promote the transcription and translation of the PRRT2 gene. These substances can be used in the present invention as substances useful for upregulating PRRT2, and thus can be used to treat or prevent epilepsy.
- the PRRT2 up-regulator includes (but is not limited to): a construct, or an expression system formed by the construct (e.g., a viral (infection) system); the construct includes: a forebrain excitatory neuron-specific expression drive system, and a PRRT2 encoding gene driven by the drive system.
- the forebrain excitatory neuron-specific expression drive system includes a forebrain excitatory neuron-specific expression promoter.
- the forebrain excitatory neuron-specific expression promoter includes: Emx1 promoter and CaMKIIa promoter.
- the construct includes: a promoter, an element for DIO, a PRRT2 encoding A gene expression cassette (preferably, a PRRT2 encoding gene is reversely linked), a forebrain excitatory neuron-specific expression promoter-driven Cre recombinase encoding gene; preferably, the element used for DIO is a LoxP/Lox2272 element, and a DIO regulation method based on LoxP and Lox2272 is used to perform cre recombinase-dependent expression of PRRT2.
- the construct comprises:
- Construct 1 comprising: a promoter (such as but not limited to beta-actin promoter), LoxP/Lox2272, a PRRT2 encoding gene, LoxP/Lox2272 connected in sequence; the PRRT2 encoding gene is reversely connected;
- a promoter such as but not limited to beta-actin promoter
- LoxP/Lox2272 a promoter (such as but not limited to beta-actin promoter)
- PRRT2 encoding gene LoxP/Lox2272 connected in sequence
- the PRRT2 encoding gene is reversely connected
- Construct 2 a promoter specifically expressing forebrain excitatory neurons and a gene encoding cre recombinase (eg, Emx1 promoter drives the expression of the gene encoding cre recombinase).
- a promoter specifically expressing forebrain excitatory neurons and a gene encoding cre recombinase eg, Emx1 promoter drives the expression of the gene encoding cre recombinase.
- the PRRT2 polynucleotide sequence can be inserted into a recombinant expression vector, so that it can be transferred into cells to overexpress PRRT2.
- any plasmid and vector can be used in the present invention.
- Methods well known to those skilled in the art can be used to construct expression vectors containing the DNA sequence of PRRT2 and appropriate transcription/translation control signals. These methods include in vitro recombinant DNA technology, DNA synthesis technology, in vivo recombination technology, etc.
- An important feature of the expression vector is that it usually contains a replication origin, a promoter, a marker gene and a translation control element.
- the expression vector includes: a viral vector, a non-viral vector; preferably, the viral vector includes (but is not limited to): an adeno-associated virus (AAV) vector, a lentiviral vector, an adenoviral vector, a retroviral vector, etc.
- AAV adeno-associated virus
- the viral vector is an AAV vector.
- AAV virion or "AAV virus” or “AAV virus particle” or “AAV vector particle” or “AAV vector” refers to a viral particle comprising at least one AAV capsid polypeptide and a polynucleotide (such as a PRRT2 polynucleotide sequence).
- AAV vectors are usually named according to the name (serotype) of the capsid polypeptide.
- the AAV vectors include (but are not limited to): PHP.eB serotype AAV vector, Cap-B10 serotype AAV vector.
- the gene encoding PRRT2 can be cloned into an appropriate vector (such as a conventional prokaryotic or eukaryotic expression vector, or a viral vector such as a herpes virus vector or an adeno-associated virus vector) by conventional methods, and the vector is introduced into a cell that can express the PRRT2, so that the cell expresses PRRT2.
- an appropriate vector such as a conventional prokaryotic or eukaryotic expression vector, or a viral vector such as a herpes virus vector or an adeno-associated virus vector
- the expression of PRRT2 can be achieved by introducing an appropriate amount of the cells into an appropriate part of the subject's body.
- the vector is administered by intraorbital injection (such as orbital vein injection) so that it selectively acts on excitatory neurons in the forebrain and reduces the influence of the blood-brain barrier.
- intracranial injection intrathecal (spinal cord) injection
- intrathecal (cerebral cistern) injection intracerebral injection
- intraventricular injection direct injection into the epileptic lesion in the hippocampus, and direct injection into the epileptic lesion in the temporal lobe.
- PHP.eB serotype AAV virus as the delivery vector of PRRT2 gene, which has good blood-brain barrier penetration ability.
- vectors that help to efficiently deliver PRRT2 gene to neurons in the brain such as Cap-B10 serotype AAV virus, etc.
- vectors that help to efficiently deliver PRRT2 gene to excitatory neurons in the forebrain can be applied to this experiment.
- PRRT2 after overexpression to produce PRRT2, methods well known to those skilled in the art can be used to detect whether the overexpression of PRRT2 is successful.
- primers that specifically amplify PRRT2 can be used; or probes that specifically recognize PRRT2 can be used to determine the presence or absence of the PRRT2 gene;
- antibodies or ligands that specifically bind to the protein encoded by PRRT2 can be used to determine the expression of the PRRT2 protein, for example, using an anti-PRRT2 antibody to bind to the PRRT2 protein, and then using a secondary antibody with a detection signal (preferably a fluorescent label) to bind to the anti-PRRT2 antibody, and judging by the presence or absence of the detection signal, or the strength of the detection signal.
- a detection signal preferably a fluorescent label
- the present invention also provides a method for treating or preventing epilepsy, comprising selectively administering an effective amount of PRRT2 or an up-regulator thereof (such as an expression vector overexpressing the PRRT2) to excitatory neurons of the forebrain of a subject.
- an effective amount of PRRT2 or an up-regulator thereof such as an expression vector overexpressing the PRRT2
- treat and “prevent” generally refer to the use of drugs or methods to reduce, eliminate or prevent the symptoms of a disease, and include achieving a therapeutic benefit and/or a prophylactic benefit.
- a therapeutic benefit refers to slowing the progression of the condition or symptoms of the condition being treated, halting the progression, reversing the progression, or eradicating or ameliorating the symptoms.
- a prophylactic benefit includes reducing the risk of a condition, delaying the progression of a condition, or reducing the likelihood of a condition occurring.
- the PRRT2 or its up-regulator can be administered to the subject by various methods well known in the art.
- gene therapy can be used, such as delivering an expression unit (such as an expression vector or virus, etc.) carrying the PRRT2 gene to the target through a certain route, and causing it to express (preferably overexpress) active PRRT2; or, the expression of endogenous PRRT2 in forebrain excitatory neurons can be enhanced by in vivo gene editing (such as CRISPR-Cas9 gene editing method), thereby achieving the purpose of treating or preventing epilepsy.
- expression unit such as an expression vector or virus, etc.
- active PRRT2 preferably overexpress
- endogenous PRRT2 in forebrain excitatory neurons can be enhanced by in vivo gene editing (such as CRISPR-Cas9 gene editing method), thereby achieving the purpose of treating or preventing epilepsy.
- the term "effective amount” refers to an amount that can achieve a desired result (e.g., a preventive or therapeutic result).
- the effective amount of PRRT2 or its up-regulator described in the present invention may vary depending on the mode of administration and the severity of the disease to be treated. The selection of the preferred effective amount can be determined by a person of ordinary skill in the art based on various factors (e.g., through clinical trials). The factors include, but are not limited to: pharmacokinetic parameters of the PRRT2 or its up-regulator such as bioavailability, metabolism, half-life, etc.; the severity of the disease to be treated by the patient, the patient's weight, the patient's immune status, the route of administration, etc. For example, depending on the urgency of the treatment situation, several divided doses may be given per day, or the dose may be reduced proportionally.
- Zebrafish PRRT2 plasmid (laboratory constructed, cloned from adult zebrafish brain tissue cDNA library).
- Lipofectamine 3000 transfection kit (ThermoFisher Scientific, #L3000001).
- Nav1.2 stably transfected cell line (Beijing Ice-Eye Biotechnology Co., Ltd., #ICE-Nav1.2-HEK).
- AAV-CAG-DIO-mCherry plasmid ( Figure 16) (provided by the Gene Editing Platform of Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences).
- Anti-PRRT2 antibody (Wiiget Biotech, #Rp3246-a).
- the AAV virus used in the present invention is produced and provided by the gene editing platform of the Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences:
- Pentylenetetrazol (Sigma-Aldrich, #P6500-25G).
- C57BL/6J mice were purchased from Shanghai Slake Laboratory Animal Co., Ltd.
- Emx1-cre mice name: B6.129S2-Emx1tm1(cre)Krj/J, from Jackson Lab, catalog number: JAX catalog number #005628.
- Nkx2.1-cre mice name: C57BL/6J-Tg(Nkx2-1-cre)2Sand/J, from Jackson Lab, catalog number: JAX catalog number #008661.
- mice Three to six mice were housed in each cage with free access to food and water in the cages under a 12-h light/dark cycle, 22-23°C, and 50-60% humidity.
- AAV virus stored at -80 degrees Celsius and thaw it on ice. Dilute the AAV virus to the injection concentration (2 ⁇ 10 12 vg/mL, vg: AAV vector genomes) with pre-cooled sterile PBS. Equilibrate the AAV virus solution to room temperature before injection and add 0.3 Aspirate 50 ⁇ L of AAV virus solution into a mL insulin needle for injection.
- mice were anesthetized by injecting sodium pentobarbital solution (90 mg/kg, intraperitoneal injection). After the mouse was anesthetized, the mouse head was fixed with fingers, and the mouse artery was avoided to be pressed. The eye skin was stretched to fully expose the eyeball. The insulin needle was inserted from the corner of the eye at a 20° angle, and the eyeball was avoided when entering the orbit. The needle tip entered the orbit to a depth of 2 mm to reach the orbital venous plexus, and 50 ⁇ L was slowly injected. After the injection, the needle was slowly withdrawn and excess fluid and blood in the corner of the eye were wiped off. The same injection was performed in the orbital vein on the other side, and a total titer of 2 ⁇ 10 11 vg of AAV virus was injected on both sides.
- sodium pentobarbital solution 90 mg/kg, intraperitoneal injection
- mice were placed back in the cages and sent back to the feeding room after they woke up.
- Electrode implantation surgery was performed two weeks after viral expression, and epilepsy evaluation experiments were performed three weeks after viral expression.
- mice were first anesthetized with 4% isoflurane, and then the mouse heads were fixed to the stereotaxic adapter.
- the isoflurane concentration was adjusted to 1.5% during the entire subsequent surgery to keep the mice in a state of continuous anesthesia.
- chlortetracycline eye ointment was used to protect the mouse eyes to avoid retinal damage caused by dryness.
- the hair on the head of the mouse was removed with a depilatory cream, and the head skin was cut along the midline to expose the skull surface.
- the surgical incision and skull surface were treated with a 2% hydrogen peroxide solution, and the connective tissue on the skull surface was scraped off.
- the skin around the exposed skull was fixed with tissue glue (Vetbond, 3M Deutschland Gmbh), and the membrane system on the skull surface was cleaned again.
- the electrode implantation position was marked by stereotaxic method, with coordinates AP-0.8mm, ML ⁇ 1.1mm.
- the skull surface was covered with a light-cured self-etching adhesive (3M ESPE Single Bound universal) to form a solidified layer.
- a small handheld cranial drill was used to open a hole at the implantation position mark, with a circular hole diameter of approximately 0.6mm. Carefully clean the bone debris at the opening and keep the surface of the cerebral cortex moist.
- two sterile ECoG recording electrodes (0.5 mm in diameter) were vertically inserted into the cerebral cortex at a depth of 0.35 mm (with the surface of the cerebral cortex as a reference) along the skull opening, and the recording electrodes and electrode interface components were fixed with dental cement.
- a lightweight titanium alloy sheet (30 mm ⁇ 2 mm ⁇ 1 mm) was additionally fixed with dental cement at the position of the mouse head near the electrode interface.
- the two metal wire electrodes used to record electromyographic signals extended from the bottom of the electrode interface component to the neck of the mouse, and the exposed ends of the 1 mm electrodes were inserted into the muscle layers on both sides of the neck, and the neck skin was sutured and disinfected after fixation.
- the mice recovered for one week after surgery.
- the mouse Before ECoG/EMG recording in the awake state, the mouse was fixed to the recording platform using a metal plate on the mouse's head, and the mouse was allowed to adapt to this head-fixed state for 30 minutes.
- the ECoG and EMG signals were amplified 100 times by a differential amplifier (Model 3000, A-M system), and then band-filtered (ECoG 1-300Hz, EMG 10-1000Hz) and collected (1000Hz) and recorded by a digital-to-analog converter (Digidata 1332A, Molecular Devices).
- mice Before the test, 3.5 mg/mL (Nkx2.1-cre mouse experiment) or 4.5 mg/mL (Emx1-cre mouse experiment) of pentylenetetrazol solution was prepared with normal saline. According to the weight of mice, pentylenetetrazol was injected intraperitoneally at a dose of 35 mg/kg or 45 mg/kg. Immediately after the injection, the mice were placed in a behavior box for video recording to observe the epileptic seizures of the mice within 30 minutes after administration.
- Level 1 The mouse lies motionless on the ground, crawling on its belly;
- Level 2 Sudden convulsions, sudden cessation of behavior, and raised tail;
- Grade 3 Myoclonus, head twisting, hand twitching
- Grade 4 Tonic-clonic, falling to the ground and twitching, jumping and running wildly;
- mice were deeply anesthetized with sodium pentobarbital, 10 mL of room temperature phosphate buffer solution (PBS) was perfused through the heart to flush the blood, and then 10 mL of 4% polyformaldehyde solution (PFA) at 4°C was perfused for tissue fixation.
- PBS room temperature phosphate buffer solution
- PFA polyformaldehyde solution
- the mouse brain tissue was dissected and removed, placed in a 4% paraformaldehyde solution and fixed overnight. The brain tissue was then placed in PBS containing 30% sucrose for dehydration for 2 days, and the sucrose solution was replaced once after 24 hours.
- the brain tissue was sagittal sliced at -20°C with a thickness of 25 ⁇ m using a freezing microtome (CM1950, Leica), and the brain slices were collected in a PBS solution and stored at 4°C. Conventional immunohistochemistry was performed using anti-PRRT2 antibodies.
- Zebrafish Prrt2 gene protein coding region, CDS sequence (SEQ ID NO: 5):
- Example 1 Verification of the effect of overexpression of PRRT2 on slow inactivation of sodium channels
- the cDNA sequence of the mouse PRRT2 protein coding region ( Figures 1 and 2) was inserted into the multiple cloning site region of the pCAG2IG plasmid (i.e., the EGFP control plasmid, Figure 3) to construct the mouse PRRT2 overexpression plasmid pCAG-mouse PRRT2(HA)-IRES-EGFP ( Figure 4).
- HEK293 cells stably transfected with Nav1.2 PRRT2 overexpression plasmid or control plasmid was transfected using Lipo3000 reagent. After expression at 37°C for 24 hours, the cells were plated on round glass slides and continued to express for 12 hours.
- Sodium channel currents were recorded by whole-cell recording (Figure 11A).
- a capillary glass tube (BF150-86-10, Sutter Instruments) was pulled into a recording electrode using a microelectrode puller (P97, Sutter Instruments). Electrode. Under an inverted microscope (MF53, Micro-shot), the microelectrode manipulator (MP225, Butter Instrument) was manipulated to contact the recording electrode to the cell surface, and negative pressure was applied to form a G ⁇ seal. After the G ⁇ seal was formed, fast capacitance compensation was performed, and then negative pressure was continued to be applied to break the cell membrane and form a whole-cell recording mode. Then slow capacitance compensation was performed and membrane capacitance and series resistance were recorded. All electrophysiological experiments were performed at room temperature. The experimental data were collected by an EPC-10 amplifier (HEKA) and stored in the PatchMaster (HEKA) software.
- HEKA EPC-10 amplifier
- HEKA PatchMaster
- the testing scheme for the effect of human PRRT2 and zebrafish PRRT2 on the slow inactivation of sodium channels is similar to the testing scheme for mouse PRRT2 mentioned above, but in the process of detecting the recovery of sodium channels from slow inactivation, the inventors used a continuous testing scheme instead of the repeated scanning test method to improve the success rate of cell electrophysiological recording.
- the initial clamping voltage of the cell is -120mV, at which time the sodium ion channel is in a closed state or a resting state.
- the clamping voltage rises from -120mV to 0mV, the cell depolarizes, the sodium ion channel opens, and extracellular sodium ions rapidly flow inward to form a sodium current ( Figure 11B).
- the sodium ion channel begins to enter an inactivated state (fast inactivation), the channel closes, and the sodium current decays ( Figures 11B and 11C).
- the sodium ion channel enters a fast inactivation state within a short depolarization period, and can be restored to an activatable resting state during a hyperpolarization process of ten to tens of milliseconds ( Figures 12A and 12B).
- sodium channels In addition to the fast inactivation mechanism, sodium channels also have the characteristics of slow inactivation. It takes longer for sodium channels to enter the slow inactivation state or recover from the slow inactivation state than the fast inactivation state, usually hundreds of milliseconds to tens of seconds. According to the above rules, after the depolarization process is completed, a brief hyperpolarization of 10 milliseconds can restore the fast inactivation state of sodium channels, thereby effectively separating the slow inactivation component of the sodium channel (Figure 13A).
- mice PRRT2 promoted the speed at which sodium ion channels entered the slow inactivation state during depolarization ( FIG. 13B ), and prolonged the time it took for sodium ion channels to recover from the slow inactivation state to the activatable resting state ( FIG. 14A and FIG. 14B ).
- PRRT2 is conserved in many species. Surprisingly, in the previous work, the inventors found that the inhibitory effect of zebrafish PRRT2 on the recovery of sodium channel slow inactivation (compared with mouse and human PRRT2, zebrafish PRRT2 overexpression makes the recovery time of sodium channel slow inactivation longer, which can be understood as having a stronger effect on sodium channel slow inactivation) was significantly stronger than that of human and mouse PRRT2.
- Figure 15C is the comparison result of zebrafish, human and mouse PRRT2.
- PRRT2 overexpression can promote the slow inactivation of sodium channels, but has no significant effect on the fast inactivation of sodium channels. Therefore, PRRT2 has the characteristics of sodium channel state-dependent regulation, and is more inclined to reduce the effective supply of cell sodium channels under continuous depolarization. This feature of PRRT2 allows it to reduce the effective supply of cell sodium channels under continuous depolarization without affecting the normal excitatory activity of cells. Reduce the abnormal excitability of cells, giving them good cell state selectivity.
- Example 2 Overexpression of PRRT2 in forebrain excitatory neurons to intervene in epileptic seizures
- anti-PRRT2 antibody and anti-HA tag antibody used to perform immunofluorescence staining on brain slice samples.
- Anti-PRRT2 antibody is used to detect PRRT2 protein including endogenous and overexpressed, while anti-HA antibody can only detect overexpressed PRRT2 protein with HA tag.
- the brain slices were rinsed three times with PBS solution for 10 minutes each time, and then incubated with blocking solution containing 0.3% Triton X-100 and 5% bovine serum protein (BSA; dissolved in PBS) at room temperature for 1 hour. After the blocking was completed, the primary antibody was incubated, and the brain slice samples were incubated in anti-PRRT2 antibody (1:500, rabbit) solution at 4°C overnight. The brain slices were rinsed three times with PBS solution and then incubated with secondary antibodies. The brain slice samples were incubated in fluorescent secondary antibodies (1:2000, donkey anti-rabbit-647nm, donkey anti-rat 488nm) and Hoechst (1:5000) solutions at room temperature for 2 hours.
- BSA bovine serum protein
- the brain slices were attached to the slides, and after drying, PBS solution containing 80% glycerol was added and the slides were sealed. The images were observed and photographed under a 10x objective lens using a fluorescence microscope (VS-120, Olympus).
- Adeno-associated virus is the most commonly used vector for gene delivery in vivo.
- PHP.eB serotype AAV virus was selected as the delivery vector of PRRT2 gene.
- the experiment showed that it has good blood-brain barrier penetration ability and is conducive to the expression of PRRT2 gene in the brain.
- PRRT2 protein Overexpression of PRRT2 protein was achieved by Cre recombinase-dependent method, by setting up DIO system (Loxp/Lox2272) in viral expression vector to achieve Cre recombinase-dependent expression ( Figure 17, pAAV-CAG-DIO-mouse PRRT2-HA). Indirectly relying on Emx1 promoter (forebrain excitatory neuron-specific expression promoter)-driven Cre recombinase expression to achieve specific expression of PRRT2 protein in forebrain excitatory neurons.
- Emx1 promoter forebrain excitatory neuron-specific expression promoter
- the inventors used a mouse strain (emx1-cre) that specifically expresses cre recombinase.
- the expression of cre recombinase depends on the Emx1 promoter.
- mice in the PRRT2 overexpression group showed stronger anti-epileptic ability in the PTZ-induced epilepsy model ( FIG. 19A ).
- mice in the group overexpressing PRRT2 in forebrain excitatory neurons after PTZ injection were significantly weaker than those of mice in the control group ( FIGS. 20A and 20B ).
- mice whose excitatory neurons overexpressed PRRT2 showed significantly higher performance in the open field test, wheel running test, and social interaction test. No significant behavioral abnormalities were observed in the experiments (Figure 21A-21C).
- Emx1-cre mice were injected intraorbitally with a total of 2 ⁇ 10 11 vg of AAV-PHP.eB-CAG-DIO-mCherry virus (control) or AAV-PHP.eB-CAG-DIO-zebra PRRT2 virus.
- Nkx2.1-cre mice the expression of cre recombinase depends on the Nkx2.1 gene promoter, which drives the selective expression of cre recombinase in forebrain inhibitory neurons (i.e., ⁇ -aminobutyric acid neurons).
- the inventors used Nkx2.1-cre mice and adopted a strategy of adeno-associated virus delivery to inject a total of 2 ⁇ 10 11 vg of AAV-PHP.eB-CAG-DIO-mouse PRRT2-HA virus into the orbital vein to overexpress PRRT2 in forebrain inhibitory neurons ( Figures 22A and 22B).
- mice in the inhibitory neuron overexpression PRRT2 group showed epilepsy susceptibility in the PTZ-induced epilepsy model ( Figure 22D and Figure 22E).
- mice in the inhibitory neuron overexpression PRRT2 group had higher epileptic seizure levels than mice in the control group ( Figure 22D and Figure 22E).
- the results of EEG recording also showed that the epileptic EEG signals of mice in the forebrain inhibitory neuron overexpression PRRT2 group were significantly stronger than those of mice in the control group after injection of subthreshold doses of PTZ ( Figure 22F and Figure 22G).
- mice were susceptible to epilepsy by selectively overexpressing PRRT2 in astrocytes. Therefore, overexpression of PRRT2 in non-forebrain excitatory neurons leads to adverse reactions.
- overexpression of PRRT2 protein in forebrain excitatory neurons can effectively enhance the anti-epileptic ability of mice without affecting normal movement and social behavior, and this selective expression strategy avoids the epilepsy susceptibility caused by overexpression of PRRT2 protein in forebrain inhibitory neurons.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Chemical & Material Sciences (AREA)
- Epidemiology (AREA)
- Engineering & Computer Science (AREA)
- Marine Sciences & Fisheries (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Gastroenterology & Hepatology (AREA)
- Zoology (AREA)
- Immunology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Neurosurgery (AREA)
- Biomedical Technology (AREA)
- Neurology (AREA)
- Pain & Pain Management (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
本发明属于生物医药领域,更具体地,本发明涉及作用于前脑兴奋性神经元的PRRT2及其上调剂的应用。The present invention belongs to the field of biomedicine, and more specifically, the present invention relates to the application of PRRT2 and its up-regulator acting on forebrain excitatory neurons.
癫痫是较为典型的由兴奋-抑制失衡所致的中枢神经系统疾病,临床表现主要有惊厥、肌痉挛、强直性抽搐、失张力以及失神等,并伴有短暂的意识丧失,发作时患者脑电呈现癫痫样放电特征。癫痫发作具有突然性和反复发作的特性,这给患者的身体和心理健康以及正常社会活动造成了严重影响。癫痫的病因较为多样,目前已知的致病因素包括发育异常、遗传突变、颅脑损伤、脑血管疾病和炎症等。除了少部分自限性癫痫之外,大部分癫痫患者需要通过治疗实现对癫痫的控制。目前在临床使用的治疗方法主要包括药物治疗、手术切除局部病灶、生酮饮食、迷走神经刺激以及深部脑刺激等,其中超过三分之二的癫痫患者主要通过抗癫痫药物进行癫痫控制。尽管从上世纪50年代至今已陆续推出了针对不同靶点的30余款抗癫痫药物,形成了较为有效的临床治疗方案,但仍有约30%的难治性癫痫患者不能从这些药物或药物组合中获益。另外,对现有药物有良好响应的部分癫痫患者也依然面临长期用药期间药物不良反应以及癫痫无法完全控制的困扰。以上未能满足的医疗需求是新机制研究以及新疗法研发的重要驱动力。Epilepsy is a typical central nervous system disease caused by excitation-inhibition imbalance. The main clinical manifestations include convulsions, muscle spasms, tonic convulsions, atonia and absence, accompanied by a brief loss of consciousness. During the attack, the patient's EEG shows epileptic discharge characteristics. Epileptic seizures are characterized by suddenness and recurrence, which has a serious impact on the patient's physical and mental health and normal social activities. The causes of epilepsy are relatively diverse. Currently known pathogenic factors include developmental abnormalities, genetic mutations, craniocerebral injury, cerebrovascular disease and inflammation. Except for a small number of self-limited epilepsy, most epilepsy patients need to achieve control of epilepsy through treatment. The treatment methods currently used in clinical practice mainly include drug therapy, surgical resection of local lesions, ketogenic diet, vagus nerve stimulation and deep brain stimulation. Among them, more than two-thirds of epilepsy patients mainly use anti-epileptic drugs for epilepsy control. Although more than 30 anti-epileptic drugs targeting different targets have been launched since the 1950s, forming a relatively effective clinical treatment plan, about 30% of patients with refractory epilepsy still cannot benefit from these drugs or drug combinations. In addition, some epilepsy patients who respond well to existing drugs still face the problem of adverse drug reactions during long-term medication and the inability to fully control epilepsy. The above unmet medical needs are an important driving force for the research of new mechanisms and the development of new therapies.
神经网络的稳态平衡依赖于兴奋性和抑制型神经元相互协作,而癫痫主要表现为神经网络稳态失衡导致的大脑异常同步性兴奋。因此,理想的癫痫干预策略是通过药物或其它治疗手段选择性的降低兴奋性神经元活动或者提高抑制型神经元活动,以此纠正神经网络的异常兴奋并使其回归正常的稳态;同时,干预方案应尽量避免或减少对非癫痫相关神经网络的干扰,如运动、觉醒、认知和情感环路等,以此降低长期用药时的不良反应。The homeostatic balance of neural networks depends on the cooperation between excitatory and inhibitory neurons, and epilepsy is mainly manifested as abnormal synchronous excitement of the brain caused by imbalance of neural network homeostasis. Therefore, the ideal epilepsy intervention strategy is to selectively reduce the activity of excitatory neurons or increase the activity of inhibitory neurons through drugs or other treatments, so as to correct the abnormal excitement of the neural network and return it to normal homeostasis; at the same time, the intervention plan should try to avoid or reduce interference with non-epilepsy-related neural networks, such as movement, arousal, cognition and emotional circuits, so as to reduce adverse reactions during long-term medication.
电压门控型钠离子通道(以下简称钠离子通道或Nav)是脑内神经元动作电位产生和脉冲传导所依赖的重要分子,是神经组织兴奋性的基础。在癫痫治疗领域,钠离子通道是抗癫痫药物的重要靶点。目前批准临床使用的抗癫痫药物中有超过四分之一的药物以此类通道为靶点,包括苯妥英钠(Sodium Phenytoin)、卡马西平(Carbamazepine)、奥卡西平(Oxcarbazepine)和拉考沙胺(Lacosamide)等。这些药物的作用机理是通过调节钠离子通道活性,降低神经网络的兴奋性,进而减少或阻止相关类型的癫痫发作。尽管钠离子通道为靶点的抗癫痫药物在癫痫治疗中已获得广泛应用,但其中枢不良反应(如头晕、嗜睡、共济失调和易怒等)仍未得到有效解决。Voltage-gated sodium channels (hereinafter referred to as sodium channels or Nav) are important molecules that the generation of action potentials and impulse conduction of neurons in the brain rely on, and are the basis of the excitability of neural tissue. In the field of epilepsy treatment, sodium channels are important targets for anti-epileptic drugs. More than a quarter of the anti-epileptic drugs currently approved for clinical use target such channels, including sodium phenytoin, carbamazepine, oxcarbazepine, and lacosamide. The mechanism of action of these drugs is to reduce the excitability of neural networks by regulating the activity of sodium channels, thereby reducing or preventing related types of epileptic seizures. Although anti-epileptic drugs targeting sodium channels have been widely used in the treatment of epilepsy, their central adverse reactions (such as dizziness, drowsiness, ataxia, and irritability) have not been effectively resolved.
缺乏细胞类型选择性以及细胞状态选择性不佳是目前小分子抗癫痫药物产生中枢不良反应的主要原因。在中枢神经系统,电压门控型钠离子通道主要有四个亚型(Nav1.1、Nav1.2、 Nav1.3和Nav1.6),它们分布于发育不同阶段的不同类型神经元内。例如,在与癫痫直接相关的前脑,Nav1.2和Nav1.6主要分布于以谷氨酸(Glutamate)为主要神经递质的兴奋性神经元,而Nav1.1则主要分布于以γ-氨基丁酸(GABA)为主要神经递质的抑制型中间神经元;在与运动调节相关的小脑皮层,Nav1.1、Nav1.2和Nav1.6分别或组合表达于GABA能中间神经元,颗粒细胞和浦肯野神经元。以钠离子通道为靶点的抗癫痫药物,由于其普遍缺乏钠离子通道成员间的选择性,在其进入中枢后会作用于非癫痫相关的神经细胞群,引起中枢不良反应。尤其值得关注的是,以钠离子通道为靶点的抗癫痫药物对前脑抑制型中间神经元的抑制,在一定程度上会降低大脑抑制性神经网络的功能,部分抵消抗癫痫药物对兴奋性神经网络的抑制功效,弱化其抗癫痫疗效,在某种情况下(如德拉韦综合征,Dravet syndrome)甚至还会恶化癫痫病情。Lack of cell type selectivity and poor cell state selectivity are the main reasons for the adverse reactions of small molecule anti-epileptic drugs. In the central nervous system, there are four main subtypes of voltage-gated sodium channels (Nav1.1, Nav1.2, Nav1.3 and Nav1.6), which are distributed in different types of neurons at different stages of development. For example, in the forebrain directly related to epilepsy, Nav1.2 and Nav1.6 are mainly distributed in excitatory neurons with glutamate as the main neurotransmitter, while Nav1.1 is mainly distributed in inhibitory interneurons with γ-aminobutyric acid (GABA) as the main neurotransmitter; in the cerebellar cortex related to motor regulation, Nav1.1, Nav1.2 and Nav1.6 are expressed separately or in combination in GABAergic interneurons, granule cells and Purkinje neurons. Antiepileptic drugs that target sodium channels generally lack selectivity among sodium channel members. After entering the central nervous system, they will act on non-epilepsy-related neural cell groups and cause adverse central reactions. It is particularly noteworthy that the inhibition of forebrain inhibitory interneurons by antiepileptic drugs targeting sodium ion channels will, to a certain extent, reduce the function of the brain's inhibitory neural network, partially offset the inhibitory effect of antiepileptic drugs on excitatory neural networks, weaken their antiepileptic efficacy, and in some cases (such as Dravet syndrome) may even worsen epilepsy.
除了缺乏细胞类型选择性导致的不良反应,药物缺乏有效的细胞状态选择性也是其产生不良反应的重要原因。细胞状态选择性是指抗癫痫药物或治疗手段能够作用于癫痫发作时呈现异常兴奋状态的细胞,而不干扰非癫痫状态下同类细胞的正常功能。考虑到癫痫治疗往往需要长期使用药物,对细胞癫痫状态选择性不佳的药物,常会导致患者在日常用药期间(非癫痫状态)感到不适,影响他们的日常生活与工作,甚至导致患者用药中断而使癫痫复发。In addition to the adverse reactions caused by the lack of cell type selectivity, the lack of effective cell state selectivity of the drug is also an important reason for its adverse reactions. Cell state selectivity refers to the ability of anti-epileptic drugs or treatments to act on cells that are abnormally excited during epileptic seizures without interfering with the normal functions of similar cells in the non-epileptic state. Considering that epilepsy treatment often requires long-term use of drugs, drugs with poor selectivity for cell epileptic states often cause patients to feel uncomfortable during daily medication (non-epileptic state), affecting their daily life and work, and even causing patients to interrupt medication and cause recurrence of epilepsy.
因此,本领域亟需以钠离子通道为靶点,研发具有细胞类型选择性以及细胞状态选择性的药物,从而增强癫痫的干预效果并减少副作用,促进癫痫的精准化治疗。Therefore, there is an urgent need in this field to develop drugs with cell type selectivity and cell state selectivity using sodium ion channels as targets, so as to enhance the intervention effect of epilepsy and reduce side effects, and promote the precision treatment of epilepsy.
发明内容Summary of the invention
本发明的目的在于提供一种选择性作用于前脑兴奋性神经元的富含脯氨酸的跨膜蛋白2(proline-rich transmembrane protein 2,PRRT2)的上调剂的用途,用于制备治疗或预防癫痫的药物。The purpose of the present invention is to provide a use of an upregulator of proline-rich transmembrane protein 2 (PRRT2) that selectively acts on forebrain excitatory neurons, for the preparation of drugs for treating or preventing epilepsy.
在本发明的第一方面,提供一种选择性作用于前脑兴奋性神经元的PRRT2的上调剂的用途,用于制备治疗或预防癫痫的药物。In a first aspect of the present invention, there is provided a use of an upregulator of PRRT2 that selectively acts on forebrain excitatory neurons for preparing a drug for treating or preventing epilepsy.
在一种或多种实施方式中,所述上调为显著性的上调、促进、增加或提高;例如上调、促进、增加或提高10%、20%、30%或更高。In one or more embodiments, the upregulation is a significant upregulation, promotion, increase or enhancement; for example, an upregulation, promotion, increase or enhancement of 10%, 20%, 30% or more.
在一种或多种实施方式中,所述选择性作用于前脑兴奋性神经元包括:选择性作用于钠离子通道(较佳为Nav1.2和Nav1.6)慢失活,和/或选择性作用于细胞异常兴奋性。In one or more embodiments, the selective action on forebrain excitatory neurons includes: selectively acting on the slow inactivation of sodium ion channels (preferably Nav1.2 and Nav1.6), and/or selectively acting on abnormal cell excitability.
在一种或多种实施方式中,所述治疗或预防癫痫包括:降低癫痫发作的次数、频率、水平和/或持续时间。In one or more embodiments, the treating or preventing epilepsy comprises reducing the number, frequency, level and/or duration of epileptic seizures.
在一种或多种实施方式中,所述治疗或预防癫痫不影响受试者的日常行为。In one or more embodiments, the treating or preventing epilepsy does not affect the subject's daily activities.
在一种或多种实施方式中,通过小鼠行为学实验评估癫痫发作的次数、频率、水平和/或持续时间。较佳地,所述行为学实验包括脑电和行为观察。In one or more embodiments, the number, frequency, level and/or duration of epileptic seizures are assessed by behavioral experiments in mice. Preferably, the behavioral experiments include EEG and behavioral observations.
在一种或多种实施方式中,通过小鼠行为学实验评估日常行为,所述行为学实验包括但 不限于:旷场测试、转轮测试、社交行为测试。In one or more embodiments, daily behavior is assessed by a mouse behavioral experiment, wherein the behavioral experiment includes but Not limited to: open field test, wheel test, social behavior test.
在一种或多种实施方式中,所述用途的受试者包括:人、非人灵长类动物和啮齿类动物。In one or more embodiments, the subjects of the use include: humans, non-human primates and rodents.
在一种或多种实施方式中,所述选择性作用于前脑兴奋性神经元的PRRT2的上调剂为构建体、或该构建体形成的表达系统(例如病毒(感染)系统);所述构建体包括:表达驱动系统,以及由该驱动系统驱动表达的PRRT2编码基因。In one or more embodiments, the upregulator of PRRT2 that selectively acts on forebrain excitatory neurons is a construct, or an expression system formed by the construct (e.g., a viral (infection) system); the construct includes: an expression drive system, and a PRRT2 encoding gene whose expression is driven by the drive system.
在一种或多种实施方式中,所述表达驱动系统包括单一、或组合形式的前脑兴奋性神经元特异性表达驱动系统。In one or more embodiments, the expression driving system comprises a single or combined forebrain excitatory neuron-specific expression driving system.
在一种或多种实施方式中,所述前脑兴奋性神经元特异性表达驱动系统包括单一形式的前脑谷氨酸能神经元特异性启动子,包括(但不限于):CaMKIIa启动子;单一形式的启动子用于直接驱动PRRT2的表达。In one or more embodiments, the forebrain excitatory neuron-specific expression driving system includes a single form of forebrain glutamatergic neuron-specific promoter, including (but not limited to): CaMKIIa promoter; a single form of promoter is used to directly drive the expression of PRRT2.
在一种或多种实施方式中,所述前脑兴奋性神经元特异性表达驱动系统包括组合形式的前脑谷氨酸能神经元特异性启动子,包括(但不限于):用于驱动Cre重组酶表达的前脑兴奋性神经元特异性启动子(如Emx1启动子或CaMKIIa启动子)和用于驱动PRRT2表达的强效启动子(如但不限于beta-actin promoter(CAG)启动子);组合形式的表达驱动系统用于兼顾前脑兴奋性神经元特异性和高效表达特性。In one or more embodiments, the forebrain excitatory neuron-specific expression driving system includes a combination of forebrain glutamatergic neuron-specific promoters, including (but not limited to): a forebrain excitatory neuron-specific promoter for driving Cre recombinase expression (such as Emx1 promoter or CaMKIIa promoter) and a strong promoter for driving PRRT2 expression (such as but not limited to beta-actin promoter (CAG) promoter); the combined expression driving system is used to take into account both forebrain excitatory neuron specificity and high-efficiency expression characteristics.
在一种或多种实施方式中,所述构建体包括依赖于Cre重组酶的操作性基因表达调控元件,如(但不限于)Double-floxed inverse orientation,DIO);较佳地,所述用于DIO的元件为LoxP/Lox2272,利用基于LoxP和Lox2272的特定识别序列进行Cre重组酶依赖的PRRT2的表达。In one or more embodiments, the construct includes an operational gene expression regulatory element that depends on Cre recombinase, such as (but not limited to) Double-floxed inverse orientation, DIO); preferably, the element for DIO is LoxP/Lox2272, and the Cre recombinase-dependent expression of PRRT2 is performed using specific recognition sequences based on LoxP and Lox2272.
在一种或多种实施方式中,所述构建体包括:构建体1,包括顺序连接的:启动子(如但不限于beta-actin promoter),LoxP/Lox2272,PRRT2编码基因,LoxP/Lox2272;所述PRRT2编码基因反向连接于两对LoxP/Lox2272序列之间;构建体2:包括顺序连接的:前脑兴奋性神经元特异性表达启动子,Cre重组酶编码基因(如Emx1启动子驱动Cre重组酶编码基因的表达)。In one or more embodiments, the construct includes: construct 1, including sequentially connected: a promoter (such as but not limited to beta-actin promoter), LoxP/Lox2272, a PRRT2 encoding gene, LoxP/Lox2272; the PRRT2 encoding gene is reversely connected between two pairs of LoxP/Lox2272 sequences; construct 2: including sequentially connected: a forebrain excitatory neuron-specific expression promoter, a Cre recombinase encoding gene (such as an Emx1 promoter driving the expression of the Cre recombinase encoding gene).
在一种或多种实施方式中,通过前脑兴奋性神经元特异性表达启动子驱动Cre重组酶编码基因表达;Cre重组酶作用于LoxP/Lox2272,使得反向的PRRT2编码基因转变成正向连接,由其启动子驱动表达。In one or more embodiments, the expression of the gene encoding Cre recombinase is driven by a forebrain excitatory neuron-specific expression promoter; the Cre recombinase acts on LoxP/Lox2272 to convert the reverse PRRT2 encoding gene into a forward connection, and its promoter drives the expression.
在一种或多种实施方式中,还可设置诱导表达系统(如但不限于Tet-on/off)以实现可调控的PRRT2表达的开闭。In one or more embodiments, an inducible expression system (such as but not limited to Tet-on/off) may also be provided to achieve the on/off of regulatable PRRT2 expression.
在一种或多种实施方式中,所述的构建体包含在表达载体中或经基因编辑直接插入在干预对象基因组中;所述表达载体包括:病毒载体,非病毒载体;较佳地,所述的病毒载体包括:腺相关病毒(AAV)载体,慢病毒载体,腺病毒载体,逆转录病毒载体;更佳地,所述腺相关病毒载体包括:PHP.eB血清型AAV载体、Cap-B10血清型AAV载体;或所述基因编辑包括(但不限于)基于CRISPR-Cas(如Cas9)技术的基因编辑。 In one or more embodiments, the construct is contained in an expression vector or is directly inserted into the genome of the intervention object through gene editing; the expression vector includes: viral vector, non-viral vector; preferably, the viral vector includes: adeno-associated virus (AAV) vector, lentiviral vector, adenoviral vector, retroviral vector; more preferably, the adeno-associated virus vector includes: PHP.eB serotype AAV vector, Cap-B10 serotype AAV vector; or the gene editing includes (but is not limited to) gene editing based on CRISPR-Cas (such as Cas9) technology.
在一种或多种实施方式中,所述的PRRT2为斑马鱼来源的PRRT2,人源的PRRT2或鼠源的PRRT2;较佳地为斑马鱼来源的PRRT2。In one or more embodiments, the PRRT2 is PRRT2 from zebrafish, PRRT2 from human or PRRT2 from mouse; preferably, PRRT2 from zebrafish.
在一种或多种实施方式中,所述PRRT2为:(a)氨基酸序列如SEQ ID NO:2、4或6所示的蛋白;(b)将(a)蛋白的氨基酸序列经过一个或多个(如1-20个;较佳地1-15个;更佳地1-10个,如5个,3个)氨基酸残基的取代、缺失或添加而形成的,且具有(a)蛋白功能的由(a)衍生的蛋白;(c)与(a)蛋白的氨基酸序列有80%以上(较佳地85%以上;更佳地90%以上;更佳95%以上,如98%,99%)同源性且具有(a)蛋白功能的由(a)衍生的蛋白;或(d)(a)~(c)任一所述多肽的N或C末端添加标签序列,或在其N末端添加信号肽序列或分泌信号序列后形成的蛋白。In one or more embodiments, the PRRT2 is: (a) a protein having an amino acid sequence as shown in SEQ ID NO: 2, 4 or 6; (b) a protein derived from (a) which is obtained by replacing, deleting or adding one or more (e.g., 1-20; preferably 1-15; more preferably 1-10, such as 5, 3) amino acid residues in the amino acid sequence of the protein (a), and has the function of the protein (a); (c) a protein derived from (a) which has more than 80% (preferably more than 85%; more preferably more than 90%; more preferably more than 95%, such as 98%, 99%) homology with the amino acid sequence of the protein (a) and has the function of the protein (a); or (d) a protein formed by adding a tag sequence to the N or C terminus of any of the polypeptides of (a) to (c), or adding a signal peptide sequence or a secretion signal sequence to its N terminus.
在本发明的另一方面,提供一种治疗或预防癫痫的构建体,包括:表达驱动系统,以及由该驱动系统驱动表达的PRRT2编码基因。In another aspect of the present invention, a construct for treating or preventing epilepsy is provided, comprising: an expression drive system, and a PRRT2 encoding gene whose expression is driven by the drive system.
在一种或多种实施方式中,所述表达驱动系统包括单一、或组合形式的前脑兴奋性神经元特异性表达驱动系统。In one or more embodiments, the expression driving system comprises a single or combined forebrain excitatory neuron-specific expression driving system.
在一种或多种实施方式中,所述前脑兴奋性神经元特异性表达驱动系统包括单一形式的前脑谷氨酸能神经元特异性启动子,包括(但不限于):CaMKIIa启动子;单一形式的启动子用于直接驱动PRRT2的表达。In one or more embodiments, the forebrain excitatory neuron-specific expression driving system includes a single form of forebrain glutamatergic neuron-specific promoter, including (but not limited to): CaMKIIa promoter; a single form of promoter is used to directly drive the expression of PRRT2.
在一种或多种实施方式中,所述前脑兴奋性神经元特异性表达驱动系统包括组合形式的前脑谷氨酸能神经元特异性启动子,包括(但不限于):用于驱动Cre重组酶表达的前脑兴奋性神经元特异性启动子(如Emx1启动子或CaMKIIa启动子)和用于驱动PRRT2表达的强效启动子(如但不限于beta-actin promoter(CAG)启动子);组合形式的表达驱动系统用于兼顾前脑兴奋性神经元特异性和高效表达特性。In one or more embodiments, the forebrain excitatory neuron-specific expression driving system includes a combination of forebrain glutamatergic neuron-specific promoters, including (but not limited to): a forebrain excitatory neuron-specific promoter for driving Cre recombinase expression (such as Emx1 promoter or CaMKIIa promoter) and a strong promoter for driving PRRT2 expression (such as but not limited to beta-actin promoter (CAG) promoter); the combined expression driving system is used to take into account both forebrain excitatory neuron specificity and high-efficiency expression characteristics.
在一种或多种实施方式中,所述构建体包括操作性连接的:依赖于Cre重组酶的操作性基因表达调控元件,如(但不限于)Double-floxed inverse orientation,DIO);较佳地,所述用于DIO的元件为LoxP/Lox2272,利用基于LoxP和Lox2272的特定识别序列进行Cre重组酶依赖的PRRT2的表达。In one or more embodiments, the construct includes: an operably linked operative gene expression regulatory element dependent on Cre recombinase, such as (but not limited to) Double-floxed inverse orientation, DIO); preferably, the element for DIO is LoxP/Lox2272, and the Cre recombinase-dependent PRRT2 expression is performed using specific recognition sequences based on LoxP and Lox2272.
在一种或多种实施方式中,所述的构建体包含在表达载体中或经基因编辑直接插入在干预对象基因组中,所述表达载体包括:病毒载体,非病毒载体;较佳地,所述的病毒载体包括:腺相关病毒(AAV)载体,慢病毒载体,腺病毒载体,逆转录病毒载体;更佳地,所述腺相关病毒载体包括:PHP.eB血清型AAV载体、Cap-B10血清型AAV载体。In one or more embodiments, the construct is contained in an expression vector or is directly inserted into the genome of the intervention object through gene editing, and the expression vector includes: a viral vector, a non-viral vector; preferably, the viral vector includes: an adeno-associated virus (AAV) vector, a lentiviral vector, an adenoviral vector, a retroviral vector; more preferably, the adeno-associated virus vector includes: a PHP.eB serotype AAV vector, a Cap-B10 serotype AAV vector.
在一种或多种实施方式中,所述基因编辑包括(但不限于)基于CRISPR-Cas(如Cas9)技术的基因编辑。In one or more embodiments, the gene editing includes (but is not limited to) gene editing based on CRISPR-Cas (such as Cas9) technology.
在本发明的另一方面,提供一种用于治疗或预防癫痫的表达系统,其为病毒系统,由所述的病毒载体包装获得。 In another aspect of the present invention, an expression system for treating or preventing epilepsy is provided, which is a viral system obtained by packaging the viral vector.
在本发明的另一方面,提供一种用于治疗或预防癫痫的药物或含有所述药物的药盒,所述药物含有所述的表达系统。In another aspect of the present invention, a drug for treating or preventing epilepsy or a drug kit containing the drug is provided, wherein the drug contains the expression system.
在本发明的另一方面,提供一种将有效量的PRRT2、或所述选择性作用于前脑兴奋性神经元的PRRT2上调剂选择性递送至前脑兴奋性神经元的方法,包括使所述前脑兴奋神经元与包含有效量的PRRT2、或其上调剂的表达载体接触;较佳地,所述表达载体包括:病毒载体,非病毒载体;更佳地,所述的病毒载体包括:腺相关病毒(AAV)载体,慢病毒载体,腺病毒载体,逆转录病毒载体;更更佳地,所述腺相关病毒载体包括:PHP.eB血清型AAV载体、Cap-B10血清型AAV载体;较佳地,所述表达载体通过眶内注射(较佳为眶静脉注射)、颅内注射、鞘内(脊髓)注射、鞘内(大脑池)注射、脑内注射、心室内注射、直接注射到大脑中的癫痫病灶的方式进行递送。In another aspect of the present invention, a method for selectively delivering an effective amount of PRRT2, or the PRRT2 upregulator that selectively acts on forebrain excitatory neurons, to forebrain excitatory neurons is provided, comprising contacting the forebrain excitatory neurons with an expression vector comprising an effective amount of PRRT2, or its upregulator; preferably, the expression vector comprises: a viral vector, a non-viral vector; more preferably, the viral vector comprises: an adeno-associated virus (AAV) vector, a lentiviral vector, an adenoviral vector, a retroviral vector; more preferably, the adeno-associated virus vector comprises: a PHP.eB serotype AAV vector, a Cap-B10 serotype AAV vector; preferably, the expression vector is delivered by intraorbital injection (preferably orbital vein injection), intracranial injection, intrathecal (spinal cord) injection, intrathecal (cerebral cisterna magna) injection, intracerebral injection, intraventricular injection, or direct injection into the epileptic focus in the brain.
在本发明的另一方面,提供一种选择性作用于钠离子通道慢失活,选择性作用于细胞异常兴奋性,和/或选择性作用于前脑兴奋性神经元的方法,包括给予受试者有效量的PRRT2、或其上调剂(较佳为过表达该PRRT2的表达载体)。In another aspect of the present invention, a method for selectively acting on slow inactivation of sodium ion channels, selectively acting on abnormal cell excitability, and/or selectively acting on forebrain excitatory neurons is provided, comprising administering to a subject an effective amount of PRRT2, or an upregulator thereof (preferably an expression vector that overexpresses the PRRT2).
本发明的其它方面由于本发明的公开内容,对本领域的技术人员而言是显而易见的。Other aspects of the present invention will be apparent to those skilled in the art from the disclosure of the present invention.
图1、小鼠Prrt2基因编码蛋白区域(CDS)序列。Figure 1. The protein coding region (CDS) sequence of the mouse Prrt2 gene.
图2、小鼠PRRT2蛋白氨基酸序列。Fig. 2. Amino acid sequence of mouse PRRT2 protein.
图3、EGFP对照质粒pCAG-empty-IRES-EGFP示意图。Figure 3. Schematic diagram of EGFP control plasmid pCAG-empty-IRES-EGFP.
图4、小鼠PRRT2过表达质粒pCAG-mouse PRRT2(HA)-IRES-EGFP示意图。Figure 4. Schematic diagram of mouse PRRT2 overexpression plasmid pCAG-mouse PRRT2(HA)-IRES-EGFP.
图5、人PRRT2基因编码蛋白区域(CDS)序列。Fig. 5. The sequence of the protein coding region (CDS) of the human PRRT2 gene.
图6、人PRRT2蛋白氨基酸序列。Fig. 6. Amino acid sequence of human PRRT2 protein.
图7、人PRRT2过表达质粒pCAG-human PRRT2(HA)-IRES-EGFP示意图。Figure 7. Schematic diagram of human PRRT2 overexpression plasmid pCAG-human PRRT2(HA)-IRES-EGFP.
图8、斑马鱼Prrt2基因编码蛋白区域(CDS)序列。Fig. 8. The protein coding region (CDS) sequence of the zebrafish Prrt2 gene.
图9、斑马鱼PRRT2蛋白氨基酸序列。Fig. 9. Amino acid sequence of zebrafish PRRT2 protein.
图10、斑马鱼PRRT2过表达质粒pCAG-zebra PRRT2(HA)-IRES-EGFP示意图。Figure 10. Schematic diagram of zebrafish PRRT2 overexpression plasmid pCAG-zebra PRRT2(HA)-IRES-EGFP.
图11、PRRT2过表达对Nav1.2钠离子通道进入快失活过程的影响。Fig. 11. Effect of PRRT2 overexpression on the fast inactivation process of Nav1.2 sodium channel.
(A)稳转Nav1.2的HEK293细胞钠通道电流记录示意图。(A) Schematic diagram of sodium channel current recording in HEK293 cells stably expressing Nav1.2.
(B)上方为激活钠离子通道的电压钳制方案示意图。下方为代表性钠通道电流曲线。电压门控型钠通道Nav1.2在细胞被钳制到0mV时通道开放,钠离子内流形成内向钠电流。钠通道在短暂开放后快速失活,通道关闭,钠电流迅速衰减,如箭头所示。(B) The upper part is a schematic diagram of the voltage clamp scheme for activating sodium ion channels. The lower part is a representative sodium channel current curve. When the voltage-gated sodium channel Nav1.2 is clamped to 0 mV, the channel opens and sodium ions flow in to form an inward sodium current. After a short opening, the sodium channel quickly inactivates, the channel closes, and the sodium current decays rapidly, as shown by the arrow.
(C)钠离子通道快失活导致钠电流衰减,衰减速度用时间常数tau进行表征。过表达PRRT2组钠电流衰减的时间常数与过表达EGFP对照组相比,未见显著差异(n.s.P>0.05)。mPRRT2:小鼠PRRT2。 (C) Rapid inactivation of sodium channels leads to sodium current decay, and the decay rate is characterized by the time constant tau. There was no significant difference in the time constant of sodium current decay between the PRRT2 overexpression group and the EGFP overexpression control group (nsP>0.05). mPRRT2: mouse PRRT2.
图12、PRRT2过表达对Nav1.2钠离子通道从快失活恢复的影响。Fig. 12. Effect of PRRT2 overexpression on the recovery of Nav1.2 sodium channel from fast inactivation.
(A)上方为检测钠离子通道从快失活恢复的电压钳制方案示意图。下方分别为对照组细胞第一次去极化时测得的钠电流示例,以及快失活的钠离子通道经历不同时长的超极化恢复期后测得的钠电流示例。(A) Schematic diagram of the voltage clamp protocol for detecting the recovery of sodium channels from fast inactivation. Below are examples of sodium currents measured in control cells during the first depolarization and in fast inactivating sodium channels after hyperpolarization recovery periods of varying lengths.
(B)钠离子通道从快失活状态恢复的过程。过表达mPRRT2组与过表达EGFP对照组相比,未见显著差异(n.s.P>0.05)。mPRRT2:小鼠PRRT2。(B) The process of sodium channel recovery from the fast inactivation state. There was no significant difference between the mPRRT2 overexpression group and the EGFP overexpression control group (n.s.P>0.05). mPRRT2: mouse PRRT2.
图13、PRRT2过表达对Nav1.2钠离子通道进入慢失活过程的影响。Fig. 13. Effect of PRRT2 overexpression on the slow inactivation process of Nav1.2 sodium channel.
(A)上方为检测钠离子通道进入慢失活状态的电压钳制方案示意图。下方分别为对照组细胞第一次去极化时测得的钠电流示例,以及钠离子通道经历不同时长的去极化后,间隔短暂超极化后测得的钠电流示例。(A) The top is a schematic diagram of the voltage clamp protocol for detecting the slow inactivation of sodium channels. The bottom is an example of the sodium current measured during the first depolarization of control cells and the sodium current measured after the sodium channels experienced depolarization of different lengths followed by a brief hyperpolarization.
(B)钠离子通道进入慢失活状态的过程。与过表达EGFP的对照组相比,过表达mPRRT2组的钠离子通道在持续的去极化过程中更快的进入慢失活状态。两组间存在显著性差异(P<0.0001)。mPRRT2:小鼠PRRT2。(B) The process of sodium channels entering the slow inactivation state. Compared with the control group overexpressing EGFP, the sodium channels in the mPRRT2 overexpression group entered the slow inactivation state faster during sustained depolarization. There was a significant difference between the two groups (P < 0.0001). mPRRT2: mouse PRRT2.
图14、PRRT2过表达对Nav1.2钠离子通道从慢失活恢复的影响。Fig. 14. Effect of PRRT2 overexpression on the recovery of Nav1.2 sodium channel from slow inactivation.
(A)上方为检测钠离子通道从慢失活恢复的电压钳制方案示意图。下方分别为第一次去极化时测得的钠电流示例,以及慢失活的钠离子通道经历不同时长的超极化恢复期后测得的钠电流示例。(A) Schematic diagram of the voltage clamp protocol for detecting the recovery of sodium channels from slow inactivation. Below are examples of sodium currents measured during the first depolarization and after the slowly inactivating sodium channels have undergone hyperpolarization recovery periods of varying lengths.
(B)钠离子通道从慢失活状态恢复的过程。与过表达EGFP的对照组相比,过表达mPRRT2组的钠离子通道从慢失活状态恢复的速度更慢。两组间存在显著差异(P<0.001)。(B) The process of sodium channel recovery from slow inactivation. Compared with the control group overexpressing EGFP, the sodium channel in the mPRRT2 overexpression group recovered more slowly from the slow inactivation state. There was a significant difference between the two groups (P < 0.001).
图15、斑马鱼PRRT2过表达对Nav1.2钠离子通道慢失活的影响。Fig. 15. Effect of zebrafish PRRT2 overexpression on slow inactivation of Nav1.2 sodium channel.
(A)小鼠、人和斑马鱼PRRT2蛋白氨基酸序列比对。小鼠和人的PRRT2蛋白氨基酸序列一致性为77.46%,斑马鱼和小鼠的PRRT2蛋白氨基酸序列一致性为43.6%。序列中底部横线标注了PRRT2的C末端(包含跨膜段)。(A) Comparison of the amino acid sequences of mouse, human and zebrafish PRRT2 proteins. The amino acid sequence identity of mouse and human PRRT2 proteins is 77.46%, and the amino acid sequence identity of zebrafish and mouse PRRT2 proteins is 43.6%. The bottom horizontal line in the sequence marks the C-terminus of PRRT2 (including the transmembrane segment).
(B)钠离子通道进入慢失活状态的过程。与过表达EGFP的对照组相比,过表达mPRRT2、hPRRT2和zPRRT2组的钠离子通道在持续的去极化过程中更快的进入慢失活状态,组间存在显著性差异(P<0.0001)。(B) The process of sodium channels entering the slow inactivation state. Compared with the control group overexpressing EGFP, the sodium channels in the mPRRT2, hPRRT2, and zPRRT2 overexpression groups entered the slow inactivation state faster during sustained depolarization, and there was a significant difference between the groups (P < 0.0001).
(C)钠离子通道从慢失活状态恢复的过程。与过表达EGFP的对照组相比,过表达mPRRT2、hPRRT2和zPRRT2组的钠离子通道从慢失活状态恢复的速度更慢,组间存在显著差异(P<0.0001)。另外,过表达zPRRT2组的钠离子通道从慢失活状态恢复的速度显著慢于mPRRT2和hPRRT2组,组间存在显著差异(P<0.0001)。mPRRT2:小鼠PRRT2,hPRRT2:人PRRT2,zPRRT2:斑马鱼PRRT2。(C) The process of sodium channel recovery from slow inactivation. Compared with the control group overexpressing EGFP, the sodium channels in the mPRRT2, hPRRT2, and zPRRT2 overexpression groups recovered more slowly from the slow inactivation state, with significant differences between the groups (P < 0.0001). In addition, the sodium channels in the zPRRT2 overexpression group recovered more slowly from the slow inactivation state than those in the mPRRT2 and hPRRT2 groups, with significant differences between the groups (P < 0.0001). mPRRT2: mouse PRRT2, hPRRT2: human PRRT2, zPRRT2: zebrafish PRRT2.
图16、腺相关病毒过表达mCherry质粒pAAV-CAG-DIO-mCherry示意图。Figure 16. Schematic diagram of adeno-associated virus overexpressing mCherry plasmid pAAV-CAG-DIO-mCherry.
图17、腺相关病毒过表达小鼠PRRT2质粒pAAV-CAG-DIO-mouse PRRT2-HA示意图。Figure 17. Schematic diagram of adeno-associated virus overexpressing mouse PRRT2 plasmid pAAV-CAG-DIO-mouse PRRT2-HA.
图18、前脑兴奋性神经元过表达PRRT2。 Figure 18. Forebrain excitatory neurons overexpress PRRT2.
(A)Emx1-cre小鼠眶静脉注射AAV-PHP.eB病毒示意图。(A) Schematic diagram of orbital vein injection of AAV-PHP.eB virus into Emx1-cre mice.
(B)在前脑兴奋性神经元中选择性过表达PRRT2示意图。PRRT2-OE:PRRT2over-expression。(B) Schematic diagram of selective overexpression of PRRT2 in forebrain excitatory neurons. PRRT2-OE: PRRT2 over-expression.
(C)荧光免疫组化验证PRRT2在前脑兴奋性神经元的表达。(C) Fluorescent immunohistochemistry verified the expression of PRRT2 in forebrain excitatory neurons.
图19、前脑兴奋性神经元过表达PRRT2抑制惊厥剂诱导的癫痫发作。FIG. 19 . Overexpression of PRRT2 in forebrain excitatory neurons inhibits convulsant-induced epileptic seizures.
(A)惊厥剂(戊四唑,PTZ,45mg/kg,腹腔注射)诱导小鼠癫痫行为代表性示例。(A) Representative examples of epileptic behaviors induced by convulsants (pentylenetetrazole, PTZ, 45 mg/kg, intraperitoneal injection) in mice.
(B)与对照组相比,前脑兴奋性神经元过表达PRRT2显著降低了戊四唑诱导的小鼠癫痫发作级别,两组的发作级别存在显著性差异(P<0.0001)。PTZ:pentylenetetrazol;PRRT2-OE:PRRT2 over-expression。(B) Compared with the control group, overexpression of PRRT2 in forebrain excitatory neurons significantly reduced the severity of epileptic seizures induced by pentylenetetrazol in mice, and there was a significant difference in the severity of seizures between the two groups (P < 0.0001). PTZ: pentylenetetrazol; PRRT2-OE: PRRT2 over-expression.
图20、前脑兴奋性神经元过表达PRRT2抑制惊厥剂诱导的癫痫样脑电。Figure 20. Overexpression of PRRT2 in forebrain excitatory neurons inhibits epileptic EEG induced by convulsants.
(A)小鼠脑电记录示意图。(A) Schematic diagram of EEG recording in mice.
(B)代表性脑电示例。戊四唑(PTZ)可以诱导小鼠大脑皮层出现癫痫样脑电信号;在前脑兴奋性神经元过表达PRRT2可以有效抑制戊四唑诱导的小鼠大脑皮层癫痫样放电事件。(B) Representative EEG examples. Pentylenetetrazol (PTZ) can induce epileptiform EEG signals in the mouse cerebral cortex; overexpression of PRRT2 in forebrain excitatory neurons can effectively inhibit PTZ-induced epileptiform discharge events in the mouse cerebral cortex.
图21、前脑兴奋性神经元过表达PRRT2不影响小鼠正常行为活动。Figure 21. Overexpression of PRRT2 in forebrain excitatory neurons does not affect the normal behavioral activities of mice.
(A)在旷场测试中,两组小鼠的活动总距离相似,未检测到显著性差异(n.s.P>0.05)。(A) In the open field test, the total distance covered by the two groups of mice was similar, and no significant difference was detected (n.s.P>0.05).
(B)在转轮测试中,两组小鼠在定速转轮上维持的时间相似,未检测到显著性差异(n.s.P>0.05)。该测试的时间上限为60秒。(B) In the wheel test, the time that the two groups of mice could maintain on the fixed-speed wheel was similar, and no significant difference was detected (n.s.P>0.05). The upper limit of the time for this test was 60 seconds.
(C)在社交行为测试中,两组动物均表现出与同伴小鼠的交流倾向,且两组动物间未检测到显著性差异(n.s.P>0.05)。Empty表示空盒组,Novel表示陌生小鼠组。(C) In the social behavior test, both groups of animals showed a tendency to communicate with their companion mice, and no significant difference was detected between the two groups (n.s.P>0.05). Empty represents the empty box group, and Novel represents the unfamiliar mouse group.
图22、前脑抑制型神经元过表达PRRT2易化惊厥剂诱导的癫痫发作。FIG. 22 . Overexpression of PRRT2 in forebrain inhibitory neurons facilitates convulsant-induced epileptic seizures.
(A)Nkx2.1-cre小鼠眶静脉注射AAV-PHP.eB病毒示意图。(A) Schematic diagram of orbital vein injection of AAV-PHP.eB virus into Nkx2.1-cre mice.
(B)在前脑抑制型神经元中选择性过表达PRRT2示意图。(B) Schematic diagram of selective overexpression of PRRT2 in forebrain inhibitory neurons.
(C)荧光免疫组化验证PRRT2在前脑抑制型神经元的表达。标尺为1mm。(C) Fluorescence immunohistochemistry verified the expression of PRRT2 in forebrain inhibitory neurons. The scale bar is 1 mm.
(D)阈下剂量的惊厥剂(戊四唑,PTZ,35mg/kg,腹腔注射)诱导Nkx2.1-cre小鼠癫痫行为的代表性示例;PTZ:pentylenetetrazol。(D) Representative examples of epileptic behaviors induced by subthreshold doses of convulsants (pentylenetetrazol, PTZ, 35 mg/kg, intraperitoneal injection) in Nkx2.1-cre mice; PTZ: pentylenetetrazol.
(E)与对照组相比,前脑抑制性神经元过表达PRRT2显著增强了阈下剂量戊四唑诱导的Nkx2.1-cre小鼠癫痫发作级别,两组的发作级别存在显著性差异(P<0.01)。PRRT2-OE:PRRT2over-expression。(E) Compared with the control group, overexpression of PRRT2 in forebrain inhibitory neurons significantly enhanced the level of epileptic seizures induced by subthreshold doses of pentylenetetrazol in Nkx2.1-cre mice, and there was a significant difference in the seizure levels between the two groups (P < 0.01). PRRT2-OE: PRRT2 over-expression.
(F)小鼠脑电记录示意图。(F) Schematic diagram of mouse EEG recording.
(G)代表性脑电示例。阈下剂量的戊四唑无法诱导对照组小鼠大脑皮层出现癫痫样脑电信号;但是在前脑抑制性神经元过表达PRRT2组小鼠中,阈下剂量的戊四唑可以诱导小鼠大脑皮层癫痫样放电事件。(G) Representative EEG examples. Subthreshold doses of PTZ could not induce epileptic EEG signals in the cerebral cortex of control mice; however, subthreshold doses of PTZ could induce epileptic discharge events in the cerebral cortex of mice overexpressing PRRT2 in forebrain inhibitory neurons.
本发明人经过深入的研究,首次发现PRRT2(富含脯氨酸的跨膜蛋白2,proline-rich transmembrane protein 2)基因编码的蛋白可以选择性作用于慢失活状态的钠离子通道,并通过增强钠离子通道的慢失活进而减少可激活钠离子通道的有效供给。进一步地,发明人通过在前脑兴奋性神经元中选择性过表达PRRT2蛋白,反馈式降低异常兴奋状态神经元的兴奋性,在小鼠癫痫模型中,实现了在不干扰日常行为的基础上有效抑制癫痫发作的目标。前脑兴奋性神经元特异性表达PRRT2是成功干预癫痫并减少副作用的关键。在前脑兴奋性神经元中表达PRRT2,同时具备细胞类型选择性和细胞状态选择性,在癫痫治疗领域具有良好应用前景。After in-depth research, the inventors discovered for the first time that the protein encoded by the PRRT2 (proline-rich transmembrane protein 2) gene can selectively act on sodium channels in a slow inactivation state, and reduce the effective supply of activatable sodium channels by enhancing the slow inactivation of sodium channels. Furthermore, the inventors selectively overexpressed the PRRT2 protein in forebrain excitatory neurons to feedback reduce the excitability of neurons in abnormally excited states, and achieved the goal of effectively suppressing epileptic seizures without interfering with daily behavior in a mouse epilepsy model. The specific expression of PRRT2 in forebrain excitatory neurons is the key to successfully intervening in epilepsy and reducing side effects. The expression of PRRT2 in forebrain excitatory neurons has both cell type selectivity and cell state selectivity, and has good application prospects in the field of epilepsy treatment.
PRRT2和钠离子通道PRRT2 and sodium channels
在本发明中,术语“PRRT2”包括具有SEQ ID NO:2(小鼠)、SEQ ID NO:4(人)、或SEQ ID NO:6(斑马鱼)的蛋白,也包括具有与PRRT2相同功能的序列的变异形式。In the present invention, the term "PRRT2" includes proteins having SEQ ID NO: 2 (mouse), SEQ ID NO: 4 (human), or SEQ ID NO: 6 (zebrafish), and also includes variant forms of sequences having the same function as PRRT2.
这些变异形式包括但并不限于:若干个(通常为1-50个,较佳地1-30个,更佳地1-20个,最佳地1-10个,还更佳如1-8个、1-5个)氨基酸的缺失、插入和/或取代,以及在C末端和/或N末端添加或缺失一个或数个(通常为20个以内,较佳地为10个以内,更佳地为5个以内)氨基酸。例如,在本领域中,用性能相近或相似的氨基酸进行取代时,通常不会改变蛋白质的功能。又比如,在C末端和/或N末端添加或缺失一个或数个氨基酸通常也不会改变蛋白质的功能。该术语还包括PRRT2的活性片段和活性衍生物。These variant forms include but are not limited to: deletion, insertion and/or substitution of several (usually 1-50, preferably 1-30, more preferably 1-20, most preferably 1-10, and even more preferably 1-8, 1-5) amino acids, and addition or deletion of one or several (usually within 20, preferably within 10, and more preferably within 5) amino acids at the C-terminus and/or N-terminus. For example, in the art, when amino acids with similar or similar properties are substituted, the function of the protein is usually not changed. For another example, adding or deleting one or several amino acids at the C-terminus and/or N-terminus usually does not change the function of the protein. The term also includes active fragments and active derivatives of PRRT2.
上述术语“PRRT2”还包括与上述SEQ ID NO:2、4或6所限定的多肽序列的同源性为80%或更高;较佳地同源性为85%或更高,如同源性90%,95%,98%或99%)的、且具有本发明中实施例中所涉及的PRRT2相同功能的蛋白也包括在本发明内。比对序列相同性的方法和工具也是本领域周知的,例如BLAST。“同源性”是指按照位置相同的百分比,两条或多条核酸或多肽之间的相似水平(即序列相似性或同一性)。The above term "PRRT2" also includes proteins having a homology of 80% or higher with the polypeptide sequence defined by the above SEQ ID NO: 2, 4 or 6; preferably a homology of 85% or higher, such as 90%, 95%, 98% or 99% homology) and having the same function as the PRRT2 involved in the embodiments of the present invention are also included in the present invention. Methods and tools for aligning sequence identity are also well known in the art, such as BLAST. "Homology" refers to the level of similarity (i.e., sequence similarity or identity) between two or more nucleic acids or polypeptides based on the percentage of identical positions.
编码PRRT2或其变异蛋白的多核苷酸序列(编码序列)也可以应用到本发明中。术语“编码基因”可以是包括编码所述蛋白的多核苷酸,也可以是还包括附加编码和/或非编码序列的多核苷酸。在一些实施方案中,编码PRRT2蛋白的多核苷酸序列(编码序列)是SEQ ID NO:1、3或5所示的多核苷酸序列。A polynucleotide sequence encoding PRRT2 or its variant protein (coding sequence) can also be applied to the present invention. The term "coding gene" can be a polynucleotide that includes the protein, or a polynucleotide that also includes additional coding and/or non-coding sequences. In some embodiments, the polynucleotide sequence encoding the PRRT2 protein (coding sequence) is a polynucleotide sequence shown in SEQ ID NO: 1, 3 or 5.
本发明人的研究工作中,发现PRRT2(proline-rich transmembrane protein 2)基因编码的蛋白可以选择性作用于慢失活状态的钠离子通道,即,PRRT2过表达可以促进钠离子通道的慢失活,但对钠离子通道快失活影响不显著。PRRT2具有钠离子通道状态依赖性调控特征,通过增强钠离子通道的慢失活,减少持续去极化状态下细胞钠离子通道的有效供给。PRRT2的该特征有利于其在不影响细胞正常兴奋性活动的情况下,降低细胞的异常兴奋性,使其具有良好的细胞状态选择性。In the research work of the inventors, it was found that the protein encoded by the PRRT2 (proline-rich transmembrane protein 2) gene can selectively act on the sodium ion channels in the slow inactivation state, that is, PRRT2 overexpression can promote the slow inactivation of sodium ion channels, but has no significant effect on the fast inactivation of sodium ion channels. PRRT2 has the characteristics of sodium ion channel state-dependent regulation. By enhancing the slow inactivation of sodium ion channels, it reduces the effective supply of cellular sodium ion channels under the state of continuous depolarization. This feature of PRRT2 is conducive to reducing the abnormal excitability of cells without affecting the normal excitatory activity of cells, so that it has good cell state selectivity.
基于此发现,发明人利用腺相关病毒作为基因递送载体,通过在前脑兴奋性神经元中选择性过表达PRRT2蛋白,反馈式降低异常兴奋状态神经元的兴奋性,通过小鼠癫痫模型的动物 试验发现,选择性靶向PRRT2治疗可以在不干扰日常行为的基础上有效抑制癫痫发作。Based on this discovery, the inventors used adeno-associated virus as a gene delivery vector, selectively overexpressed PRRT2 protein in the excitatory neurons of the forebrain, and feedback-reduced the excitability of abnormally excited neurons. The trial found that selectively targeting PRRT2 therapy can effectively suppress epileptic seizures without interfering with daily behavior.
本发明中,术语“电压门控型钠离子通道”、“钠离子通道”、“Nav”可互换使用。In the present invention, the terms "voltage-gated sodium ion channel", "sodium ion channel" and "Nav" are used interchangeably.
作用于前脑兴奋性神经元的PRRT2及其上调剂的应用PRRT2 acting on forebrain excitatory neurons and application of its upregulators
基于本发明人的上述新发现,本发明提供了一种选择性作用于前脑兴奋性神经元的PRRT2的上调剂的用途,用于制备治疗或预防癫痫的药物。Based on the above new discovery of the inventors, the present invention provides a use of an up-regulator of PRRT2 that selectively acts on forebrain excitatory neurons, for preparing a drug for treating or preventing epilepsy.
本发明所述的“受试者”包括但不限于:人、非人灵长类动物、啮齿类动物(如小鼠)。The "subject" described in the present invention includes but is not limited to: humans, non-human primates, and rodents (such as mice).
本领域技术人员知晓,“癫痫”发作可以分为不同水平,包括但不限于:强直-阵挛、强直、阵挛、肌阵挛、失神或失张力发作。在小鼠模型中,癫痫发作可以分为六级,分别是:一级:小鼠伏地不动,腹部贴地爬行;二级:突发性抽搐,行为突然停止,尾巴翘起;三级:肌肉阵挛,头部扭转,抬手抽搐;四级:强直-阵挛,倒地抽搐,疯跳疯跑;五级:倒地并发展至四肢强直性伸直;六级:死亡。Those skilled in the art know that "epileptic" seizures can be divided into different levels, including but not limited to: tonic-clonic, tonic, clonic, myoclonic, absence or atonic seizures. In the mouse model, epileptic seizures can be divided into six levels, namely: Level 1: The mouse lies motionless on the ground, crawling on the belly; Level 2: sudden convulsions, sudden cessation of behavior, and the tail is raised; Level 3: muscle clonus, head twisting, hand convulsions; Level 4: tonic-clonic, falling to the ground and convulsions, jumping and running wildly; Level 5: falling to the ground and developing to rigid extension of limbs; Level 6: death.
在一种或多种实施方式中,通过PRRT2的上调剂,选择性作用于前脑兴奋性神经元,可以将受试者所经历的癫痫发作的次数、频率、水平和/或持续时间降低约5%、约10%、约10%、约15%、约20%、约25%、约30%、约35%、约40%、约45%、约50%、约55%、约60%、约65%、约70%、约75%、约80%、约85%、约90%、约95%、约99%或100%,包括其间的所有范围和子范围。In one or more embodiments, upregulators of PRRT2, acting selectively on forebrain excitatory neurons, can reduce the number, frequency, level and/or duration of epileptic seizures experienced by a subject by about 5%, about 10%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 99% or 100%, including all ranges and subranges therebetween.
所述“选择性作用”是相对的,例如,相对于非前脑兴奋性神经元,对前脑兴奋性神经元的作用增加约5%、约10%、约10%、约15%、约20%、约25%、约30%、约35%、约40%、约45%、约50%、约55%、约60%、约65%、约70%、约75%、约80%、约85%、约90%、约95%、约99%或100%,包括其间的所有范围和子范围。The "selective effect" is relative, for example, the effect on forebrain excitatory neurons is increased by about 5%, about 10%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 99% or 100%, relative to non-forebrain excitatory neurons, including all ranges and subranges therebetween.
所述“兴奋性神经元”是指可以释放神经递质例如谷氨酸的一种神经元,其可以导致连接的突触后神经元变得更可能被激发。所述“抑制性神经元”是指可以释放神经递质例如γ氨基丁酸的一种神经元,其可以导致连接的突触后神经元变得更不容易被激发。The "excitatory neuron" is a neuron that can release neurotransmitters such as glutamate, which can cause the connected postsynaptic neuron to become more likely to be excited. The "inhibitory neuron" is a neuron that can release neurotransmitters such as gamma-aminobutyric acid, which can cause the connected postsynaptic neuron to become less likely to be excited.
如本发明所用,所述的PRRT2的上调剂包括了激动剂、促进剂、兴奋剂等,这些术语可互换使用。所述的PRRT2的上调剂是指任何可提高PRRT2的活性、增强PRRT2的稳定性、上调PRRT2的表达、增加PRRT2有效作用时间、或促进PRRT2基因的转录和翻译的物质,这些物质均可用于本发明,作为对于上调PRRT2有用的物质,从而可用于治疗或预防癫痫。As used in the present invention, the PRRT2 upregulator includes agonists, promoters, stimulants, etc., and these terms can be used interchangeably. The PRRT2 upregulator refers to any substance that can increase the activity of PRRT2, enhance the stability of PRRT2, upregulate the expression of PRRT2, increase the effective action time of PRRT2, or promote the transcription and translation of the PRRT2 gene. These substances can be used in the present invention as substances useful for upregulating PRRT2, and thus can be used to treat or prevent epilepsy.
在一种或多种实施方式中,所述的PRRT2的上调剂包括(但不限于):构建体、或该构建体形成的表达系统(例如病毒(感染)系统);所述构建体包括:前脑兴奋性神经元特异性表达驱动系统,以及由该驱动系统驱动表达的PRRT2编码基因。通常,所述前脑兴奋性神经元特异性表达驱动系统包括前脑兴奋性神经元特异性表达启动子。作为本发明的优选方式,所述前脑兴奋性神经元特异性表达启动子包括:Emx1启动子和CaMKIIa启动子。In one or more embodiments, the PRRT2 up-regulator includes (but is not limited to): a construct, or an expression system formed by the construct (e.g., a viral (infection) system); the construct includes: a forebrain excitatory neuron-specific expression drive system, and a PRRT2 encoding gene driven by the drive system. Typically, the forebrain excitatory neuron-specific expression drive system includes a forebrain excitatory neuron-specific expression promoter. As a preferred embodiment of the present invention, the forebrain excitatory neuron-specific expression promoter includes: Emx1 promoter and CaMKIIa promoter.
在一种或多种实施方式中,所述构建体包括:启动子,用于DIO的元件,PRRT2编码 基因表达盒(较佳地,PRRT2编码基因反向连接),前脑兴奋性神经元特异性表达启动子驱动的Cre重组酶编码基因;较佳地,所述用于DIO的元件为LoxP/Lox2272元件,利用基于LoxP和Lox2272的DIO调控方法进行cre重组酶依赖的PRRT2的表达。In one or more embodiments, the construct includes: a promoter, an element for DIO, a PRRT2 encoding A gene expression cassette (preferably, a PRRT2 encoding gene is reversely linked), a forebrain excitatory neuron-specific expression promoter-driven Cre recombinase encoding gene; preferably, the element used for DIO is a LoxP/Lox2272 element, and a DIO regulation method based on LoxP and Lox2272 is used to perform cre recombinase-dependent expression of PRRT2.
作为本发明的优选方式,所述构建体包括:As a preferred embodiment of the present invention, the construct comprises:
构建体1,包括顺序连接的:启动子(如但不限于beta-actin promoter),LoxP/Lox2272,PRRT2编码基因,LoxP/Lox2272;所述PRRT2编码基因反向连接;Construct 1, comprising: a promoter (such as but not limited to beta-actin promoter), LoxP/Lox2272, a PRRT2 encoding gene, LoxP/Lox2272 connected in sequence; the PRRT2 encoding gene is reversely connected;
构建体2:前脑兴奋性神经元特异性表达启动子,cre重组酶编码基因(如Emx1启动子驱动cre重组酶编码基因的表达)。Construct 2: a promoter specifically expressing forebrain excitatory neurons and a gene encoding cre recombinase (eg, Emx1 promoter drives the expression of the gene encoding cre recombinase).
本发明中,PRRT2多核苷酸序列可插入到重组表达载体中,从而可将之转入到细胞中,过表达产生PRRT2。只要能在宿主体内复制和稳定,任何质粒和载体都可以用于本发明。本领域的技术人员熟知的方法能用于构建含PRRT2的DNA序列和合适的转录/翻译控制信号的表达载体。这些方法包括体外重组DNA技术、DNA合成技术、体内重组技术等。表达载体的一个重要特征是通常含有复制起点、启动子、标记基因和翻译控制元件。例如,所述的表达载体包括:病毒载体,非病毒载体;较佳地,所述的病毒载体包括(但不限于):腺相关病毒(AAV)载体,慢病毒载体,腺病毒载体,逆转录病毒载体等。In the present invention, the PRRT2 polynucleotide sequence can be inserted into a recombinant expression vector, so that it can be transferred into cells to overexpress PRRT2. As long as it can replicate and be stable in the host, any plasmid and vector can be used in the present invention. Methods well known to those skilled in the art can be used to construct expression vectors containing the DNA sequence of PRRT2 and appropriate transcription/translation control signals. These methods include in vitro recombinant DNA technology, DNA synthesis technology, in vivo recombination technology, etc. An important feature of the expression vector is that it usually contains a replication origin, a promoter, a marker gene and a translation control element. For example, the expression vector includes: a viral vector, a non-viral vector; preferably, the viral vector includes (but is not limited to): an adeno-associated virus (AAV) vector, a lentiviral vector, an adenoviral vector, a retroviral vector, etc.
本发明中,由于所述载体选择性作用于前脑兴奋性神经元,因此,所述载体具有良好血脑屏障穿透能力,例如能够穿透血脑屏障的载体占总载体的2%以上、5%以上、10%以上、20%以上、30%以上或更多。在本发明的一种或多种实施方式中,所述病毒载体为AAV载体。“AAV病毒体”或“AAV病毒”或“AAV病毒颗粒”或“AAV载体颗粒”或“AAV载体”是指包含至少一种AAV衣壳多肽和多核苷酸(例如PRRT2多核苷酸序列)的病毒颗粒。AAV载体通常根据衣壳多肽的名称(血清型)命名。所述AAV载体包括(但不限于):PHP.eB血清型AAV载体、Cap-B10血清型AAV载体。In the present invention, since the vector selectively acts on the excitatory neurons of the forebrain, the vector has a good blood-brain barrier penetration ability, for example, the vector that can penetrate the blood-brain barrier accounts for more than 2%, more than 5%, more than 10%, more than 20%, more than 30% or more of the total vector. In one or more embodiments of the present invention, the viral vector is an AAV vector. "AAV virion" or "AAV virus" or "AAV virus particle" or "AAV vector particle" or "AAV vector" refers to a viral particle comprising at least one AAV capsid polypeptide and a polynucleotide (such as a PRRT2 polynucleotide sequence). AAV vectors are usually named according to the name (serotype) of the capsid polypeptide. The AAV vectors include (but are not limited to): PHP.eB serotype AAV vector, Cap-B10 serotype AAV vector.
作为本发明的一种实施方式,可将编码PRRT2的基因通过常规的方法克隆到适当的载体(如常规原核或真核表达载体、或病毒载体如疱疹病毒载体或腺相关病毒载体)中,将所述的载体导入到可表达所述PRRT2的细胞中,使所述的细胞表达PRRT2。可通过将适量的所述细胞引入到受试者身体的适当部位,实现PRRT2的表达。作为本发明的优选方式,采用眶内注射(例如眶静脉注射)的方式给予所述载体,使其选择性作用于前脑兴奋性神经元,减少血脑屏障的影响。本领域技术人员已知,其他的给药方式也可以用于本发明,例如但不限于:颅内注射、鞘内(脊髓)注射、鞘内(大脑池)注射、脑内注射、心室内注射、直接注射到海马中的癫痫病灶、直接注射到颞叶中的癫痫病灶。As an embodiment of the present invention, the gene encoding PRRT2 can be cloned into an appropriate vector (such as a conventional prokaryotic or eukaryotic expression vector, or a viral vector such as a herpes virus vector or an adeno-associated virus vector) by conventional methods, and the vector is introduced into a cell that can express the PRRT2, so that the cell expresses PRRT2. The expression of PRRT2 can be achieved by introducing an appropriate amount of the cells into an appropriate part of the subject's body. As a preferred embodiment of the present invention, the vector is administered by intraorbital injection (such as orbital vein injection) so that it selectively acts on excitatory neurons in the forebrain and reduces the influence of the blood-brain barrier. It is known to those skilled in the art that other modes of administration can also be used in the present invention, such as but not limited to: intracranial injection, intrathecal (spinal cord) injection, intrathecal (cerebral cistern) injection, intracerebral injection, intraventricular injection, direct injection into the epileptic lesion in the hippocampus, and direct injection into the epileptic lesion in the temporal lobe.
本发明人经过研究分析,选择PHP.eB血清型AAV病毒来作为PRRT2基因的递送载体,其具有良好血脑屏障穿透能力。考虑到基因递送载体正处于快速迭代和发展的时期,使用有助于将PRRT2基因高效递送至脑内神经元的载体(如Cap-B10血清型AAV病毒等)以及有助于将PRRT2基因高效递送至前脑兴奋性神经元的载体均可应用于该实验。 After research and analysis, the inventors selected PHP.eB serotype AAV virus as the delivery vector of PRRT2 gene, which has good blood-brain barrier penetration ability. Considering that gene delivery vectors are in a period of rapid iteration and development, vectors that help to efficiently deliver PRRT2 gene to neurons in the brain (such as Cap-B10 serotype AAV virus, etc.) and vectors that help to efficiently deliver PRRT2 gene to excitatory neurons in the forebrain can be applied to this experiment.
本发明中,过表达产生PRRT2后,可以使用本领域技术人员熟知的方法检测过表达PRRT2是否成功。优选的,当进行基因水平的检测时,可以采用特异性扩增PRRT2的引物;或特异性识别PRRT2的探针来确定PRRT2基因的存在与否;当进行蛋白水平的检测时,可以采用特异性结合PRRT2编码的蛋白的抗体或配体来确定PRRT2蛋白的表达情况,例如利用抗PRRT2抗体与PRRT2蛋白结合,然后使用带有检测信号(优选为荧光标签)的二抗与抗PRRT2抗体结合,通过检测信号的有无,或检测信号的强弱进行判断。In the present invention, after overexpression to produce PRRT2, methods well known to those skilled in the art can be used to detect whether the overexpression of PRRT2 is successful. Preferably, when performing gene level detection, primers that specifically amplify PRRT2 can be used; or probes that specifically recognize PRRT2 can be used to determine the presence or absence of the PRRT2 gene; when performing protein level detection, antibodies or ligands that specifically bind to the protein encoded by PRRT2 can be used to determine the expression of the PRRT2 protein, for example, using an anti-PRRT2 antibody to bind to the PRRT2 protein, and then using a secondary antibody with a detection signal (preferably a fluorescent label) to bind to the anti-PRRT2 antibody, and judging by the presence or absence of the detection signal, or the strength of the detection signal.
本发明还提供了一种治疗或预防癫痫的方法,包括选择性给予受试者前脑兴奋性神经元有效量的PRRT2、或其上调剂(如过表达该PRRT2的表达载体)。The present invention also provides a method for treating or preventing epilepsy, comprising selectively administering an effective amount of PRRT2 or an up-regulator thereof (such as an expression vector overexpressing the PRRT2) to excitatory neurons of the forebrain of a subject.
术语“治疗”和“预防”通常指使用药物或方法来减少、消除或预防疾病的症状,并且包括实现治疗益处和/或预防益处。治疗益处是指减缓所治疗的病症或病症的症状的进展、停止进展、逆转进展、或根除或改善症状。预防益处包括降低病症的风险、延缓病症的进展或降低病症发生的可能性。The terms "treat" and "prevent" generally refer to the use of drugs or methods to reduce, eliminate or prevent the symptoms of a disease, and include achieving a therapeutic benefit and/or a prophylactic benefit. A therapeutic benefit refers to slowing the progression of the condition or symptoms of the condition being treated, halting the progression, reversing the progression, or eradicating or ameliorating the symptoms. A prophylactic benefit includes reducing the risk of a condition, delaying the progression of a condition, or reducing the likelihood of a condition occurring.
在得知了所述的PRRT2的用途后,可以采用本领域熟知的多种方法来将所述的PRRT2或其上调剂给药于受试者。优选的,可采用基因治疗的手段进行,比如可以通过一定的途径将携带PRRT2基因的表达单位(比如表达载体或病毒等)递送到靶点上,并使之表达(较佳为过表达)活性的PRRT2;或者,也可以通过体内基因编辑的方式(如CRISPR-Cas9基因编辑方法)增强前脑兴奋性神经元内源PRRT2的表达,从而达到治疗或预防癫痫的目的。After knowing the use of the PRRT2, the PRRT2 or its up-regulator can be administered to the subject by various methods well known in the art. Preferably, gene therapy can be used, such as delivering an expression unit (such as an expression vector or virus, etc.) carrying the PRRT2 gene to the target through a certain route, and causing it to express (preferably overexpress) active PRRT2; or, the expression of endogenous PRRT2 in forebrain excitatory neurons can be enhanced by in vivo gene editing (such as CRISPR-Cas9 gene editing method), thereby achieving the purpose of treating or preventing epilepsy.
术语“有效量”是指可以实现期望结果(例如预防或治疗结果)的量。本发明所述的PRRT2或其上调剂的有效量可随给药的模式和待治疗的疾病的严重程度等而变化。优选的有效量的选择可以由本领域普通技术人员根据各种因素来确定(例如通过临床试验)。所述的因素包括但不限于:所述的PRRT2或其上调剂的药代动力学参数例如生物利用率、代谢、半衰期等;患者所要治疗的疾病的严重程度、患者的体重、患者的免疫状况、给药的途径等。例如,由治疗状况的迫切要求,可每天给予若干次分开的剂量,或将剂量按比例地减少。The term "effective amount" refers to an amount that can achieve a desired result (e.g., a preventive or therapeutic result). The effective amount of PRRT2 or its up-regulator described in the present invention may vary depending on the mode of administration and the severity of the disease to be treated. The selection of the preferred effective amount can be determined by a person of ordinary skill in the art based on various factors (e.g., through clinical trials). The factors include, but are not limited to: pharmacokinetic parameters of the PRRT2 or its up-regulator such as bioavailability, metabolism, half-life, etc.; the severity of the disease to be treated by the patient, the patient's weight, the patient's immune status, the route of administration, etc. For example, depending on the urgency of the treatment situation, several divided doses may be given per day, or the dose may be reduced proportionally.
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件如J.萨姆布鲁克等编著,分子克隆实验指南,科学出版社中所述的条件,或按照制造厂商所建议的条件。The present invention is further described below in conjunction with specific examples. It should be understood that these examples are only used to illustrate the present invention and are not intended to limit the scope of the present invention. The experimental methods in the following examples where specific conditions are not specified are usually carried out according to conventional conditions such as those described in J. Sambrook et al., Molecular Cloning Experiment Guide, Science Press, or according to the conditions recommended by the manufacturer.
材料和方法Materials and methods
1、实验材料1. Experimental Materials
小鼠PRRT2质粒(Origene,#MR214094)。Mouse PRRT2 plasmid (Origene, #MR214094).
人类PRRT2质粒(Origene,#RC202304)。Human PRRT2 plasmid (Origene, #RC202304).
斑马鱼PRRT2质粒(实验室构建,克隆自成年斑马鱼脑组织cDNA库)。Zebrafish PRRT2 plasmid (laboratory constructed, cloned from adult zebrafish brain tissue cDNA library).
pCAG2IG质粒(Addgene,#122292)。 pCAG2IG plasmid (Addgene, #122292).
Lipofectamine 3000转染试剂盒(ThermoFisher Scientific,#L3000001)。Lipofectamine 3000 transfection kit (ThermoFisher Scientific, #L3000001).
Nav1.2稳转细胞株(北京爱思益普生物科技股份有限公司,#ICE-Nav1.2-HEK)。Nav1.2 stably transfected cell line (Beijing Ice-Eye Biotechnology Co., Ltd., #ICE-Nav1.2-HEK).
二氧化碳恒温培养箱(ThermoFisher Scientific,#3111)。CO2 constant temperature incubator (ThermoFisher Scientific, #3111).
膜片钳记录系统(HEKA,#EPCI0)。Patch clamp recording system (HEKA, #EPCIO).
细胞外液:140mM NaCl,3.5mM KCl,1mM MgCl2,2mM CaCl2,10mM D-Glucose,1.25mM NaH2PO4,10mM HEPES(pH=7.4NaOH调节)。Extracellular fluid: 140mM NaCl, 3.5mM KCl, 1mM MgCl2, 2mM CaCl2, 10mM D-Glucose, 1.25mM NaH2PO4, 10mM HEPES (pH=7.4 adjusted with NaOH).
电极内液:50mM CsCl,10mM NaCl,10mM HEPES,20mM EGTA,60mM CsF,pH=7.2(CsOH)。Electrode solution: 50mM CsCl, 10mM NaCl, 10mM HEPES, 20mM EGTA, 60mM CsF, pH=7.2 (CsOH).
AAV-CAG-DIO-mCherry质粒(图16)(中国科学院脑科学与智能技术卓越创新中心基因编辑平台提供)。AAV-CAG-DIO-mCherry plasmid (Figure 16) (provided by the Gene Editing Platform of Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences).
pAAV-CAG-DIO-mouse PRRT2-HA质粒(图17)(中国科学院脑科学与智能技术卓越创新中心基因编辑平台提供)。pAAV-CAG-DIO-mouse PRRT2-HA plasmid (Figure 17) (provided by the Gene Editing Platform of Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences).
抗PRRT2抗体(Wiiget Biotech,#Rp3246-a)。Anti-PRRT2 antibody (Wiiget Biotech, #Rp3246-a).
抗HA抗体(ROAHAHA Roche,#11867423)。Anti-HA antibody (ROAHAHA Roche, #11867423).
本发明所使用的AAV病毒由中国科学院脑科学与智能技术卓越创新中心基因编辑平台生产提供:The AAV virus used in the present invention is produced and provided by the gene editing platform of the Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences:
AAV-PHP.eB-CAG-DIO-mouse PRRT2-HA;AAV-PHP.eB-CAG-DIO-mouse PRRT2-HA;
AAV-PHP.eB-CAG-DIO-mCherry;AAV-PHP.eB-CAG-DIO-mCherry;
戊四唑(Pentylenetetrazol,PTZ)(Sigma-Aldrich,#P6500-25G)。Pentylenetetrazol (PTZ) (Sigma-Aldrich, #P6500-25G).
C57BL/6J小鼠,购自上海斯莱克实验动物有限责任公司。C57BL/6J mice were purchased from Shanghai Slake Laboratory Animal Co., Ltd.
Emx1-cre小鼠,名称:B6.129S2-Emx1tm1(cre)Krj/J,来自Jackson Lab,货号:JAX货号#005628。Emx1-cre mice, name: B6.129S2-Emx1tm1(cre)Krj/J, from Jackson Lab, catalog number: JAX catalog number #005628.
Nkx2.1-cre小鼠,名称:C57BL/6J-Tg(Nkx2-1-cre)2Sand/J,来自Jackson Lab,货号:JAX货号#008661。Nkx2.1-cre mice, name: C57BL/6J-Tg(Nkx2-1-cre)2Sand/J, from Jackson Lab, catalog number: JAX catalog number #008661.
2、实验动物饲养2. Animal husbandry
每笼饲养3-6只小鼠,小鼠可在笼内自由获取食物和水,饲养期间12小时光照/黑暗循环,22-23℃,湿度50-60%。Three to six mice were housed in each cage with free access to food and water in the cages under a 12-h light/dark cycle, 22-23°C, and 50-60% humidity.
所有实验动物的操作和使用均符合中国科学院脑科学与智能技术卓越创新中心动物管理委员会对于实验动物操作和动物福利的要求。All operations and use of experimental animals complied with the requirements of the Animal Care Committee of the Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences on experimental animal operations and animal welfare.
3、眶静脉注射AAV病毒3. Orbital vein injection of AAV virus
取出-80摄氏度保存的AAV病毒,冰上融解。用预冷的无菌PBS稀释AAV病毒至注射用浓度(2×1012vg/mL,vg:AAV vector genomes)。注射前将AAV病毒溶液平衡至室温,用0.3 mL体积胰岛素针吸取AAV病毒溶液50μL准备注射。Take out the AAV virus stored at -80 degrees Celsius and thaw it on ice. Dilute the AAV virus to the injection concentration (2×10 12 vg/mL, vg: AAV vector genomes) with pre-cooled sterile PBS. Equilibrate the AAV virus solution to room temperature before injection and add 0.3 Aspirate 50 μL of AAV virus solution into a mL insulin needle for injection.
注射戊巴比妥钠溶液(90mg/kg,腹腔注射)麻醉小鼠。待小鼠麻醉后,手指固定小鼠头部,且避免按压小鼠动脉,撑开眼部皮肤充分暴露眼球。持胰岛素针从眼角处呈20°角插入,进入眼眶内部时避免伤及眼球。注射针针尖进入眼眶2mm深度到达眶静脉丛,缓慢注射50μL。注射结束后缓慢退针,拭去眼角多余液体和血液。在另一侧眶静脉进行相同的注射,两侧共计注射总滴度为2×1011vg的AAV病毒。Mice were anesthetized by injecting sodium pentobarbital solution (90 mg/kg, intraperitoneal injection). After the mouse was anesthetized, the mouse head was fixed with fingers, and the mouse artery was avoided to be pressed. The eye skin was stretched to fully expose the eyeball. The insulin needle was inserted from the corner of the eye at a 20° angle, and the eyeball was avoided when entering the orbit. The needle tip entered the orbit to a depth of 2 mm to reach the orbital venous plexus, and 50 μL was slowly injected. After the injection, the needle was slowly withdrawn and excess fluid and blood in the corner of the eye were wiped off. The same injection was performed in the orbital vein on the other side, and a total titer of 2×10 11 vg of AAV virus was injected on both sides.
注射完毕后将小鼠放回笼盒内,待其苏醒后送回饲养间。After the injection, the mice were placed back in the cages and sent back to the feeding room after they woke up.
病毒表达两周后进行电极植入手术,并在病毒表达三周后开展癫痫评价实验。Electrode implantation surgery was performed two weeks after viral expression, and epilepsy evaluation experiments were performed three weeks after viral expression.
4、EEG/EMG电极植入及脑电记录4. EEG/EMG electrode implantation and EEG recording
先用4%的异氟烷麻醉实验小鼠,然后将小鼠头部固定于立体定位仪适配器。在后续整个手术期间将异氟烷浓度调整为1.5%,使小鼠处于持续麻醉状态。另外,用金霉素眼膏保护小鼠眼部,避免干燥导致视网膜损伤。使用脱毛膏将小鼠头部毛发去除,沿中线切开头部皮肤,暴露颅骨表面。使用2%的过氧化氢溶液处理手术切口及颅骨表面,并刮除颅骨表面的结缔组织。用组织胶(Vetbond,3M Deutschland Gmbh)将暴露颅骨四周的皮肤进行固定,并再次清理颅骨表面的膜系。通过立体定位的方式标记电极植入位置,坐标AP-0.8mm,ML±1.1mm。待颅骨表面干燥后,用光固化自蚀粘合剂(3M ESPE Single Bound universal)覆盖颅骨表面,形成固化层。使用小型手持颅钻在植入位置标记处开孔,圆孔直径约为0.6mm。小心清理开孔处的骨屑,并保持大脑皮层表面的湿润,然后将两个无菌处理的ECoG记录电极(直径0.5mm)分别沿颅骨开孔处垂直插入大脑皮层0.35mm深度(以大脑皮层表面为参考),用牙科水泥固定记录电极和电极接口部件。为了便于小鼠清醒状态的电生理记录,在小鼠头部靠近电极接口的位置用牙科水泥额外固定一片轻质的钛合金片(30mm×2mm×1mm)。用于记录肌电信号的两条金属丝电极从电极接口部件底部分别延伸到小鼠颈部,将1mm电极裸露端分别插入颈部两侧的肌肉层,固定后缝合颈部皮肤并消毒。小鼠手术后恢复一周。The experimental mice were first anesthetized with 4% isoflurane, and then the mouse heads were fixed to the stereotaxic adapter. The isoflurane concentration was adjusted to 1.5% during the entire subsequent surgery to keep the mice in a state of continuous anesthesia. In addition, chlortetracycline eye ointment was used to protect the mouse eyes to avoid retinal damage caused by dryness. The hair on the head of the mouse was removed with a depilatory cream, and the head skin was cut along the midline to expose the skull surface. The surgical incision and skull surface were treated with a 2% hydrogen peroxide solution, and the connective tissue on the skull surface was scraped off. The skin around the exposed skull was fixed with tissue glue (Vetbond, 3M Deutschland Gmbh), and the membrane system on the skull surface was cleaned again. The electrode implantation position was marked by stereotaxic method, with coordinates AP-0.8mm, ML±1.1mm. After the skull surface was dry, the skull surface was covered with a light-cured self-etching adhesive (3M ESPE Single Bound universal) to form a solidified layer. A small handheld cranial drill was used to open a hole at the implantation position mark, with a circular hole diameter of approximately 0.6mm. Carefully clean the bone debris at the opening and keep the surface of the cerebral cortex moist. Then, two sterile ECoG recording electrodes (0.5 mm in diameter) were vertically inserted into the cerebral cortex at a depth of 0.35 mm (with the surface of the cerebral cortex as a reference) along the skull opening, and the recording electrodes and electrode interface components were fixed with dental cement. In order to facilitate electrophysiological recording of mice in the awake state, a lightweight titanium alloy sheet (30 mm × 2 mm × 1 mm) was additionally fixed with dental cement at the position of the mouse head near the electrode interface. The two metal wire electrodes used to record electromyographic signals extended from the bottom of the electrode interface component to the neck of the mouse, and the exposed ends of the 1 mm electrodes were inserted into the muscle layers on both sides of the neck, and the neck skin was sutured and disinfected after fixation. The mice recovered for one week after surgery.
进行清醒状态ECoG/EMG记录前,利用小鼠头部的金属片将小鼠固定于记录台,使小鼠适应此头部固定状态30分钟。ECoG和EMG信号经由差分放大器(Model 3000,A-M system)放大100倍,然后分别经过波段滤波后(ECoG 1-300Hz,EMG 10-1000Hz)由数模转换器(Digidata 1332A,Molecular Devices)进行采集(1000Hz)和记录。Before ECoG/EMG recording in the awake state, the mouse was fixed to the recording platform using a metal plate on the mouse's head, and the mouse was allowed to adapt to this head-fixed state for 30 minutes. The ECoG and EMG signals were amplified 100 times by a differential amplifier (Model 3000, A-M system), and then band-filtered (ECoG 1-300Hz, EMG 10-1000Hz) and collected (1000Hz) and recorded by a digital-to-analog converter (Digidata 1332A, Molecular Devices).
5、PTZ诱导癫痫小鼠模型5. PTZ-induced epilepsy mouse model
测试前用生理盐水配制3.5mg/mL(Nkx2.1-cre小鼠实验)或4.5mg/mL(Emx1-cre小鼠实验)的戊四唑溶液。根据小鼠体重,以35mg/kg或45mg/kg的剂量腹腔注射戊四唑,注射结束后立即将小鼠放入行为箱内进行视频记录,观察小鼠在给药后30分钟内的癫痫发作状况。Before the test, 3.5 mg/mL (Nkx2.1-cre mouse experiment) or 4.5 mg/mL (Emx1-cre mouse experiment) of pentylenetetrazol solution was prepared with normal saline. According to the weight of mice, pentylenetetrazol was injected intraperitoneally at a dose of 35 mg/kg or 45 mg/kg. Immediately after the injection, the mice were placed in a behavior box for video recording to observe the epileptic seizures of the mice within 30 minutes after administration.
小鼠癫痫评级标准: Mouse epilepsy rating criteria:
一级:小鼠伏地不动,腹部贴地爬行;Level 1: The mouse lies motionless on the ground, crawling on its belly;
二级:突发性抽搐,行为突然停止,尾巴翘起;Level 2: Sudden convulsions, sudden cessation of behavior, and raised tail;
三级:肌肉阵挛,头部扭转,抬手抽搐;Grade 3: Myoclonus, head twisting, hand twitching;
四级:强直-阵挛,倒地抽搐,疯跳疯跑;Grade 4: Tonic-clonic, falling to the ground and twitching, jumping and running wildly;
五级:倒地并发展至四肢强直性伸直;Grade 5: Falling to the ground and developing tonic extension of limbs;
六级:死亡。Level 6: Death.
6、免疫组化检测PRRT2在脑内的表达6. Immunohistochemical detection of PRRT2 expression in the brain
将小鼠用戊巴比妥钠深度麻醉后,经心脏灌注10mL常温磷酸盐缓冲溶液(PBS)冲洗血液,然后灌注10mL 4℃的4%多聚甲醛溶液(polyformaldehyde,PFA)进行组织固定。解剖并取出小鼠脑组织,放入4%多聚甲醛溶液中后固定过夜。然后将脑组织放入含30%蔗糖的PBS中脱水2天,并在24小时后更换一次蔗糖溶液。脱水结束后,脑组织用冷冻切片机(CM1950,Leica)在-20℃进行矢状(sagittal)切片,厚度为25μm,脑片收集于PBS溶液中,4℃保存。利用抗PRRT2抗体进行常规免疫组化检测。After the mice were deeply anesthetized with sodium pentobarbital, 10 mL of room temperature phosphate buffer solution (PBS) was perfused through the heart to flush the blood, and then 10 mL of 4% polyformaldehyde solution (PFA) at 4°C was perfused for tissue fixation. The mouse brain tissue was dissected and removed, placed in a 4% paraformaldehyde solution and fixed overnight. The brain tissue was then placed in PBS containing 30% sucrose for dehydration for 2 days, and the sucrose solution was replaced once after 24 hours. After dehydration, the brain tissue was sagittal sliced at -20°C with a thickness of 25 μm using a freezing microtome (CM1950, Leica), and the brain slices were collected in a PBS solution and stored at 4°C. Conventional immunohistochemistry was performed using anti-PRRT2 antibodies.
7、序列信息7. Sequence information
(1)小鼠Prrt2基因(编码蛋白区域,CDS)序列(SEQ ID NO:1):
(1) Mouse Prrt2 gene (protein coding region, CDS) sequence (SEQ ID NO: 1):
(2)小鼠PRRT2蛋白氨基酸序列(346个氨基酸)(SEQ ID NO:2):
(2) Mouse PRRT2 protein amino acid sequence (346 amino acids) (SEQ ID NO: 2):
(3)人Prrt2基因(编码蛋白区域,CDS)序列(SEQ ID NO:3):
(3) Human Prrt2 gene (protein coding region, CDS) sequence (SEQ ID NO: 3):
(4)人PRRT2蛋白氨基酸序列(340个氨基酸)(SEQ ID NO:4):
(4) Human PRRT2 protein amino acid sequence (340 amino acids) (SEQ ID NO: 4):
(5)斑马鱼Prrt2基因(编码蛋白区域,CDS)序列(SEQ ID NO:5):
(5) Zebrafish Prrt2 gene (protein coding region, CDS) sequence (SEQ ID NO: 5):
(6)斑马鱼PRRT2蛋白氨基酸序列(226个氨基酸)(SEQ ID NO:6):
(6) Zebrafish PRRT2 protein amino acid sequence (226 amino acids) (SEQ ID NO: 6):
实施例1、验证过表达PRRT2对钠离子通道慢失活的作用Example 1: Verification of the effect of overexpression of PRRT2 on slow inactivation of sodium channels
将小鼠PRRT2蛋白编码区cDNA序列(图1和图2)插入pCAG2IG质粒(即EGFP对照质粒,图3)的多克隆位点区,构建小鼠PRRT2过表达质粒pCAG-mouse PRRT2(HA)-IRES-EGFP(图4)。The cDNA sequence of the mouse PRRT2 protein coding region (Figures 1 and 2) was inserted into the multiple cloning site region of the pCAG2IG plasmid (i.e., the EGFP control plasmid, Figure 3) to construct the mouse PRRT2 overexpression plasmid pCAG-mouse PRRT2(HA)-IRES-EGFP (Figure 4).
以同样的方式,构建人源PRRT2过表达质粒pCAG-human PRRT2(HA)-IRES-EGFP(图5、图6和图7)。In the same way, the human PRRT2 overexpression plasmid pCAG-human PRRT2(HA)-IRES-EGFP was constructed (Figures 5, 6 and 7).
以同样的方式,构建斑马鱼源PRRT2过表达质粒pCAG-zebra PRRT2(HA)-IRES-EGFP(图8、图9和图10)。In the same way, the zebrafish PRRT2 overexpression plasmid pCAG-zebra PRRT2(HA)-IRES-EGFP was constructed (Figures 8, 9 and 10).
在稳转Nav1.2的HEK293细胞中,利用Lipo3000试剂转染PRRT2过表达质粒或对照质粒,37℃表达24小时后将细胞铺种至圆形玻片上,继续表达12小时。In HEK293 cells stably transfected with Nav1.2, PRRT2 overexpression plasmid or control plasmid was transfected using Lipo3000 reagent. After expression at 37°C for 24 hours, the cells were plated on round glass slides and continued to express for 12 hours.
钠通道电流的记录采用全细胞记录(whole-cell recording)的方式(图11A)。首先,用微电极拉制仪(P97,Sutter Instruments)将毛细玻璃管(BF150-86-10,Sutter Instruments)拉制成记录 电极。在倒置显微镜(MF53,Micro-shot)下操纵微电极操纵仪(MP225,Butter Instrument)将记录电极接触到细胞表面,给予负压抽吸,形成吉欧姆(GΩ)封接。形成吉欧姆封接后进行快速电容补偿,然后继续给予负压,吸破细胞膜,形成全细胞记录模式。然后进行慢速电容的补偿并记录膜电容及串联电阻。所有电生理实验均在室温下进行。实验数据由EPC-10放大器(HEKA)进行采集并储存于PatchMaster(HEKA)软件中。Sodium channel currents were recorded by whole-cell recording (Figure 11A). First, a capillary glass tube (BF150-86-10, Sutter Instruments) was pulled into a recording electrode using a microelectrode puller (P97, Sutter Instruments). Electrode. Under an inverted microscope (MF53, Micro-shot), the microelectrode manipulator (MP225, Butter Instrument) was manipulated to contact the recording electrode to the cell surface, and negative pressure was applied to form a GΩ seal. After the GΩ seal was formed, fast capacitance compensation was performed, and then negative pressure was continued to be applied to break the cell membrane and form a whole-cell recording mode. Then slow capacitance compensation was performed and membrane capacitance and series resistance were recorded. All electrophysiological experiments were performed at room temperature. The experimental data were collected by an EPC-10 amplifier (HEKA) and stored in the PatchMaster (HEKA) software.
钠离子通道快失活状态的进入和恢复采用程序1:快失活(fast inactivation,图11B)和程序2:从快失活恢复(Recover from fast inactivation,图12A)进行测试。钠离子通道慢失活状态的进入和恢复分别采用程序3:进入慢失活(Entry in slow inactivation,图13A)和程序4:从慢失活恢复(Recover from slow inactivation,图14A)进行记录。The entry and recovery of the fast inactivation state of the sodium ion channel were tested using Procedure 1: fast inactivation (Figure 11B) and Procedure 2: Recover from fast inactivation (Figure 12A). The entry and recovery of the slow inactivation state of the sodium ion channel were recorded using Procedure 3: Entry in slow inactivation (Figure 13A) and Procedure 4: Recover from slow inactivation (Figure 14A), respectively.
人PRRT2和斑马鱼PRRT2对钠离子通道慢失活作用的测试方案与上述小鼠PRRT2的测试方案类同,但在检测钠离子通道从慢失活恢复的过程中,发明人采用连续测试方案取代重复扫描测试法以改善细胞电生理记录的成功率。The testing scheme for the effect of human PRRT2 and zebrafish PRRT2 on the slow inactivation of sodium channels is similar to the testing scheme for mouse PRRT2 mentioned above, but in the process of detecting the recovery of sodium channels from slow inactivation, the inventors used a continuous testing scheme instead of the repeated scanning test method to improve the success rate of cell electrophysiological recording.
在记录过程中,细胞的初始钳制电压为-120mV,此时钠离子通道处于关闭状态或称静息状态。当钳制电压从-120mV升至0mV时,细胞发生去极化,钠离子通道开放,细胞外钠离子迅速内流形成钠电流(图11B)。随后(约0.5-1ms)钠离子通道开始进入失活状态(快失活),通道关闭,钠电流衰减(图11B和图11C)。钠离子通道在短暂的去极化时程内进入快失活状态,并可以在十至数十毫秒超极化过程中恢复至可激活的静息态(图12A和图12B)。During the recording process, the initial clamping voltage of the cell is -120mV, at which time the sodium ion channel is in a closed state or a resting state. When the clamping voltage rises from -120mV to 0mV, the cell depolarizes, the sodium ion channel opens, and extracellular sodium ions rapidly flow inward to form a sodium current (Figure 11B). Subsequently (about 0.5-1ms), the sodium ion channel begins to enter an inactivated state (fast inactivation), the channel closes, and the sodium current decays (Figures 11B and 11C). The sodium ion channel enters a fast inactivation state within a short depolarization period, and can be restored to an activatable resting state during a hyperpolarization process of ten to tens of milliseconds (Figures 12A and 12B).
结果显示,小鼠PRRT2过表达对钠离子通道快失活的形成和从快失活恢复均无显著影响(图11-图12)。The results showed that overexpression of mouse PRRT2 had no significant effect on the formation of fast inactivation of sodium ion channels and the recovery from fast inactivation ( FIGS. 11-12 ).
除了快失活机制,钠离子通道还具有慢失活的特性。钠离子通道进入慢失活状态或从慢失活状态恢复都需要比快失活更长时间,通常为几百毫秒至数十秒。根据以上规律,在去极化过程结束后通过10毫秒的短暂超极化,可以使快失活状态的钠离子通道得到恢复,从而有效分离出钠离子通道的慢失活成分(图13A)。In addition to the fast inactivation mechanism, sodium channels also have the characteristics of slow inactivation. It takes longer for sodium channels to enter the slow inactivation state or recover from the slow inactivation state than the fast inactivation state, usually hundreds of milliseconds to tens of seconds. According to the above rules, after the depolarization process is completed, a brief hyperpolarization of 10 milliseconds can restore the fast inactivation state of sodium channels, thereby effectively separating the slow inactivation component of the sodium channel (Figure 13A).
结果显示,小鼠PRRT2过表达促进了钠离子通道在去极化过程中进入慢失活状态的速度(图13B),并且延长了钠离子通道从慢失活恢复至可激活静息态的时间(图14A和图14B)。The results showed that overexpression of mouse PRRT2 promoted the speed at which sodium ion channels entered the slow inactivation state during depolarization ( FIG. 13B ), and prolonged the time it took for sodium ion channels to recover from the slow inactivation state to the activatable resting state ( FIG. 14A and FIG. 14B ).
人源PRRT2对钠离子通道慢失活的调控效果与小鼠PRRT2类似(图15A-图15C)。The regulatory effect of human PRRT2 on slow inactivation of sodium channels is similar to that of mouse PRRT2 ( Figure 15A-Figure 15C ).
PRRT2在较多的物种中具有保守性。令人意外的是,前期工作中,发明人针对多种多样的不同物种来源的PRRT2对钠离子通道慢失活作用过程中,发现斑马鱼来源的PRRT2对钠离子通道慢失活恢复的抑制作用(与小鼠和人源的PRRT2相比斑马鱼PRRT2过表达使得钠离子通道慢失活恢复时间更长,可以理解为对钠离子通道慢失活具有更强的作用)显著强于人来源和小鼠来源的PRRT2,图15C即为斑马鱼与人源以及小鼠PRRT2的比较结果。PRRT2 is conserved in many species. Surprisingly, in the previous work, the inventors found that the inhibitory effect of zebrafish PRRT2 on the recovery of sodium channel slow inactivation (compared with mouse and human PRRT2, zebrafish PRRT2 overexpression makes the recovery time of sodium channel slow inactivation longer, which can be understood as having a stronger effect on sodium channel slow inactivation) was significantly stronger than that of human and mouse PRRT2. Figure 15C is the comparison result of zebrafish, human and mouse PRRT2.
综上所述,PRRT2过表达可以促进钠离子通道的慢失活,但对钠离子通道快失活影响不显著。因此,PRRT2具有钠离子通道状态依赖性调控特征,更倾向于减少持续去极化状态下细胞钠离子通道的有效供给。PRRT2的该特征使之在不影响细胞正常兴奋性活动的情况下, 降低细胞的异常兴奋性,使其具有良好的细胞状态选择性。In summary, PRRT2 overexpression can promote the slow inactivation of sodium channels, but has no significant effect on the fast inactivation of sodium channels. Therefore, PRRT2 has the characteristics of sodium channel state-dependent regulation, and is more inclined to reduce the effective supply of cell sodium channels under continuous depolarization. This feature of PRRT2 allows it to reduce the effective supply of cell sodium channels under continuous depolarization without affecting the normal excitatory activity of cells. Reduce the abnormal excitability of cells, giving them good cell state selectivity.
实施例2、在前脑兴奋性神经元过表达PRRT2干预癫痫发作Example 2: Overexpression of PRRT2 in forebrain excitatory neurons to intervene in epileptic seizures
为了检测PRRT2蛋白的表达和分布,发明人用抗PRRT2抗体和抗HA标签抗体对脑片样本进行了免疫荧光染色。抗PRRT2抗体用于检测包括内源性和过表达在内的PRRT2蛋白,而抗HA抗体仅能检测过表达的带有HA标签的PRRT2蛋白。In order to detect the expression and distribution of PRRT2 protein, the inventors used anti-PRRT2 antibody and anti-HA tag antibody to perform immunofluorescence staining on brain slice samples. Anti-PRRT2 antibody is used to detect PRRT2 protein including endogenous and overexpressed, while anti-HA antibody can only detect overexpressed PRRT2 protein with HA tag.
首先将脑片用PBS溶液漂洗三遍,每遍10分钟,然后用含0.3%Triton X-100和5%牛血清蛋白(bovine serum protein,BSA;溶于PBS)的封闭液室温孵育1小时。封闭结束后进行一抗孵育,脑片样本在抗PRRT2抗体(1:500,rabbit)溶液中4℃孵育过夜。脑片用PBS溶液漂洗三遍,然后进行二抗孵育。脑片样本在荧光二抗(1:2000,donkey anti-rabbit-647nm,donkey anti-rat 488nm)和Hoechst(1:5000)溶液中室温孵育2小时。用PBS溶液漂洗三遍后将脑片贴附于载玻片,晾干后滴加含80%甘油的PBS溶液并封片。用荧光显微镜(VS-120,Olympus)在10倍物镜下进行观察和拍摄。First, the brain slices were rinsed three times with PBS solution for 10 minutes each time, and then incubated with blocking solution containing 0.3% Triton X-100 and 5% bovine serum protein (BSA; dissolved in PBS) at room temperature for 1 hour. After the blocking was completed, the primary antibody was incubated, and the brain slice samples were incubated in anti-PRRT2 antibody (1:500, rabbit) solution at 4°C overnight. The brain slices were rinsed three times with PBS solution and then incubated with secondary antibodies. The brain slice samples were incubated in fluorescent secondary antibodies (1:2000, donkey anti-rabbit-647nm, donkey anti-rat 488nm) and Hoechst (1:5000) solutions at room temperature for 2 hours. After rinsing three times with PBS solution, the brain slices were attached to the slides, and after drying, PBS solution containing 80% glycerol was added and the slides were sealed. The images were observed and photographed under a 10x objective lens using a fluorescence microscope (VS-120, Olympus).
腺相关病毒(AAV)是最常用的在体内进行基因递送的载体。本实验选择PHP.eB血清型AAV病毒作为PRRT2基因的递送载体,实验中显示,其具有良好血脑屏障穿透能力、有助于PRRT2基因在脑内的表达。Adeno-associated virus (AAV) is the most commonly used vector for gene delivery in vivo. In this experiment, PHP.eB serotype AAV virus was selected as the delivery vector of PRRT2 gene. The experiment showed that it has good blood-brain barrier penetration ability and is conducive to the expression of PRRT2 gene in the brain.
PRRT2蛋白的过表达采用cre重组酶依赖的方式,通过在病毒表达载体中设置DIO系统(Loxp/Lox2272)来实现cre重组酶依赖的表达(图17,pAAV-CAG-DIO-mouse PRRT2-HA)。间接地依赖Emx1启动子(前脑兴奋性神经元特异性表达启动子)驱动的cre重组酶表达来实现在前脑兴奋性神经元内的PRRT2蛋白的特异性表达。Overexpression of PRRT2 protein was achieved by Cre recombinase-dependent method, by setting up DIO system (Loxp/Lox2272) in viral expression vector to achieve Cre recombinase-dependent expression (Figure 17, pAAV-CAG-DIO-mouse PRRT2-HA). Indirectly relying on Emx1 promoter (forebrain excitatory neuron-specific expression promoter)-driven Cre recombinase expression to achieve specific expression of PRRT2 protein in forebrain excitatory neurons.
同时,由于实验动物为小鼠,本发明人运用特异性表达cre重组酶的小鼠品系(emx1-cre),该品系的小鼠,cre重组酶的表达依赖Emx1启动子。At the same time, since the experimental animals are mice, the inventors used a mouse strain (emx1-cre) that specifically expresses cre recombinase. In this strain of mice, the expression of cre recombinase depends on the Emx1 promoter.
实验结果显示,在Emx1-cre小鼠中,眶静脉注射总量2×1011vg的AAV-PHP.eB-CAG-DIO-mCherry病毒,可以实现mCherry荧光蛋白在前脑兴奋性神经元中的过表达。注射相同总量的AAV-PHP.eB-CAG-DIO-mouse PRRT2-HA病毒,则可观察到前脑兴奋性神经元中PRRT2蛋白的过表达(图18A-图18C)。The experimental results showed that in Emx1-cre mice, orbital vein injection of a total of 2×10 11 vg of AAV-PHP.eB-CAG-DIO-mCherry virus could achieve overexpression of mCherry fluorescent protein in forebrain excitatory neurons. Injection of the same total amount of AAV-PHP.eB-CAG-DIO-mouse PRRT2-HA virus could observe overexpression of PRRT2 protein in forebrain excitatory neurons (Figure 18A-Figure 18C).
与对照组小鼠相比,过表达PRRT2组的小鼠在PTZ诱导的癫痫模型中表现出更强的抗癫痫能力(图19A)。Compared with the control group mice, the mice in the PRRT2 overexpression group showed stronger anti-epileptic ability in the PTZ-induced epilepsy model ( FIG. 19A ).
在基于癫痫行为等级的评价中,前脑兴奋性神经元过表达PRRT2组小鼠在腹腔注射PTZ(45mg/kg)之后,平均发作等级为1.4级,而对照组小鼠的平均发作等级为4级,两组间的癫痫发作等级存在非常显著的统计学差异(图19B)。In the evaluation based on epileptic behavior grade, after intraperitoneal injection of PTZ (45 mg/kg), the average seizure grade of mice in the forebrain excitatory neuron overexpression group was 1.4, while the average seizure grade of mice in the control group was 4. There was a very significant statistical difference in the epileptic seizure grade between the two groups (Figure 19B).
脑电记录的结果也显示,前脑兴奋性神经元过表达PRRT2组小鼠在注射PTZ之后癫痫样脑电信号明显弱于对照组小鼠(图20A和图20B)。The results of EEG recording also showed that the epileptic-like EEG signals of mice in the group overexpressing PRRT2 in forebrain excitatory neurons after PTZ injection were significantly weaker than those of mice in the control group ( FIGS. 20A and 20B ).
与对照组小鼠相比,兴奋性神经元过表达PRRT2的小鼠在旷场实验、转轮实验以及社交 实验中均未表现出显著的行为异常(图21A-图21C)。Compared with control mice, mice whose excitatory neurons overexpressed PRRT2 showed significantly higher performance in the open field test, wheel running test, and social interaction test. No significant behavioral abnormalities were observed in the experiments (Figure 21A-21C).
以同样的方式,在Emx1-cre小鼠中,眶静脉注射总量2×1011vg的AAV-PHP.eB-CAG-DIO-mCherry病毒(对照)或AAV-PHP.eB-CAG-DIO-zebra PRRT2病毒。In the same manner, Emx1-cre mice were injected intraorbitally with a total of 2×10 11 vg of AAV-PHP.eB-CAG-DIO-mCherry virus (control) or AAV-PHP.eB-CAG-DIO-zebra PRRT2 virus.
结果如表1所示,令人意外地,前脑兴奋性神经元过表达斑马鱼PRRT2能够极为显著地抑制PTZ诱导的癫痫发作。斑马鱼来源的PRRT2呈现比其它物种来源显著更优的效果。The results are shown in Table 1. Surprisingly, overexpression of zebrafish PRRT2 in forebrain excitatory neurons can significantly inhibit PTZ-induced epileptic seizures. Zebrafish-derived PRRT2 exhibits significantly better effects than PRRT2 from other species.
表1.前脑兴奋性神经元过表达斑马鱼PRRT2抑制惊厥剂诱导的癫痫发作
Table 1. Overexpression of zebrafish PRRT2 in forebrain excitatory neurons inhibits convulsant-induced epileptic seizures
实施例3、非前脑兴奋性神经元中过表达PRRT2的影响Example 3: Effects of overexpressing PRRT2 in non-forebrain excitatory neurons
为了验证选择性表达PRRT2的重要性,发明人测试了在非前脑兴奋性神经元中过表达PRRT2,分析是否引发不良反应。To verify the importance of selective expression of PRRT2, the inventors tested whether overexpression of PRRT2 in non-forebrain excitatory neurons would induce adverse reactions.
在Nkx2.1-cre小鼠中,cre重组酶的表达依赖Nkx2.1基因启动子,该启动子驱动cre重组酶选择性表达于前脑抑制性神经元(即γ氨基丁酸能神经元)。发明人利用Nkx2.1-cre小鼠,采用腺相关病毒递送的策略,眶静脉注射总量2×1011vg的AAV-PHP.eB-CAG-DIO-mouse PRRT2-HA病毒,进行前脑抑制型神经元的PRRT2过表达(图22A和图22B)。In Nkx2.1-cre mice, the expression of cre recombinase depends on the Nkx2.1 gene promoter, which drives the selective expression of cre recombinase in forebrain inhibitory neurons (i.e., γ-aminobutyric acid neurons). The inventors used Nkx2.1-cre mice and adopted a strategy of adeno-associated virus delivery to inject a total of 2×10 11 vg of AAV-PHP.eB-CAG-DIO-mouse PRRT2-HA virus into the orbital vein to overexpress PRRT2 in forebrain inhibitory neurons (Figures 22A and 22B).
发明人发现,在NKX2.1-cre小鼠中,与对照组小鼠相比,抑制型神经元过表达PRRT2组的小鼠在PTZ诱导的癫痫模型中表现出癫痫易感(图22D和图22E)。抑制型神经元过表达PRRT2组小鼠在腹腔注射阈下剂量PTZ(35mg/kg)之后,癫痫发作等级高于对照组小鼠(图22D和图22E)。脑电记录的结果也显示,前脑抑制性神经元过表达PRRT2组小鼠在注射阈下剂量PTZ之后癫痫样脑电信号明显强于对照组小鼠(图22F和图22G)。The inventors found that in NKX2.1-cre mice, compared with control mice, mice in the inhibitory neuron overexpression PRRT2 group showed epilepsy susceptibility in the PTZ-induced epilepsy model (Figure 22D and Figure 22E). After intraperitoneal injection of subthreshold doses of PTZ (35 mg/kg), mice in the inhibitory neuron overexpression PRRT2 group had higher epileptic seizure levels than mice in the control group (Figure 22D and Figure 22E). The results of EEG recording also showed that the epileptic EEG signals of mice in the forebrain inhibitory neuron overexpression PRRT2 group were significantly stronger than those of mice in the control group after injection of subthreshold doses of PTZ (Figure 22F and Figure 22G).
另外,本发明人在星型胶质细胞中选择性过表达PRRT2也观察到小鼠癫痫易感的现象。因此,在非前脑兴奋性神经元中过表达PRRT2导致不良反应的产生。In addition, the inventors also observed that mice were susceptible to epilepsy by selectively overexpressing PRRT2 in astrocytes. Therefore, overexpression of PRRT2 in non-forebrain excitatory neurons leads to adverse reactions.
综上所述,在前脑兴奋性神经元中过表达PRRT2蛋白可以在不影响正常运动和社交行为的基础上有效增强小鼠的抗癫痫能力,并且该选择性表达策略避免了在前脑抑制型神经元中过表达PRRT2蛋白导致的癫痫易感。In summary, overexpression of PRRT2 protein in forebrain excitatory neurons can effectively enhance the anti-epileptic ability of mice without affecting normal movement and social behavior, and this selective expression strategy avoids the epilepsy susceptibility caused by overexpression of PRRT2 protein in forebrain inhibitory neurons.
以上所述实施例仅表达了本发明的几种实施方式,其描述较为具体和详细,但并不能因此而理解为对本发明专利范围的限制。应当指出的是,对于本领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干变形和改进,这些都属于本发明的保护范围。因此,本发明专利的保护范围应以所附权利要求为准。同时,在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。 The above-mentioned embodiments only express several implementation methods of the present invention, and the description thereof is relatively specific and detailed, but it cannot be understood as limiting the scope of the patent of the present invention. It should be pointed out that, for a person of ordinary skill in the art, several variations and improvements can be made without departing from the concept of the present invention, and these all belong to the protection scope of the present invention. Therefore, the protection scope of the patent of the present invention shall be subject to the attached claims. At the same time, all the documents mentioned in the present invention are cited as references in this application, just as each document is cited as reference separately.
Claims (15)
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN202311118635.7 | 2023-08-31 | ||
| CN202311118635.7A CN117122688B (en) | 2023-08-31 | 2023-08-31 | PRRT2 acting on forebrain excitatory neurons and application of its upregulators |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025044677A1 true WO2025044677A1 (en) | 2025-03-06 |
Family
ID=88856134
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2024/109426 Pending WO2025044677A1 (en) | 2023-08-31 | 2024-08-02 | Use of prrt2 acting on forebrain excitatory neuron and up-regulator thereof |
Country Status (2)
| Country | Link |
|---|---|
| CN (1) | CN117122688B (en) |
| WO (1) | WO2025044677A1 (en) |
Families Citing this family (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN117122688B (en) * | 2023-08-31 | 2024-06-04 | 中国科学院脑科学与智能技术卓越创新中心 | PRRT2 acting on forebrain excitatory neurons and application of its upregulators |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20140304846A1 (en) * | 2011-10-28 | 2014-10-09 | Itek Ventures Pty Ltd | Gene and mutations thereof associated with seizure and movement disorders |
| CN107955818A (en) * | 2016-10-17 | 2018-04-24 | 中国科学院上海生命科学研究院 | A kind of method for building up of non-human primate neurological disease animal model and application thereof |
| CN114921494A (en) * | 2022-04-12 | 2022-08-19 | 西北工业大学 | RNAi expression vector for Cre enzyme-dependent rAAV expression and application thereof |
| CN117122688A (en) * | 2023-08-31 | 2023-11-28 | 中国科学院脑科学与智能技术卓越创新中心 | PRRT2 acting on forebrain excitatory neurons and use of upregulation thereof |
Family Cites Families (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2010037143A1 (en) * | 2008-09-29 | 2010-04-01 | The University Of Montana | Vectors and methods of treating brain seizures |
| WO2017106382A1 (en) * | 2015-12-14 | 2017-06-22 | Cold Spring Harbor Laboratory | Compositions and methods for treatment of central nervous system diseases |
| WO2022221420A2 (en) * | 2021-04-13 | 2022-10-20 | Capsida, Inc. | Selected aav compositions having preferred brain enrichment |
-
2023
- 2023-08-31 CN CN202311118635.7A patent/CN117122688B/en active Active
-
2024
- 2024-08-02 WO PCT/CN2024/109426 patent/WO2025044677A1/en active Pending
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20140304846A1 (en) * | 2011-10-28 | 2014-10-09 | Itek Ventures Pty Ltd | Gene and mutations thereof associated with seizure and movement disorders |
| CN107955818A (en) * | 2016-10-17 | 2018-04-24 | 中国科学院上海生命科学研究院 | A kind of method for building up of non-human primate neurological disease animal model and application thereof |
| CN114921494A (en) * | 2022-04-12 | 2022-08-19 | 西北工业大学 | RNAi expression vector for Cre enzyme-dependent rAAV expression and application thereof |
| CN117122688A (en) * | 2023-08-31 | 2023-11-28 | 中国科学院脑科学与智能技术卓越创新中心 | PRRT2 acting on forebrain excitatory neurons and use of upregulation thereof |
Non-Patent Citations (2)
| Title |
|---|
| MICHETTI, C. ET AL.: "T he PRRT2 Knockout Mouse Recapitulates the Neurological Diseases Associated with PRRT2 Mutations", NEUROBIOLOGY OF DISEASE, vol. 99, 20 December 2016 (2016-12-20), pages 66 - 83, XP029914777, DOI: 10.1016/j.nbd.2016.12.018 * |
| MICHETTI, C. ET AL.: "The PRRT2 Knockout Mouse Recapitulates the Neurological Diseases Associated with PRRT2 Mutations", NEUROBIOLOGY OF DISEASE, vol. 99, 20 December 2016 (2016-12-20), pages 66 - 83, XP029914777, DOI: 10.1016/j.nbd.2016.12.018 * |
Also Published As
| Publication number | Publication date |
|---|---|
| CN117122688A (en) | 2023-11-28 |
| CN117122688B (en) | 2024-06-04 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP6590828B2 (en) | Combination of modified receptor vector and its exogenous agonist in the treatment of seizures | |
| Koilkonda et al. | Leber′ s Hereditary Optic Neuropathy‐Gene Therapy: From Benchtop to Bedside | |
| JP2022522996A (en) | AAV-mediated gene therapy to restore the otoferin gene | |
| RS62359B1 (en) | Treatment of amd using aav sflt-1 | |
| JP2021519609A (en) | Compositions and Methods for Delivering Nucleic Acids to Cochlear and Vestibular Cells | |
| US20160256571A1 (en) | Invention | |
| JP6949867B2 (en) | Methods and Compositions for Treating Nerve Hyperexcitability | |
| JP2008505602A (en) | Methods and compositions for treating neurological diseases | |
| JP2018515622A (en) | Synergistic combination of neuronal survival factors and uses thereof | |
| KR20180072713A (en) | Targeted expression of chloride channels and their use | |
| CN117122688B (en) | PRRT2 acting on forebrain excitatory neurons and application of its upregulators | |
| CN114381465A (en) | Optimized CPY4V2 gene and application thereof | |
| RS64553B1 (en) | Vectors for the treatment of friedreich's ataxia | |
| JP2025529884A (en) | Compositions for use in treating Fabry disease | |
| US10898550B2 (en) | Compositions and methods of treating root avulsion injury | |
| JP7077343B2 (en) | Expression vector containing the engineered gene | |
| WO2021050991A1 (en) | Compositions and methods for treatment of friedreich's ataxia | |
| US20090281163A1 (en) | Regulatory elements that mediate retinal cell-specific gene expression | |
| KR20080007968A (en) | Neuropathic pain relief composition comprising a recombinant vector expressing YAD65 | |
| JP2019031511A (en) | Composition having hypoxia-inducible factor-1 alpha and method of using the composition | |
| JP2025508336A (en) | Recombinant viruses expressing TPK and their use in treating Alzheimer's disease | |
| EP3883591B1 (en) | Relaxin receptor 1 for use in treatment and prevention of heart failure | |
| CN120899949A (en) | Regeneration of functional neurons for the treatment of spinal cord injury and ALS | |
| RU2804196C1 (en) | Treatment method for amyotrophic lateral sclerosis | |
| US20230338581A1 (en) | G-protein-gated-k+ channel-mediated enhancements in light sensitivity in rod-cone dystrophy (rcd) |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24858220 Country of ref document: EP Kind code of ref document: A1 |