Compounds for the treatment of parkinson's disease

文档序号:23419 发布日期:2021-09-21 浏览:22次 中文

阅读说明:本技术 用于治疗帕金森病的化合物 (Compounds for the treatment of parkinson's disease ) 是由 斯蒂芬·弗朗茨·托马斯·韦斯 埃洛伊斯·范德莫维 凯特琳·卡勒 莫妮可·比格努斯 杰西卡·伯 于 2019-11-28 设计创作,主要内容包括:本发明的领域涉及用于治疗和/或预防帕金森病(PD)的LRP/LR。本发明扩展至用于治疗帕金森病(PD)的包含LRP/LR的药物组合物并扩展至维持人或动物体内多巴胺浓度水平的方法。(The field of the invention relates to LRP/LR for the treatment and/or prevention of Parkinson's Disease (PD). The invention extends to pharmaceutical compositions comprising LRP/LR for use in the treatment of Parkinson's Disease (PD) and to methods of maintaining dopamine concentration levels in a human or animal body.)

1. A 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof for use in the treatment and/or prevention of parkinson's disease, wherein the LRP/LR and/or the fragment thereof is for administration to a subject in need thereof.

2. The LRP/LR and/or fragments thereof for use according to claim 1, wherein the LRP/LR comprises the peptide/protein sequence Listing as set forth in SEQ ID NO 1 and/or SEQ ID NO 2, or a fragment thereof as set forth in SEQ ID NO 4 and/or SEQ ID NO 5.

3. The LRP/LR and/or fragments thereof for use according to claim 1 or 2, wherein the LRP/LR comprises a peptide/protein sequence Listing having at least 80% homology with the sequence as set forth in SEQ ID NO 1 or SEQ ID NO 2, or at least 80% homology with the fragment thereof as set forth in SEQ ID NO 4 and/or SEQ ID NO 5.

4. The LRP/LR and/or fragments thereof for use according to any of claims 1-3, wherein the LRP/LR is combined with levodopa and/or dopamine to provide a composition for treating and/or preventing Parkinson's disease.

5. The LRP/LR and/or fragments thereof for use according to claim 4, wherein the composition further comprises a monoamine oxidase B inhibitor.

6. A pharmaceutical composition comprising a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof and a vector for use in treating parkinson's disease, wherein the pharmaceutical composition is for administration to a subject in need thereof.

7. The pharmaceutical composition according to claim 6, wherein the LRP/LR comprises the peptide/protein sequence Listing as set forth in SEQ ID NO 1 or SEQ ID NO 2, or a fragment thereof as set forth in SEQ ID NO 4 and/or SEQ ID NO 5.

8. The pharmaceutical composition according to claim 6, wherein the LRP/LR comprises a peptide/protein sequence Listing having at least 80% homology to the sequence as set forth in SEQ ID NO 1 or SEQ ID NO 2, or a fragment thereof.

9. The pharmaceutical composition according to any one of claims 6 to 8, further comprising levodopa and/or dopamine.

10. The pharmaceutical composition of claim 9, further comprising a monoamine oxidase B inhibitor.

11. The pharmaceutical composition according to any one of claims 6 to 10, wherein the pharmaceutical composition is suitable for parenteral or non-parenteral administration to the subject, wherein non-parenteral administration comprises at least one of the group of: oral, intranasal, rectal, vaginal, ocular and transdermal administration, and wherein parenteral administration comprises at least one of the following groups: intravenous, subcutaneous and intramuscular administration.

12. A method of maintaining dopamine levels in cells of the human or animal body, said method comprising the steps of:

(i) transfecting the cell to produce a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof, thereby increasing the cellular level of LRP/LR and/or fragment thereof; or

(ii) Providing the cell with LRP/LR and/or a fragment thereof to increase the cellular level of LRP/LR and/or a fragment thereof,

such that, when used, the LRP/LR increases the C-terminal domain (CTD) of alpha-synuclein in the human or animal body, thereby preventing aggregation thereof to ameliorate and/or treat and/or prevent PD, and/or such that LRP/LR reduces cellular damage and/or maintains dopamine levels in the human or animal body and/or increases the levels of aureolin in the cells, thereby ameliorating and/or treating and/or preventing PD.

13. The method of claim 12, wherein the LRP/LR comprises the peptide/protein sequence listing as set forth in SEQ ID No. 1 or SEQ ID No. 2, or a fragment thereof as set forth in SEQ ID No. 4 and/or SEQ ID No. 5.

14. The method of claim 12 or claim 13, further comprising the step of providing levodopa and/or dopamine and/or monoamine oxidase B inhibitor.

Technical Field

The field of the invention relates to compounds for the treatment and/or prevention of Parkinson's Disease (PD). The invention extends to pharmaceutical compositions comprising said compounds and a pharmaceutical carrier.

Background

Parkinson's Disease (PD) is the most common motor-activity neurodegenerative disease, with over 1000 million people worldwide suffering from the disease.

There is currently no cure for PD, but only palliative treatment to help control the symptoms. These treatment options include drugs such as levodopa, dopamine, and monoamine oxidase B inhibitors. The first line treatment for PD is levodopa (levodopa/L-Dopa), paired with a peripheral decarboxylase inhibitor to prevent external turnover to dopamine. However, high dose or long-term levodopa use can lead to adverse side effects such as dyskinesias, motor fluctuations, and impulse control disorders. Other non-drug symptomatic therapies include functional stereotactic neurosurgery (deep brain stimulation), physical therapy, speech therapy, and dietary changes.

There is a need for new and inventive compounds, pharmaceutical compositions and/or methods of treating and/or preventing Parkinson's Disease (PD).

Disclosure of Invention

Broadly, according to a first aspect of the present disclosure there is provided a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof for use in the treatment and/or prevention of parkinson's disease, wherein the LRP/LR and/or fragment thereof is for administration to a subject in need thereof.

When used, the LRP/LR increases the C-terminal domain (CTD) of alpha-synuclein in a human or animal, thereby preventing its aggregation to improve and/or treat and/or prevent PD. When used, increasing CTD may also provide a concomitant reduction in cell damage and/or maintenance of dopamine levels, thereby improving and/or treating and/or preventing PD.

LRP/LR may comprise the peptide/protein sequence listing as set forth in SEQ ID NO 1 and/or SEQ ID NO 2, or fragments thereof, which may be set forth in SEQ ID NO 4 and/or SEQ ID NO 5.

LRP/LR may comprise a peptide/protein sequence listing having at least 80% homology to the sequence as set forth in SEQ ID NO:1 or SEQ ID NO:2, or fragments thereof.

LRP/LR may comprise homologues or fragments thereof, as well as homologues of the fragments, wherein LRP/LR may comprise the peptide/protein sequence Listing as set forth in SEQ ID NO:1 or SEQ ID NO: 2.

1 may be the peptide/protein sequence of human LRP/LR and may have the following sequence:

MSGALDVLQMKEEDVLKFLAAGTHLGGTNLDFQMEQYIYKRKSDGIYIINLKRTWEKLLL

AARAIVAIENPADVSVISSRNTGQRAVLKFAAATGATPIAGRFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

TQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTDWS

SEQ ID NO:2 may be the peptide/protein sequence of mouse (Mus musculus) LRP/LR and may have the following sequence:

MSGALDVLQMKEEDVLKFLAAGTHLGGTNLDFQMEQYIYKRKSDGIYIINLKRTWEKLLL

AARAIVAIENPADVSVISSRNTGQRAVLKFAAATGATPIAGRFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

AQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTEWS

it is understood that LRP/LR is highly conserved, and homologues or fragments of SEQ ID NO:1 and SEQ ID NO:2, and/or homologues of the fragments, may also be utilized in order to practice the invention described, illustrated and/or exemplified herein.

The peptide/protein sequence of LRP/LR, or homologues or fragments or homologues of fragments thereof, may bind to, or be bonded to, or be linked to, or be conjugated to, or be associated with additional protein sequences, amino acid sequences, peptides, proteins or antibodies. Alternatively and/or additionally, the protein sequence of LRP/LR may form part of a larger and/or longer protein sequence. In particular embodiments of the invention, LRP/LR may be bound to, or bonded to, or linked to, or conjugated or associated with a FLAG protein such that, when used, the LRP/LR may be tagged with a FLAG. The FLAG protein may comprise a peptide sequence comprising at least one sequence motif DYKDDDDK (SEQ ID NO: 3).

Exemplary embodiments of LRP/LR fragments are exemplified as sequence proteins/peptides having the sequence listed in SEQ ID No. 4 corresponding to the fragment from amino acid residue 102 to amino acid residue 295 as SEQ ID No. 1, and/or SEQ ID No. 5 corresponding to the fragment from amino acid residue 102 to amino acid residue 295 as SEQ ID No. 2.

4 can be the peptide/protein sequence of a human LRP/LR fragment and can have the following sequence:

RFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

TQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTDWS

SEQ ID No. 5 may be a peptide/protein sequence of a mouse LRP/LR fragment and may have the following sequence:

RFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

AQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTEWS

peptides/proteins having the sequence listing as set forth in SEQ ID NO 4 and/or SEQ ID NO 5 are provided for use in the treatment of Parkinson's disease.

The 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragments thereof may be combined with levodopa (levo-dopa/L-dopa) (or derivatives thereof) to provide a composition for use in the treatment and/or prevention of parkinson's disease, such that LRP/LRP and/or fragments thereof, when used, help to maintain dopamine levels in a subject. LRP/LR (or composition) may alternatively or additionally be used in combination with dopamine and/or monoamine oxidase B inhibitors for the treatment and/or prevention of parkinson's disease. Maintaining dopamine levels in the subject avoids frequent use of levodopa and/or, in turn, inhibits adverse side effects associated with long-term use of levodopa by the subject. Further, maintaining dopamine level inhibition in a subject requires the continued use of high levels of levodopa. Thus, by maintaining levodopa levels through LRP/LR, relatively less levodopa can be administered to a subject in need thereof, while producing a more sustained effect. LRP/LR may also increase parkin levels, which may be beneficial in the treatment and/or prevention of PD. Applicants have found that this has significant advantages, providing an improved parkinson's disease treatment and/or prevention regimen.

The subject may be a human, animal, reptile, bird, amphibian, or plant. Typically, the subject may be a human and/or an animal, preferably a human.

The LRP/LR and/or fragments thereof may be formulated into a pharmaceutical composition that may further include a pharmaceutical carrier for parenteral or non-parenteral administration to a subject. Non-parenteral administration may include, but is not limited to, at least one of the following groups: oral, intranasal, rectal, vaginal, ocular and transdermal administration. Typically, parenteral administration may be oral. Parenteral administration may include at least one of intravenous, subcutaneous, and intramuscular administration. Typically, parenteral administration can be intravenous.

It is very surprising for the applicant that providing LRP/LR to the human or animal body, i.e. up-regulating its expression, increases the C-terminal domain (CTD) of alpha-synuclein. Providing LRP/LR as part of a PD treatment regimen will significantly limit the chance of any unwanted side effects as it is a naturally occurring protein/peptide. This certainly improves at least one of the problems known in the prior art.

According to a second aspect of the invention, there is provided a pharmaceutical composition comprising a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof and a carrier for use in the treatment of parkinson's disease, wherein the pharmaceutical composition is for administration to a subject in need thereof.

When used, the LRP/LR of the pharmaceutical composition increases the C-terminal domain (CTD) of alpha-synuclein in a human or animal, thereby preventing its aggregation to improve and/or treat and/or prevent PD. When used, increasing CTD may also provide a concomitant reduction in cell damage and/or maintenance of dopamine levels, thereby improving and/or treating and/or preventing PD. LRP/LR may also increase parkin levels, thereby facilitating the treatment and/or prevention of PD.

LRP/LR may comprise the peptide/protein sequence listing as set forth in SEQ ID NO 1 or SEQ ID NO 2, or fragments thereof, which fragments may be as set forth in SEQ ID NO 4 and/or SEQ ID NO 5.

LRP/LR may comprise a peptide/protein sequence listing having at least 80% homology to the sequence as set forth in SEQ ID NO:1 or SEQ ID NO:2, or fragments thereof.

LRP/LR may comprise homologues or fragments thereof, as well as homologues of the fragments, wherein LRP/LR may comprise the peptide/protein sequence Listing as set forth in SEQ ID NO:1 or SEQ ID NO: 2.

The peptide/protein sequence of LRP/LR, or homologues or fragments or homologues of fragments thereof, may bind to, or be bonded to, or be linked to, or be conjugated to, or be associated with additional protein sequences, amino acid sequences, peptides, proteins or antibodies. Alternatively and/or additionally, the peptide/protein sequence of LRP/LR may form part of a larger and/or longer peptide/protein sequence. In particular embodiments of the invention, LRP/LR may be bound to, or bonded to, or linked to, or conjugated or associated with a FLAG protein such that, when used, the LRP/LR may be tagged with a FLAG. The FLAG protein may comprise a peptide sequence comprising at least one sequence motif DYKDDDDK (SEQ ID NO: 3).

Exemplary embodiments of fragments of the peptide/protein sequence listing are illustrated as SEQ ID NO. 4 corresponding to a fragment from 102 to 295 of SEQ ID NO. 1 and/or SEQ ID NO. 5 corresponding to a fragment from 102 to 295 of SEQ ID NO. 2.

4 can be the peptide/protein sequence of a human LRP/LR fragment and can have the following sequence:

RFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

TQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTDWS

SEQ ID No. 5 may be a peptide/protein sequence of a mouse LRP/LR fragment and may have the following sequence:

RFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

AQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTEWS

the pharmaceutical composition may further comprise levodopa (levo-dopa/L-dopa) (or derivatives thereof). When used, LRP/LRP and/or fragments thereof are beneficial for maintaining dopamine levels in a subject. Maintaining dopamine levels in a subject avoids frequent use of levodopa and/or, in turn, inhibits adverse side effects associated with long-term use of levodopa by a subject. Further, maintaining dopamine levels in the subject inhibits the need for sustained use of high levels of levodopa by the subject. The pharmaceutical composition when in use allows for the administration of relatively less levodopa to a subject and has a more sustained effect. The pharmaceutical composition may further comprise dopamine and/or monoamine oxidase B inhibitors and/or excipients. When used, LRP/LRP and/or fragments thereof facilitate an increase in parkinsonian protein levels, which facilitates the treatment and/or prevention of parkinson's disease by blocking the aggregation of alpha-synuclein. An increase in parkinsonian levels will also increase the cell viability of parkinson's disease.

The subject may be a human, animal, reptile, bird, amphibian, or plant. Typically, the subject may be a human and/or an animal, preferably a human.

The pharmaceutical composition may be suitable for parenteral or non-parenteral administration to a subject. Non-parenteral administration may include, but is not limited to, at least one of the following groups: oral, intranasal, rectal, vaginal, ocular and transdermal administration. Typically, parenteral administration may be oral. Parenteral administration may include at least one of intravenous, subcutaneous, and intramuscular administration. Typically, parenteral administration can be intravenous.

According to a third aspect of the invention there is provided a method of increasing the C-terminal domain (CTD) of alpha-synuclein in a cell of a human or animal body and/or a method of reducing cell damage and/or a method of maintaining dopamine levels and/or a method of increasing the level of parkin in a human or animal body, the method comprising the steps of:

(i) transfecting a cell to produce a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof, thereby increasing the cellular level of LRP/LR and/or fragment thereof; or

(ii) Providing LRP/LR and/or a fragment thereof to a cell to increase the cellular level of LRP/LR and/or a fragment thereof, such that when used, the LRP/LR increases the C-terminal domain (CTD) of alpha-synuclein in a human or animal body, thereby preventing aggregation thereof to ameliorate and/or treat and/or prevent PD, and/or such that LRP/LR reduces cellular damage and/or maintains dopamine levels in the human or animal body, and/or such that LRP/LR causes an increase in parkinsonine levels/concentrations, thereby ameliorating and/or treating and/or preventing PD.

LRP/LR may comprise the peptide/protein sequence listing as set forth in SEQ ID NO 1 or SEQ ID NO 2, or fragments thereof.

LRP/LR may comprise a peptide/protein sequence listing having at least 80% homology to the sequence as set forth in SEQ ID NO:1 or SEQ ID NO:2, or fragments thereof.

LRP/LR may comprise homologues or fragments thereof, as well as homologues of the fragments, wherein LRP/LR may comprise the peptide/protein sequence Listing as set forth in SEQ ID NO:1 or SEQ ID NO: 2.

The peptide/protein sequence of LRP/LR, or homologues or fragments or homologues of fragments thereof, may bind to, or be bonded to, or be linked to, or be conjugated to, or be associated with additional protein sequences, amino acid sequences, peptides, proteins or antibodies. Alternatively and/or additionally, the peptide/protein sequence of LRP/LR may form part of a larger and/or longer peptide/protein sequence. In particular embodiments of the invention, LRP/LR may be bound to, or bonded to, or linked to, or conjugated or associated with a FLAG protein such that, when used, the LRP/LR may be tagged with a FLAG. The FLAG protein may comprise a peptide/protein sequence that includes at least one sequence motif DYKDDDDK (SEQ ID NO: 3).

It is understood that the step of transfecting the cells to produce a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment may be performed via procedures known in the art, including introducing a transfection agent into the cells. The step of transfecting the cells may upregulate LRP/LR.

Exemplary embodiments of fragments of the peptide/protein sequence listing are illustrated as SEQ ID NO. 4 corresponding to a fragment from 102 to 295 of SEQ ID NO. 1 and/or SEQ ID NO. 5 corresponding to a fragment from 102 to 295 of SEQ ID NO. 2.

4 can be the peptide/protein sequence of a human LRP/LR fragment and can have the following sequence:

RFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

TQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTDWS

SEQ ID No. 5 may be a peptide/protein sequence of a mouse LRP/LR fragment and may have the following sequence:

RFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

AQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTEWS

the subject may be a human, animal, reptile, bird, amphibian, or plant. Typically, the subject may be a human and/or an animal, preferably a human.

The LRP/LR and/or fragments thereof may be formulated into a pharmaceutical composition that may further include a pharmaceutical carrier for parenteral or non-parenteral administration to a subject. Non-parenteral administration may include, but is not limited to, at least one of the following groups: oral, intranasal, rectal, vaginal, ocular and transdermal administration. Typically, parenteral administration may be oral. Parenteral administration may include at least one of intravenous, subcutaneous, and intramuscular administration. Typically, parenteral administration can be intravenous. Alternatively and/or additionally, the transfection agent may be formulated as a pharmaceutical composition, wherein the pharmaceutical composition may further comprise a pharmaceutical carrier for parenteral or non-parenteral administration to a subject.

The method of maintaining dopamine levels in a human or animal body can further comprise the step of providing levodopa (l-dopa/levo dopa) and LRP/LR or a fragment thereof to the subject. When used, LRP/LRP and/or fragments thereof are beneficial for maintaining dopamine levels in a subject. Maintaining dopamine levels in the subject avoids frequent use of levodopa and/or, in turn, inhibits adverse side effects associated with long-term use of levodopa by the subject. Further, maintaining dopamine level inhibition in a subject requires the continued use of high levels of levodopa. This method allows for the administration of relatively less levodopa to a subject and has a more sustained effect. The combination of LRP/LR and/or fragments thereof and levodopa can be formulated as a pharmaceutical composition for administration to a subject in need thereof. The method may further comprise the step of providing dopamine and/or monoamine oxidase B inhibitor to the subject.

According to a fourth aspect of the present invention there is provided a method of treatment and/or prevention of parkinson's disease, the method comprising the step of administering to a subject in need thereof a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof, such that, in use, the LRP/LR and/or fragment thereof increases the C-terminal domain (CTD) of alpha-synuclein in a human or animal, thereby preventing aggregation thereof to improve and/or treat and/or prevent PD, and/or such that the LRP/LR reduces cellular damage and/or maintains dopamine levels in the human or animal body, and/or such that the LRP/LR increases parkin levels, thereby improving and/or treating and/or preventing PD.

LRP/LR may comprise the peptide/protein sequence listing as set forth in SEQ ID NO 1 or SEQ ID NO 2, or fragments thereof.

LRP/LR may comprise a peptide/protein sequence listing having at least 80% homology to the sequence as set forth in SEQ ID NO:1 or SEQ ID NO:2, or fragments thereof.

LRP/LR may comprise homologues or fragments thereof, as well as homologues of the fragments, wherein LRP/LR may comprise the peptide/protein sequence Listing as set forth in SEQ ID NO:1 or SEQ ID NO: 2.

The peptide/protein sequence of LRP/LR, or homologues or fragments or homologues of fragments thereof, may bind to, or be bonded to, or be linked to, or be conjugated to, or be associated with additional protein sequences, amino acid sequences, peptides, proteins or antibodies. Alternatively and/or additionally, the peptide/protein sequence of LRP/LR may form part of a larger and/or longer peptide/protein sequence. In particular embodiments of the invention, LRP/LR may be bound to, or bonded to, or linked to, or conjugated or associated with a FLAG protein such that, when used, the LRP/LR may be tagged with a FLAG. The FLAG protein may comprise a peptide/protein sequence that includes at least one sequence motif DYKDDDDK (SEQ ID NO: 3).

Exemplary embodiments of fragments of the peptide/protein sequence listing are illustrated as SEQ ID NO. 4 corresponding to a fragment from 102 to 295 of SEQ ID NO. 1 and/or SEQ ID NO. 5 corresponding to a fragment from 102 to 295 of SEQ ID NO. 2.

SEQ ID No. 4 may be the peptide/protein sequence of fragment human LRP/LR and may have the following sequence:

RFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

TQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTDWS

SEQ ID No. 5 may be a peptide/protein sequence of a mouse LRP/LR fragment and may have the following sequence:

RFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

AQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTEWS

the method of treating and/or preventing Parkinson's disease may further comprise the step of administering levodopa (l-dopa/levodopa) to the subject. This step may occur simultaneously with, or in addition to, the step of administering to the subject in need thereof a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof. When used, LRP/LRP and/or fragments thereof typically facilitate maintenance of dopamine levels in a subject when administered concurrently with or in addition to administration of levodopa. Maintaining dopamine levels in the subject avoids frequent use of levodopa and/or, in turn, inhibits adverse side effects associated with long-term use of levodopa by the subject.

The subject may be a human, animal, reptile, bird, amphibian, or plant. Typically, the subject may be a human and/or an animal, preferably a human.

The LRP/LR and/or fragments thereof may be formulated into a pharmaceutical composition that may further include a pharmaceutical carrier for parenteral or non-parenteral administration to a subject. Non-parenteral administration may include, but is not limited to, at least one of the following groups: oral, intranasal, rectal, vaginal, ocular and transdermal administration. Typically, parenteral administration may be oral. Parenteral administration may include at least one of intravenous, subcutaneous, and intramuscular administration. Typically, parenteral administration can be intravenous. The combination of LRP/LR and/or fragments thereof and levodopa can be formulated as a pharmaceutical composition for administration to a subject in need thereof. The pharmaceutical composition may further comprise dopamine and/or monoamine oxidase B inhibitors and/or excipients.

Broadly, according to a fifth aspect of the present disclosure there is provided a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof for use in the treatment of trauma, wherein the LRP/LR and/or fragment thereof is for administration to a subject in need thereof. The wound may be an external skin lesion. The wound may be an internal lesion and/or an injury of an internal organ in the human or animal body.

LRP/LR may comprise the peptide/protein sequence listing as set forth in SEQ ID NO 1 and/or SEQ ID NO 2, or fragments thereof as set forth in SEQ ID NO and/or SEQ ID NO 5.

LRP/LR may comprise a peptide/protein sequence listing having at least 80% homology to the sequence as set forth in SEQ ID NO:1 or SEQ ID NO:2, or fragments thereof.

LRP/LR may comprise homologues or fragments thereof, as well as homologues of the fragments, wherein LRP/LR may comprise the peptide/protein sequence Listing as set forth in SEQ ID NO:1 or SEQ ID NO: 2.

1 may be the peptide/protein sequence of human LRP/LR and may have the following sequence:

MSGALDVLQMKEEDVLKFLAAGTHLGGTNLDFQMEQYIYKRKSDGIYIINLKRTWEKLLL

AARAIVAIENPADVSVISSRNTGQRAVLKFAAATGATPIAGRFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

TQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTDWS

2 can be the peptide/protein sequence of mouse (mus musculus) LRP/LR and can have the following sequence:

MSGALDVLQMKEEDVLKFLAAGTHLGGTNLDFQMEQYIYKRKSDGIYIINLKRTWEKLLL

AARAIVAIENPADVSVISSRNTGQRAVLKFAAATGATPIAGRFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

AQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTEWS

it is understood that LRP/LR is highly conserved, and homologues or fragments of SEQ ID NO:1 and SEQ ID NO:2, and/or homologues of the fragments, may also be utilized in order to practice the invention described, illustrated and/or exemplified herein.

The peptide/protein sequence of LRP/LR, or homologues or fragments or homologues of fragments thereof, may bind to, or be bonded to, or be linked to, or be conjugated to, or be associated with additional protein sequences, amino acid sequences, peptides, proteins or antibodies. Alternatively and/or additionally, the protein sequence of LRP/LR may form part of a larger and/or longer protein sequence. In particular embodiments of the invention, LRP/LR may be bound to, or bonded to, or linked to, or conjugated or associated with a FLAG protein such that, when used, the LRP/LR may be tagged with a FLAG. The FLAG protein may comprise a peptide sequence comprising at least one sequence motif DYKDDDDK (SEQ ID NO: 3).

Exemplary embodiments of LRP/LR fragments are exemplified as proteins/peptides having the sequences listed in SEQ ID No. 4 corresponding to the fragment from amino acid residue 102 to amino acid residue 295 as SEQ ID No. 1 and/or SEQ ID No. 5 corresponding to the fragment from amino acid residue 102 to amino acid residue 295 as SEQ ID No. 2.

4 can be the peptide/protein sequence of a human LRP/LR fragment and can have the following sequence:

RFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

TQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTDWS

SEQ ID No. 5 may be a peptide/protein sequence of a mouse LRP/LR fragment and may have the following sequence:

RFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

AQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTEWS

peptides/proteins having the sequence listing as set out in SEQ ID NO 4 and/or SEQ ID NO 5 are provided for use in the treatment of wounds, preferably external skin lesions of the human or animal body.

The subject may be a human and/or an animal, preferably a human.

The LRP/LR and/or fragments thereof may be formulated into a pharmaceutical composition that may further include a pharmaceutical carrier for parenteral or non-parenteral administration to a subject. Non-parenteral administration may include, but is not limited to, at least one of the following groups: oral, intranasal, rectal, vaginal, ocular and transdermal administration. Typically, parenteral administration may be oral. Parenteral administration may include at least one of intravenous, subcutaneous, and intramuscular administration. Typically, parenteral administration can be intravenous.

Typically, the compositions are formulated for topical administration to an external skin lesion of the human and/or animal body. Such topical administration may be, but is not limited to, ointments, creams, gels, lotions, liquids, bandages, plasters, creams and/or combinations of the foregoing.

Further provided is a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof, substantially as described herein, for use in the treatment of parkinson's disease, described and/or exemplified with reference to any one of the examples and/or drawings.

There is further provided a pharmaceutical composition substantially as described, illustrated and/or exemplified herein with reference to any one of the examples and/or figures.

Further provided are methods of increasing the C-terminal domain (CTD) of alpha-synuclein in a human or animal body and/or methods of reducing cellular damage and/or methods of maintaining dopamine levels in a human or animal body, the methods being substantially as described, illustrated and/or exemplified herein with reference to any one of the example(s) and/or figures.

Drawings

Embodiments of the present disclosure will now be described, by way of example only, with reference to the accompanying drawings, in which:

FIG. 1 shows the full-length structure of alpha (α) -synuclein;

FIGS. 2A and B show Western blot and densitometric analysis of protein lysates of HEK293 cells, performed on pCIneo-LRP:FLAGtransfected HEK293 and untransfected HEK293 cells. The primary antibody used was raised against LRP and FLAG. Secondary antibody was conjugated with HRP. HRP-conjugated primary antibody was used to detect β -actin. pCIneo-LRP:FLAGtransfection caused LRP:FLAGoverexpression and increase in total LRP levels. Densitometric analysis was performed followed by statistical analysis using student's t-test. Optical density and statistical analysis revealed pCIneo-LRP-FLAG transfection caused an 71.24% increase in total LRP levels;

FIGS. 3A to N show confocal microscopy results at 630 magnification, indicating expression levels and localization of LRP and alpha-synuclein;

fig. 4A and B show western blot and optical density analysis of protein lysates. Western blot analysis was performed on pCIneo-LRP:FLAGtransfected HEK293 and untransfected HEK293 cells. The primary antibody used was raised against alpha-synuclein. Secondary antibody was conjugated with HRP. HRP-conjugated primary antibody was used to detect β -actin. pCIneo-LRP:: FLAG transfection caused an increase in the level of the C-terminal domain of alpha-synuclein, LRP:: FLAG overexpression and an increase in total LRP levels. Densitometric analysis was performed followed by statistical analysis using student's t-test. Optical density and statistical analysis revealed pCIneo-LRP that FLAG transfection caused a significant 165.22% increase in the level of the C-terminal domain of alpha-synuclein;

FIG. 5 shows MTT assay analysis of LRP:FLAGtransfected and untransfected HEK293 cells, indicating an increase in cell viability of LRP:FLAGoverexpressing HEK293 cells compared to untransfected HEK293 cells in the presence of 200nM and 500nM recombinant alpha-synuclein;

FIG. 6 shows MTT assay analysis in LRP:FLAGtransfected and untransfected SH-SY5Y cells, indicating increased cell viability of LRP:FLAGoverexpressing SH-SY5Y cells compared to untransfected SH-SY5Y cells in the presence of 200nM and 500nM recombinant alpha-synuclein;

FIGS. 7A and B show MTT assay analysis of pCIneo-molLRP:FLAGtransfected and untransfected HEK293 cells, indicating a significant increase in cell viability of LRP:FLAGoverexpressing HEK293 cells at 500uM and 750nM 1-methyl-4-phenylpyridine iodide (MPP +) for (A)48 hours and (B)72 hours; and

figure 8 shows confocal microscopy results at 630 magnification, indicating expression levels and localization of LRP and parkin. The primary antibody used was raised against LRP, parkin and hTERT. Secondary antibody was conjugated to FITC, Cy3 and alexaflur 647. Panels A to F represent DLD-1 cells, and Panels G to L represent DLD-1 cells that have been transfected with pCIneo-LRP FLAG plasmid. Pangin is shown in yellow for panels A and G, while LRP/LR is shown in green for panels B and H. Panels D and I show the combination of parkin and LRP/LR. Panels E and K represent co-localization of the two proteins, with white indicating overlap and possible interaction between parkin and LRP/LR. Co-localization occurred intracellularly, as shown by the diagonal in the 2D cell fluorescence map (F), where 59% of the parkin was co-localized with 54% of the LRP/LR in DLD-1 cells, and 99% of the parkin was co-localized with 22% of the LRP in DLD-1 cells overexpressing LRP:FLAG.

Detailed Description

The summary above is hereby incorporated by reference herein in its entirety and not repeated in its entirety to avoid lengthy repetition.

Parkinson's Disease (PD) is the most common motor-activity neurodegenerative disease, with over 1000 million people worldwide suffering from the disease. Alpha-synuclein is implicated in PD and is encoded by the gene SNCA. In PD, alpha-synuclein forms aggregates and is the major component of the lewy body. Mutations in the SNCA gene cause aggregation of alpha-synuclein and thus cause familial forms of parkinson's disease. The C-terminal domain (CTD) of alpha-synuclein prevents aggregation of alpha-synuclein (see fig. 1).

LRP/LR is a multifunctional receptor protein that plays a crucial role in the pathogenesis of neurodegenerative disorders, including alzheimer's disease and prion disorders. It has previously been shown, according to the disclosure of PCT/IB2012/054968, that knockout (down-regulation) of LRP/LR is a useful treatment regimen for alzheimer's disease.

In contrast and surprisingly to the applicant, it has now been found that overexpression of LRP/LR is a beneficial therapeutic regimen for the treatment of Parkinson's Disease (PD). This was completely unexpected in view of the previous findings. Without being limited by theory, this is indicative of the complexity and unpredictable nature of the biochemical pathways involved in neurodegenerative disorders, particularly PD.

The diagnosis of PD relies on the observation of the above-mentioned disease properties, since there are no specific tests available for diagnosing PD. Non-motor symptoms are used to diagnose the disease at an early stage, as they are more prominent. There is currently no cure for PD, but only palliative treatment to help control the symptoms. These treatment options include drugs such as levodopa, dopamine, and monoamine oxidase B inhibitors. The first line treatment for PD is levodopa (levodopa/L-Dopa), paired with a peripheral decarboxylase inhibitor to prevent external turnover to dopamine. However, high dose or long-term levodopa use can lead to adverse side effects such as dyskinesias, motor fluctuations, and impulse control disorders. Other non-drug symptomatic therapies include functional stereotactic neurosurgery (deep brain stimulation), physical therapy, speech therapy, and dietary changes. There is a need for new compounds for the treatment of PD.

PD is mainly defined by the absence of dopamine-producing neurons located in the Substantia Nigra Pars Compacta (SNPC) and ultimately incurs prominent motor characteristics of PD (Dexter & Jenner, 2013). The absence of these dopamine-producing neurons located in SNPCs causes not only organ atrophy, but also the presence of lewy bodies (Dauer & Prezedborski, 2003). Lewy bodies are intracellular protein inclusion bodies consisting of more than 76 different components (Wakabayashi et al, 2007). Motor and sleep regulation are two major functions of SNPC, and thus the absence of these neurons leads to dysregulation of these and other functions of SNPC (Lima et al, 2009). In PD, oxidative toxins released by glial cells during oxidative stress can also lead to neuronal cell death (Jenner, 2003).

Sporadic forms of PD account for 90% of the total number of PD cases (Yasuda & Mochizuki, 2010) and may be caused by unknown degenerative disease processes, toxins, metabolic disorders, or drugs (Dickson, 2012). Only a few PD cases were familial and resulted from mutations in the genes encoding the protein, alpha-synuclein and parkin (Yasuda & Mochizuki, 2010). Alpha-synuclein is the major component of lewy bodies and is encoded by the gene SNCA (Yasuda & Mochizuki, 2010). Mutations in the SNCA gene cause aggregation of alpha-synuclein and thus the formation of families of PD (Yasuda & Mochizuki, 2010). Parkin is an E3 ubiquitin ligase that is involved in ubiquitin-mediated protein degradation (Seirafi et al, 2015). The PARK2 gene encodes parkin and mutations in this gene cause loss of function of parkin (Kitada et al, 1998). Parkin is responsible for ubiquitination of O-glycosylated forms of α -synuclein, and thus loss of function of parkin causes aggregation of α -synuclein (Shimura et al, 2000). Yang et al (2003) demonstrated that overexpression of parkin reduced the toxicity of alpha-synuclein and thus protected cells from the destructive effects of alpha-synuclein (Yang et al, 2003).

The 19kDa protein (α -synuclein) had no defined structure in aqueous solution (Stefanis et al, 2012). Alpha-synuclein is found mainly in neuronal cells and nuclei of synaptic vesicles (Auluck et al, 2010). This protein consists of ctd (ctd), the non-amyloid β component of plaques (NAC) domain and ntd (ntd) (Xu & Pu, 2016). NTD is involved in membrane binding (Vamvaca et al, 2009). The NAC domain is the domain responsible for aggregation (Rajagopalan et al, 2001) and CTD interacts with the NAC domain to inhibit aggregation (emazadeh, 2016). The NAC region of alpha-synuclein is also a component of plaques found in Alzheimer's Disease (AD) (Bendor et al, 2013). Alpha-synuclein has multiple functions, including neuronal differentiation via the ERK/MAPK pathway (Fu et al, 2000), leading to down-regulation of dopamine biosynthesis by inhibition of tyrosine hydroxylase (Rodriguez-Araujo et al, 2015).

The major component of lewy bodies is the fibrillar form of α -synuclein due to mutation and replication (Dolgacheva et al, 2018). Patients with PD also have high levels of phosphorylated and nitrated forms of alpha-synuclein, which may indicate that these post-translational modifications may promote aggregation of alpha-synuclein, thereby promoting PD (Rocha et al, 2018). Dysfunctional lysosomal clearance and mutated lysosomal hydrolases favor α -synuclein aggregation, and thus the probability of PD progression is higher in these individuals (Sidransky et al, 2009). Autophagy-lysosomal pathway (ALP) and dysregulation of the ubiquitin-proteasome system (UPS) favour α -synuclein aggregation, as these systems cause α -synuclein degradation (Rocha et al, 2018). Recent studies have also demonstrated that CTD truncation favors the aggregation of α -synuclein (van der Wateren et al, 2018).

In the following, applicants revealed that human embryonic kidney (HEK293) cells were stably transfected with pCIneo-LRP:: FLAG, which resulted in successful LRP:: FLAG overexpression. LRP FLAG overexpression causes: (i) increased levels of alpha-synuclein, (ii) co-localization between alpha-synuclein and LRP/LR, (iii) co-localization between LRP/LR and parkin, and (iv) increased cell viability in the presence of MPP + (Parkinson's disease inducer) of LRP:FLAGoverexpressing HEK293 cells.

Western blot analysis revealed LRP that CTD levels of alpha-synuclein were significantly increased after FLAG overexpression. MTT assay after treatment with recombinant α -synuclein revealed an increase in cell viability of LRP:FLAGtransfected HEK293 and SH-SY5Y cells compared to untransfected cells. Without being limited by theory, applicants provide LRP/LR for use in the treatment and/or prevention of PD, wherein LRP/LR is for administration to a human or animal in need thereof, and wherein the LRP/LR increases the C-terminal domain (CTD) of alpha-synuclein, thereby preventing aggregation thereof to improve PD. When used, increasing CTD may also simultaneously reduce cell damage and/or maintain dopamine levels. Maintaining dopamine level inhibition in the subject is a need to continue using high levels of levodopa as a treatment regimen for parkinson's disease. Thus, by maintaining levodopa levels through LRP/LR, relatively less levodopa can be administered to a subject in need thereof, while having a more sustained effect. LRP/LR was also shown to increase cell viability. Applicants have found that this is a significant advantage, providing an improved parkinson's disease treatment and/or prevention regimen.

MTT assay analysis of FLAG-transfected and untransfected HEK293 cells indicated that in the presence of 500uM and 750nM 1-methyl-4-phenylpyridine iodide (MPP +) (PD inducer), LRP:FLAGoverexpressing HEK293 cells, cell viability was significantly increased as shown in FIGS. 7A and 7B. Increased levels of parkin facilitated by LRP/LR overexpression may further provide for maintaining alpha-synuclein levels and/or inhibiting or blocking aggregation of alpha-synuclein levels.

According to a first aspect of the present invention there is provided a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof for use in the treatment and/or prevention of parkinson's disease, wherein the LRP/LR and/or fragment thereof is for administration to a subject in need thereof. LRP/LR may comprise a peptide/protein sequence Listing as set forth in SEQ ID NO:1 or SEQ ID NO:2, or fragments thereof, such as fragments having a sequence Listing as set forth in SEQ ID NO:4 and/or SEQ ID NO: 5. Alternatively and/or additionally, the LRP/LR may comprise a peptide/protein sequence listing having at least 80% homology to the sequence as set forth in SEQ ID NO:1 or SEQ ID NO:2 or SEQ ID NO:4 or SEQ ID NO:5, or a fragment thereof. Still further, the LRP/LR may comprise homologues or fragments thereof, and homologues of the fragments, wherein the LRP/LR may comprise the peptide/protein sequence Listing as set forth in SEQ ID NO 1 or SEQ ID NO 2 or SEQ ID NO 4 or SEQ ID NO 5.

When used, the LRP/LR increases the C-terminal domain (CTD) of alpha-synuclein in a human or animal, thereby preventing its aggregation to improve and/or treat and/or prevent PD. When used, increasing CTD may also provide a concomitant reduction in cell damage and/or maintenance of dopamine levels, thereby improving and/or treating and/or preventing PD. LRP/LR can be used in combination with levodopa (levodopa/dopa/l-dopa) for the treatment and/or prevention of PD, wherein LRP/LR is beneficial for maintaining dopamine levels, thereby requiring less levodopa and inhibiting the negative side effects of high levodopa doses and/or sustained use of levodopa. Increased levels of parkin that LRP/LR over-expression is beneficial may further provide for maintaining alpha-synuclein levels and/or inhibiting or blocking aggregation of alpha-synuclein levels.

1 may be the peptide/protein sequence of human LRP/LR and may have the following sequence:

MSGALDVLQMKEEDVLKFLAAGTHLGGTNLDFQMEQYIYKRKSDGIYIINLKRTWEKLLL

AARAIVAIENPADVSVISSRNTGQRAVLKFAAATGATPIAGRFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

TQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTDWS

2 can be the peptide/protein sequence of mouse (mus musculus) LRP/LR and can have the following sequence:

MSGALDVLQMKEEDVLKFLAAGTHLGGTNLDFQMEQYIYKRKSDGIYIINLKRTWEKLLL

AARAIVAIENPADVSVISSRNTGQRAVLKFAAATGATPIAGRFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

AQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTEWS

it is understood that LRP/LR is highly conserved, and homologues or fragments of SEQ ID NO:1 and SEQ ID NO:2, and/or homologues of the fragments, may also be utilized in order to practice the invention described, illustrated and/or exemplified herein.

The peptide/protein sequence of LRP/LR, or homologues or fragments or homologues of fragments thereof, may bind to, or be bonded to, or be linked to, or be conjugated to, or be associated with additional protein sequences, amino acid sequences, peptides, proteins or antibodies. Alternatively and/or additionally, the peptide/protein sequence of LRP/LR may form part of a larger and/or longer protein sequence. In particular embodiments of the invention, LRP/LR may be bound to, or bonded to, or linked to, or conjugated or associated with a FLAG protein such that, when used, the LRP/LR may be tagged with a FLAG. The FLAG protein may comprise a peptide/protein sequence that includes at least one sequence motif DYKDDDDK (SEQ ID NO: 3). FLAG is used to help evaluate and/or quantify and/or explain the experiments in the examples section below. Although used in the examples, this is not necessary in order to practice the claimed invention. However, one skilled in the art may wish to include a tag such as FLAG.

Exemplary embodiments of LRP/LR fragments are exemplified as proteins/peptides having the sequences listed in SEQ ID No. 4 corresponding to the fragment from amino acid residue 102 to amino acid residue 295 as SEQ ID No. 1 and/or SEQ ID No. 5 corresponding to the fragment from amino acid residue 102 to amino acid residue 295 as SEQ ID No. 2.

RFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

TQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTDWS

SEQ ID No. 5 may be a peptide/protein sequence of a mouse LRP/LR fragment and may have the following sequence:

RFTPGTFTNQIQAAFREPR

LLVVTDPRADHQPLTEASYVNLPTIALCNTDSPLRYVDIAIPCNNKGAHSVGLMWWMLAR

EVLRMRGTISREHPWEVMPDLYFYRDPEIEKEEQAAAEKAVTKEEFQGEWTAPAPEFTA

AQPEVADWSEGVQVPSVPIQQFPTEDWSAQPATEDWSAAPTAQATEWVGATTEWS

the LRP/LR and/or fragments thereof may be formulated into a pharmaceutical composition that may further include a pharmaceutical carrier for parenteral or non-parenteral administration to a subject. Non-parenteral administration may include, but is not limited to, at least one of the following groups: oral, intranasal, rectal, vaginal, ocular and transdermal administration. Typically, parenteral administration is oral. Parenteral administration may include at least one of intravenous, subcutaneous, and intramuscular administration. Typically, parenteral administration can be intravenous. The pharmaceutical composition may further comprise levodopa. The pharmaceutical composition may further comprise dopamine and/or monoamine oxidase B inhibitors and/or excipients.

According to a second aspect of the present invention, there is provided a pharmaceutical composition comprising a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof and a carrier for use in the treatment and/or prevention of parkinson's disease, wherein the pharmaceutical composition is for administration to a subject in need thereof. The pharmaceutical composition may also include levodopa (levodopa/l-dopa). The pharmaceutical composition may further comprise dopamine and/or monoamine oxidase B inhibitors and/or excipients.

According to a third aspect of the invention there is provided a method of increasing the C-terminal domain (CTD) of alpha-synuclein and/or a method of reducing cellular damage and/or a method of maintaining dopamine levels and/or increasing resident protein levels in a human or animal body, the method comprising the steps of:

(i) transfecting a cell to produce a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof, thereby increasing the cellular level of LRP/LR and/or fragment thereof; or

(ii) Providing LRP/LR and/or a fragment thereof to a cell to increase the cellular level of LRP/LR and/or a fragment thereof, such that when used, the LRP/LR increases the C-terminal domain (CTD) of alpha-synuclein in a human or animal body, thereby preventing aggregation thereof to ameliorate and/or treat and/or prevent PD, and/or such that LRP/LR reduces cellular damage and/or maintains dopamine levels in the human or animal body, and/or such that LRP/LR increases parkin levels/concentrations, thereby ameliorating and/or treating and/or preventing PD.

It is understood that the step of transfecting the cells to produce a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment may be performed via procedures known in the art, including introducing a transfection agent into the cells.

The method of maintaining dopamine levels in a human or animal body can further comprise the step of providing levodopa (l-dopa/levo dopa) and LRP/LR or a fragment thereof to the subject. When used, LRP/LRP and/or fragments thereof are beneficial for maintaining dopamine levels in a subject. Maintaining dopamine levels in the subject avoids frequent use of levodopa and/or, in turn, inhibits adverse side effects associated with long-term use of levodopa by the subject. Further, maintaining dopamine level inhibition in a subject requires the continued use of high levels of levodopa. This method allows for the administration of relatively less levodopa to a subject and has a more sustained effect. The combination of LRP/LR and/or fragments thereof and levodopa can be formulated as a pharmaceutical composition for administration to a subject in need thereof. The method may further comprise the step of providing dopamine and/or monoamine oxidase B inhibitor to the subject.

According to a fourth aspect of the present invention there is provided a method of treatment and/or prevention of parkinson's disease, the method comprising the step of administering to a subject in need thereof 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragments thereof.

When used, the LRP/LR increases the C-terminal domain (CTD) of alpha-synuclein in a human or animal, thereby preventing its aggregation to improve and/or treat and/or prevent PD. When used, increasing CTD may also provide a concomitant reduction in cell damage and/or maintenance of dopamine levels, thereby improving and/or treating and/or preventing PD. The method may further comprise the step of providing levodopa (levodopa/dopa/l-dopa) and/or dopamine and/or monoamine oxidase B inhibitor to the subject.

According to a fifth aspect of the invention, there is provided a 37kDa/67kDa laminin receptor precursor/high affinity laminin receptor (LRP/LR) and/or fragment thereof for use in the treatment of trauma, wherein the LRP/LR and/or fragment thereof is for administration to a subject in need thereof. LRP/LR may comprise a peptide/protein sequence Listing as set forth in SEQ ID NO:1 or SEQ ID NO:2, or a fragment thereof, such as a fragment having a sequence Listing as set forth in SEQ ID NO:4 or SEQ ID NO: 5. Alternatively and/or additionally, the LRP/LR may comprise a peptide/protein sequence listing having at least 80% homology to the sequence as set forth in SEQ ID NO:1 or SEQ ID NO:2 or SEQ ID NO:4 or SEQ ID NO:5, or a fragment thereof. Still further, the LRP/LR may comprise homologues or fragments thereof, and homologues of the fragments, wherein the LRP/LR may comprise the peptide/protein sequence Listing as set forth in SEQ ID NO 1 or SEQ ID NO 2 or SEQ ID NO 4 or SEQ ID NO 5. The wound is preferably a skin lesion, but may also be a visceral wound.

Specific but non-limiting embodiments of the invention will now be described. Applicants contemplate further work including, but not limited to, in vivo and in vitro experiments.

Example (b):

the following examples serve to further illustrate the invention and are not limiting in its scope.

Example 1-Compounds for the prevention of Parkinson's disease.

List of abbreviations

AD Alzheimer's disease

ALP autophagy-lysosomal pathway

BCA bicinchoninic acid

BSA bovine serum albumin

CTD C-terminal Domain

DAPI diamidino-2-phenylindole

DLD-1 late colorectal cancer cell

DMEM Dulbecco modified eagle medium

DMSO dimethyl sulfoxide

EDTA ethylene diamine tetraacetic acid

ELISA enzyme-linked immunosorbent assay

FBS fetal bovine serum

HEK293 human embryonic Kidney

HRP horse radish peroxidase

hTERT human telomerase reverse transcriptase

L-Dopa L-Dopa

LRP/LR 37kDa laminin receptor precursor/67 kDa high affinity laminin receptor

MTT 3- (4, 5-Dimethylthiazol-2-yl) -2, 5-diphenyltetrazolium bromide

Non-amyloid-like protein-beta component of NAC plaque

PBS phosphate buffered saline

PCA protocatechuic acid

PD Parkinson's disease

P/S penicillin/streptomycin

PVDF polyvinylidene fluoride

RIPA radioimmunoprecipitation assay

SDS PAGE Polyacrylamide gel electrophoresis

SNPC black dense part

UPS ubiquitin-proteasome system

Experimental procedure (example 1):

cell culture

The HEK293 cell line was used because it is commonly used for Parkinson's Disease (PD) studies (schlachettzki et al, 2013). The cell culture medium consisted of Dulbecco's Modified Eagle Medium (DMEM) (Hyclone) and Ham's F12 in a ratio of 1: 1. The mixture was supplemented with 1% penicillin/streptomycin (P/S) and 15% Fetal Bovine Serum (FBS) to create a supplemented medium that would provide the cells with the necessary nutrients, including growth factors and P/S, to prevent bacterial and fungal contamination. The nutrient medium is renewed when necessary. The cells were cultured in an incubator maintained at humidity, pH, 37 ℃ and 5% CO2Atmospheric composition to simulate in vivo conditions. Once the cells reached 80% confluence, they were passaged by washing with Phosphate Buffered Saline (PBS) followed by resuspension by addition of trypsin-EDTA. After cell resuspension, media was added to inactivate trypsin, and the cell suspension was diluted into a new flask. Fresh medium was added to both flasks. Transfected and untransfected cells were harvested for further use. The SH-SY5Y cell line is another cell line widely used in PD studies because it displays a catecholaminergic phenotype. SH-SY5Y cells were also confirmed to be received after transfection. Cells were maintained under the same conditions as HEK293 cells. SH-SY5Y cells were used for MTT cell viability assay. LRP in the presence of 1-methyl-4-phenylpyridine iodide (MPP +) (PD inducer) FLAG overexpressing HEK293 cells was also studied. Late colorectal cancer (DLD-1) cells were used for confocal microscopy studies of certain parkin proteins.

Cell transfection

Transfection of cells is used to overexpress the protein of interest. Untransfected HEK293 cells will be used as controls. In this study, FLAG (Vana) was prepared by using a DNA construct, pCIneo-molLRP&Weiss, 2006), transfection was used to stably overexpress LRP:flag. By overexpressing LRP FLAG, the effect on alpha-synuclein can be determined. Once HEK293 cells reached 50-70% confluence according to the Clontech XfectTM transfection protocol, they were placed in T25 flasks (25 cm)2) In (c) performing transfection. Briefly, 10. mu.g of pCIneo-mol LRP FLAG plasmid DNA was made into a final volume of 200. mu.l of Xfet Reaction Buffer, followed by the addition of 3. mu.l of Xfet Polymer. Then will beThis solution was incubated at room temperature for 10 minutes to allow formation of nanoparticle complexes. The nanoparticle complexes containing plasmid DNA were then added to the sub-confluent cell cultures, followed by resuspension and humidified incubator at 37 ℃ and 5% CO2The cells were incubated for 48 hours. After 48 hours of incubation all media was removed and replaced with fresh complete growth media. The transfected cells were then treated with 800ng/ml Geneticin as a selective treatment and thereafter treated with 400ng/ml to maintain the transfected cell population.

Confocal microscope

Confocal microscopy is a visualization tool used to determine the localization of a fluorescently labeled protein of interest. Confocal microscopy was used in this study to determine whether LRP/LR co-localized with alpha-synuclein. In 6-well plates, cells were plated at 1.2X 10 per well5The density of individual cells was seeded on a coverslip (Labocare-18X18 mm; 0.19mm thick). The cells were then incubated at 37 ℃ and 5% CO in a humidified incubator2Incubate overnight to allow cells to attach. The remainder of the experiment was performed with gentle shaking. After incubation, cells were washed in 2ml 1 × PBS for 3 minutes. Cells were then fixed on coverslips with 2ml 4% formaldehyde for 20 minutes at room temperature. Once the cells were fixed, the cells were washed 3 times as above and then permeabilized with 3ml of 0.1% Triton X-100 for 20 minutes at room temperature. Cells were washed as described above and subsequently blocked with 2ml of 0.5% Bovine Serum Albumin (BSA) in 1XPBS for 25 minutes. Once the cells were blocked, a wash with 2ml PBS was performed once for 2 minutes. Primary antibodies were diluted in 500 μ l 0.5% BSA (1/200) and added to the cells, followed by incubation at 4 ℃ overnight. After incubation, cells were washed 3 times for 2 minutes each with 2ml of 0.5% BSA in 1 XPBS. The secondary antibody was then diluted in 500 μ l 0.5% BSA (1/500) and added to the cells, followed by incubation for one hour at room temperature in the dark. The cells were then washed 3 times with 2ml of 1X PBS in the dark for 2 minutes each. To stain the nuclei, 1ml of 0.1. mu.l/ml diamidino-2-phenylindole (DAPI) was added to the cells. The coverslips were then incubated at room temperature for 5 minutes in the dark, followed by washing 4 times with 2ml of 1X PBS in the dark for 2 minutes each. The cover glass is then mounted on the tapeGlass microscope slides with Sigma Fluorocount (Sigma) were incubated in the dark for one and a half hours. The samples were then stored at 4 ℃ until visualization was required using a Zeiss LSM 780 confocal microscope. See table 1 for a list of all antibodies used.

Table 1: list of antibodies used in confocal microscopy

Extraction of proteins from cells:

protein samples were extracted from cell lysates to determine protein concentration and western blot analysis was performed. To extract a protein sample from the cells, a cell lysate is prepared. Cells were first harvested by removing all media from the flask and washing with 5ml 1 XPBS. The cells were then resuspended in culture flasks by the addition of 2ml trypsin/EDTA and incubated at 37 ℃ for 5 minutes. To inactivate trypsin/EDTA, 8ml of medium was added to the flask and the resuspended cells were transferred to a 15ml Falcon centrifuge tube and centrifuged at 5000rpm for 10 minutes. The supernatant was then discarded, followed by addition of 100-. RIPA buffer consisted of 1% Triton X-100, 150mM NaCl, 0.1% Sodium Dodecyl Sulfate (SDS), 0.5% sodium deoxycholate, 50mM Tris-HCl (pH 8.0), and 5mM ethylenediaminetetraacetic acid (EDTA) (pH 8.0). The amount of RIPA buffer added depends on the size of the cell pellet. The pelleted cells were then resuspended in RIPA buffer and subsequently incubated at 4 ℃ for 15 minutes. To determine the protein concentration of the prepared cell lysates, BCA assays were performed prior to western blot analysis.

BCA assay

The total protein concentration from the cell lysate was determined by performing a bicinchoninic acid content assay (BCA). This determination is made by focusing on Cu2+Ions to determine protein concentration. When Cu2+When reduced by peptide bonds in proteins, Cu is formed+. Then, Cu+The ions form complexes with BCA, which can be generated byViolet product of absorbance measurement. Briefly, BSA protein standards were prepared at final concentrations of 0, 0.2, 0.4, 0.6, 0.8 and l mg/ml. 10 μ l of each standard was added to wells of a 96-well plate in triplicate. Protein samples of unknown concentration were then added in triplicate at 3 μ l per well. Distilled water was then added to the protein sample to reach a final volume of 10 μ Ι. BCA and CuSO4BCA and CuSO respectively429/30 and 1/30 ratios of (g) were added to each well. The samples were then incubated at 37 ℃ for 30 minutes. The absorbance at 562nm was read using an enzyme-linked immunosorbent assay (ELISA) microplate reader. BCA protein standard curves were constructed using the optical density of BSA protein standards. The protein concentration was then inferred using a standard curve. The samples were then normalized to 2mg/ml for western blot analysis.

SDS PAGE

Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS PAGE) analysis was performed to separate prepared protein samples according to protein molecular weight prior to western blotting. SDS binds to proteins, causing them to denature and to take a net negative charge. The total net charge will be proportional to the molecular weight of the protein. Electrophoresis of proteins causes the proteins to migrate from the negative end to the positive end. Migration allows proteins to pass through the pore according to their molecular weight. After quantification of the extracted proteins, protein samples were added to Blue Loading Buffer containing 40mM dithiothreitol to obtain alpha-synuclein and LRP-a 25. mu.g final protein concentration of FLAG and a 10. mu.g final protein concentration detected for LRP. For the detection of alpha-synuclein and LRP FLAG, 15% (w/v) polyacrylamide gel and 12% (w/v) polyacrylamide gel were used for the detection of LRP. Pink Plus Prestained Protein Ladder (GeneDireX) was loaded onto each gel together with the Protein samples. The proteins were then separated for 45 minutes at a voltage of 150V per gel. Western blot analysis was then performed.

Western blotting method

To determine the relative level of LRP:FLAG, LRP and alpha-synuclein western blotting was performed. Western blotting is used for immunological detection of proteins by using labeled antibodies. First, protein extraction was performed, followed by separation on SDS PAGE gels by electrophoresisA protein. After protein separation, the filter paper was soaked in 1X transfer buffer and polyvinylidene fluoride (PVDF) membrane was cut to size and activated in methanol. The transfer buffer consisted of 192mM glycine, 20% (w/v) methanol and 25mM Tris base. A semi-dry type electrophoretic transfer device is used. Three filter papers were placed on the transfer device followed by the PVDF membrane. The polyacrylamide gel was then placed on top of the PDVF film, followed by three additional filter papers. The transfer device was closed after filling to the desired amount with 1X transfer buffer. Proteins were transferred at 300mA for 50 min. The membranes were then blocked using 3% BSA in PBS-TWEEN for 1 hour, followed by five washes in 0.1% PBS-TWEEN, each for 5 minutes. The membrane was fixed with 0.4% formaldehyde before blocking to detect alpha-synuclein because of its small molecular weight. Primary antibodies were diluted in 3% BSA in PBS-TWEEN, added to the blot and incubated overnight at 4 ℃. Five additional washes (5 minutes each) were performed and the secondary antibody was diluted with 3% BSA, then added, followed by 1 hour incubation at room temperature. Five washes were then performed, followed by ClarityTMVisualization was performed by Western ECL Blotting Substrate (Bio-Rad) and ChemiDocTM imaging system (Bio-Rad). Densitometry analysis was performed using Image Lab 5.1 software (Bio-Rad) and all values were normalized to loading control β -actin.

Table 2: antibodies for detecting LRP in Western blotting FLAG, alpha-synuclein, and LRP/LR

MTT assay

The effect of treating cells with a particular substance or protein on cell viability can be determined by the MTT (3- (4, 5-dimethylthiazol-2-yl) -2, 5-diphenyltetrazolium bromide) assay (Riss et al, 2016). MTT is a yellow tetrazolium salt that is converted to purple formazan crystals by the mitochondrial enzyme succinate dehydrogenase (Stockert et al, 2012). Succinate dehydrogenase is only active in living cells, so only living cells can convert MTT to formazan (Stockert et al, 2012). The MTT assay is superior to it due to its high throughput screeningHe cell viability assays, such as ATP assays, azurin reduction assays, and protease viability marker assays. For this study, HEK293 cells were first plated at 1.4 × 10 per well4Individual cells were seeded in 24-well plates, whereas SH-SY5Y cells grew at 1.8X 10 cells due to their slower growth rate4And (4) inoculating the cells. The cells were then incubated overnight to allow for reattachment. Then 200nM and 500nM alpha-synuclein were treated again to induce PD-like state. After treatment, cells were incubated for 48 hours to determine the effect of α -synuclein on cell viability and incubated for 48 hours. Protocatechuic acid (PCA), a known inducer of apoptosis, was used as a positive control. After incubation, 200 μ l of 1mg/ml MTT was then added to each well and incubated at 37 ℃ for 2 hours to allow formazan crystals to form. The remaining MTT was discarded and the formazan crystals were dissolved by adding 200 μ l dimethyl sulfoxide (DMSO). Samples were then added in duplicate to a 96-well plate at 100 μ Ι and absorbance was read at 570nm using an ELISA plate reader. High absorbance indicates a large number of viable cells.

Statistical evaluation

Statistical evaluation of the data was performed by performing student t-tests with 95% confidence intervals, and therefore, the results were considered significant when the p-value was below 0.05 (p <0.05, p <0.01 and p <0.001) (Johnston, 1970).

Results (example 1)

Overexpression of LRP FLAG in HEK293 cells

The role of LRP/LR in PD has not been established. To determine the role of LRP/LR in PD, HEK293 cells were stably transfected to overexpress LRP:FLAG. pCIneo-molLRP:FLAGplasmid was used to achieve this. After cell transfection, western blot analysis was performed to confirm successful transfection (fig. 6A). In addition, total LRP protein levels were examined in transfected and untransfected HEK293 cell lines and loading controls (using β -actin) (fig. 2A).

Western blot analysis was performed to detect LRP:flagand determine total LRP protein levels in transfected and untransfected HEK293 cells. LRP:FLAGwas detected only in transfected HEK293 cells (FIG. 2A: lanes 4-6). Thus, transfection was successful and HEK293 transfected cells overexpressed LRP:FLAG. An increase in LRP/LR protein levels was seen when comparing LRP protein levels of untransfected (FIG. 2A: lanes 1-3) and transfected (FIG. 2A: lanes 4-6) HEK293 cells. Optical density and statistical analysis revealed a significant increase in total LRP levels and showed LRP:, over-expression of FLAG increased the LRP level by 71.24% (fig. 2B). A loading control (β -actin) was used.

Overexpression of FLAG in HEK293 cells increases alpha-synuclein levels and LRP/LR is co-localized with alpha-synuclein in HEK293 cells

To determine whether alpha-synuclein and LRP/LR co-localized, confocal microscopy was performed. Transfected HEK293 and untransfected cells were observed under a confocal microscope at magnification 630. Relative protein levels were also examined and compared in transfected and untransfected HEK293 cells. Co-localization of α -synuclein and LRP/LR occurred at the cell surface of the cytoplasm as well as untransfected and transfected HEK293 cells. Furthermore, FLAG transfected HEK293 cells were found to express more alpha-synuclein as shown by the increase in fluorescence intensity in FIG. 3H compared to FIG. 3A. Co-localization images confirmed co-localization between alpha-synuclein and LRP/LR and revealed that cells overexpressing LRP-FLAG had a greater degree of co-localization. The diagonal lines seen in the 2D cell fluorescence plots in fig. 3G and 3N also confirm co-localization between a-synuclein and LRP/LR.

10kDa CTD of alpha-synuclein was increased in LRP:FLAGoverexpressing cells

To determine the effect of over-expressing LRP, FLAG on α -synuclein levels, western blot analysis was performed on transfected and untransfected HEK293 cell lines (fig. 4). When comparing the α -synuclein levels in untransfected (FIG. 4A: lanes 1-3) and transfected (FIG. 4A: lanes 4-6) HEK293 cells, no difference in 19kDa full length α -synuclein protein levels was observed. However, the 10kDa CTD of alpha-synuclein increased. Densitometric analysis of western blots confirmed this and showed a 165.22% increase in CTD for α -synuclein levels (fig. 4B).

Over-expression of FLAG increases cell viability of HEK293 and SH-SY5Y cells treated with 200nM and 500nM alpha-synuclein

To determine the effect of over-expressing LRP, FLAG on cell viability of α -synuclein-treated cells, MTT assays were performed. MTT assays were performed on HEK293 and SH-SY5Y transfected and untransfected cell lines. Cells were treated with 200nM and 500nM alpha-synuclein to induce a PD-like state. MTT analysis showed a significant reduction in cell viability of HEK293 and SH-SY5Y transfected and untransfected cell lines when cells were treated with the positive control PCA (known inducer of apoptosis) (fig. 5 and 6), as expected. When we treated cells with 200nM and 500nM α -synuclein, cell viability of HEK293 untransfected cells was significantly reduced (fig. 5). However, no significant increase or decrease was seen in SH-SY5Y untransfected cells treated with 200nM and 500nM α -synuclein (FIG. 6). A slight increase in cell viability was seen in both HEK293 and SH-SY5Y LRP treated with 200nM and 500nM α -synuclein, however, this increase was not significant in FLAG transfected cells (FIGS. 5 and 6). Due to time limitations, the SH-SY5Y cell line was used only for MTT cell viability assays.

Over-expression of FLAG increased cell viability in HEK293 cells treated with 500uM and 750nM 1-methyl-4-phenylpyridine iodide (MPP +)

MTT analysis of FLAG-transfected and untransfected HEK293 cells was performed only, indicating that LRP:FLAGoverexpressing HEK293 cells had significantly increased cell viability in the presence of 500uM and 750nM 1-methyl-4-phenylpyridine iodide (MPP +) for 48 and 72 hours (see FIGS. 7A and 7B).

Discussion (example 1)

FLAG was overexpressed in the HEK293 in vitro PD model to determine if there was a relationship between LRP/LR and alpha-synuclein and LRP/LR and parkin withdrawal.

LRP/LR colocalization with alpha-synuclein and parkin

Once LRP:FLAGexpression in HEK293 cells was confirmed by Western blot analysis (FIG. 2), LRP levels were examined. Western blot analysis confirmed that over-expression of FLAG caused an increase in total LRP protein levels of 71.24% compared to untransfected cells (FIG. 2). Thereafter, confocal microscopy was performed to determine whether LRP/LR and alpha-synuclein co-localized. Confocal microscopy demonstrated that LRP/LR and a-synuclein were confocal on both the cytoplasm and cell surface of untransfected HEK293 cells (figure 3). Co-localization was confirmed by pooling, co-localization and diagonal lines in the 2D cell fluorescence image. Several studies have demonstrated that alpha-synuclein is predominantly present in the cytoplasm (Guardia-Laguarta et al, 2015). Other studies have shown that α -synuclein is able to bind to membranes after stimulation (Eliezer et al, 2001; Jao et al, 2004). It is not clear at present what the exact stimulus is. This provides evidence that there is an interaction between LRP/LR and a-synuclein, as they are located in the same subcellular compartment. Without being limited by theory, LRP/LR may act as a stimulator of membrane binding.

Confocal microscopy revealed not only co-localization between LRP/LR and alpha-synuclein, but also LRP-LRP/LR and alpha-synuclein levels increased after FLAG transfection (FIG. 4). Because of the increased protein levels of alpha-synuclein and LRP/LR, co-localization between LRP/LR and alpha-synuclein was more apparent in LRP:FLAGoverexpressing HEK293 cells compared to untransfected HEK293 cells. The increase in α -synuclein levels observed in LRP:FLAGtransfected HEK293 cells is further evidence that there may be an interaction between LRP/LR and α -synuclein.

In FIG. 3, the primary antibodies used were raised against LRP and alpha-synuclein. The Cy 3-coupled primary antibody was used to detect FLAG. Secondary antibody was conjugated to FITC and alexaflur 647. Panels A and H represent α -synuclein in red, while Panels B and I represent LRP/LR in green. LRP:FLAGis shown in panels C and J. Panels E and L represent the pooling of alpha-synuclein and LRP/LR. Yellow fluorescence indicates the presence of overlap and possible interaction between alpha-synuclein and LRP/LR. Panels a and B: LRP and a-synuclein are predominantly expressed on the cell surface where co-localization occurs. Panels H and I: pCIneo-LRP-LRP/LR and α -synuclein levels were increased after FLAG transfection. LRP/LR co-localized in HEK293 cells (panels E-F) and this effect was more pronounced in LRP:, FLAG overexpressing cells (L-N). Co-localization occurred on the cell surface and within the cell as shown by the diagonal lines in the 2D cell fluorescence maps (G and N).

Figure 8 shows a confocal microscope at 630 magnification, indicating the expression levels and localization of LRP and parkin. The primary antibody used was raised against LRP, parkin and hTERT. Secondary antibody was conjugated to FITC, Cy3 and alexaflur 647. Panels A to F represent DLD-1 cells, and Panels G to L represent DLD-1 cells that have been transfected with pCIneo-LRP FLAG plasmid. Pangin is shown in yellow for panels A and G, while LRP/LR is shown in green for panels B and H. Panels D and I show the combination of parkin and LRP/LR. Panels E and K represent co-localization of the two proteins, with white indicating overlap and possible interaction between parkin and LRP/LR. Co-localization occurred intracellularly, as shown by the diagonal in the 2D cell fluorescence map (F), where 59% of the parkin was co-localized with 54% of the LRP/LR in DLD-1 cells, and 99% of the parkin was co-localized with 22% of the LRP in DLD-1 cells overexpressing LRP:FLAG. This supports that overexpression of LRP/LR increases parkin levels in cells.

FLAG overexpression increases CTD of alpha-synuclein and increases parkin

Western blot analysis was also used to confirm the increase in alpha-synuclein observed in confocal microscopy. However, no increase in the full-length 19kDa alpha-synuclein level was observed after LRP:, FLAG transfection (FIG. 5). Interestingly, in LRP:FLAGtransfected HEK293 cells, a 165.22% increase was seen in the second band below the full length 19kDa alpha-synuclein. Previous studies showed that the CTD of α -synuclein is 10kDa and migrates below the full-length 19kDa α -synuclein (Xu et al, 2015). CTD plays an important role in preventing alpha-synuclein aggregation by interacting with the NAC domain (emazadeh, 2016). Most studies have focused on NTD of α -synuclein, since many mutations are found in this domain, and thus CTD is not much studied (Xu & Pu 2016). A recent study demonstrated that truncation of CTD alters the mechanism of α -synuclein aggregation and the properties of the fibrils formed (van der Wateren et al, 2018). The same study also demonstrated that truncation of CTD favours alpha-synuclein aggregation (van der Wateren et al, 2018). This indicates that the results seen from western blot in this study are evidence that LRP/LR increases CTD of a-synuclein, which prevents a-synuclein aggregation.

FLAG overexpression increases cell viability

To determine whether the CTD increase of alpha-synuclein was positive or negative, MTT cell viability assays were performed. MTT assays were performed on HEK293 untransfected and transfected and SH-SY5Y untransfected and transfected cell lines. The SH-SY5Y cell line is widely used in PD studies because it produces both dopamine and norepinephrine, thereby showing a catecholaminergic phenotype (Xicoy et al, 2017). Treatment of LRP with 200nM and 500nM α -synuclein FLAG over-expressed and untransfected HEK293 cells caused a slight decrease in cell viability, however, no decrease was observed in LRP:FLAGover-expressed and untransfected SH-SY5Y cells. Interestingly, a slight increase in cell viability was observed in both HEK293 and SH-SY5Y cell lines when comparing cell viability of LRP overexpressing FLAG versus untransfected cells. When LRP:flagwas overexpressed, no negative effects were observed in cell viability. However, these results may indicate that the CTD increase of α -synuclein following LRP:FLAGtransfection and thus over-expression of FLAG protects cells from α -synuclein treatment. CTD is crucial for maintaining the structure of full-length α -synuclein and prevents conformational changes important during fibrosis (Hong et al, 2011). The mechanism behind this protection needs to be investigated.

MTT assay analysis of FLAG-transfected and untransfected HEK293 cells indicated that cell viability of LRP:FLAGoverexpressing HEK293 cells was significantly increased in the presence of 500uM and 750nM 1-methyl-4-phenylpyridine iodide (MPP +) for 48 hours (as shown in FIG. 7A) and 72 hours (as shown in FIG. 7B).

Conclusion (example 1)

In summary, LRP/LR increases the C-terminus of alpha-synuclein, thereby preventing accumulation and/or aggregation and thus providing effective compounds for the treatment of Parkinson's Disease (PD). Applicants demonstrated that LRP/LR not only colocalizes with alpha-synuclein, but that over-expression of FLAG increased the level of CTD. Any observed increase in CTD has a protective effect on cells treated with alpha-synuclein. LRP/LR co-localization with parkin was also shown. Therefore, therapies aimed at increasing LRP/LR protein levels may be potential treatments for PD.

Reference (example 1)

Ahmed,S.,Passos,J.F.,Birket,M.J.,Beckmann,T.,Brings,S.,Peters,H.,Birch-Machin,M.A.,von Zglinicki,T.and Saretzki,G.,2008.Telomerase does not counteract telomere shortening but protects mitochondrial function under oxidative stress.Journal of cell science,121(7),pp.1046-1053.

Ardini,E.,Pesole,G.,Tagliabue,E.,Magnifico,A.,Castronovo,V.,Sobel,M.E.,Colnaghi,M.I.and Menard,S.,1998.The 67-kDa laminin receptor originated from a ribosomal protein that acquired a dual function during evolution.Molecular biology and evolution,15(8),pp.1017-1025.

Auluck,P.K.,Caraveo,G.and Lindquist,S.,2010.a-Synuclein:membrane interactions and toxicity in Parkinson's disease.Annual review of cell and developmental biology,26,pp.211-233.

Bendor,J.T.,Logan,T.P.and Edwards,R.H.,2013.The function of a-Synuclein.Neuron,79(6),pp.1044-1066.

Chai,C.and Lim,K.L.,2013.Genetic insights into sporadic Parkinson's disease pathogenesis.Current genomics,14(8),pp.486-501.

Chauhan,A.and Jeans,A.F.,2015.Is Parkinson’s disease truly a prion-like disorderAn appraisal of current evidence.Neurology research international,2015.

Chen,M.S.,Fornace Jr,A.and Laszlo,A.,1996.Characterization of an hsp70 related clone encoding a 33 kDa protein with homology to a protein which associates with polysomes.Biochimica et Biophysica Acta(BBA)-Protein Structure and Molecular Enzymology,1297(2),pp.124-126.

Chetty,C.J.,Ferreira,E.,Jovanovic,K.and Weiss,S.F.,2017.Knockdown of LRP/LR induces apoptosis in pancreatic cancer and neuroblastoma cells through activation of caspases.Experimental cell research,360(2),pp.264-272.

Dauer,W.and Przedborski,S.,2003.Parkinson's disease:mechanisms and models.Neuron,39(6),pp.889-909.

Dexter,D.T.and Jenner,P.,2013.Parkinson disease:from pathology to molecular disease mechanisms.Free Radical Biology and Medicine,62,pp.132-144.

Dias,B.D.C.,Jovanovic,K.,Gonsalves,D.,Moodley,K.,Reusch,U.,Knackmuss,S.,Penny,C.,Weinberg,M.S.,Little,M.and Weiss,S.F.,2013.Anti-LRP/LR specific antibody IgGl-iS18 and knock-down of LRP/LR by shRNAs rescue cells from Ap 42 induced cytotoxicity.Scientific reports,3,p.2702.

Dias,B.D.C.,Jovanovic,K.,Gonsalves,D.,Moodley,K.,Reusch,U.,Knackmuss,S.,Weinberg,M.S.,Little,M.and Weiss,S.F.,2014.The 37 kDa/67 kDa laminin receptor acts as a receptor for Ap 42 internalization.Scientific reports,4,p.5556.

Dickson,D.W.,2012.Parkinson’s disease and parkinsonism:neuropathology.Cold Spring Harbor perspectives in medicine,p.a009258.

DiGiacomo,V.and Meruelo,D.,2016.Looking into laminin receptor:critical discussion regarding the non-integrin 37/67-kDa laminin receptor/RPSA protein.Biological Reviews,91(2),pp.288-310.

Dolgacheva,L.P.,Fedotova,E.I.,Abramov,A.Y.and Berezhnov,A.V.,2018.Alpha-Synuclein and Mitochondrial Dysfunction in Parkinson’s Disease.Biochemistry (Moscow),Supplement Series A:Membrane and Cell Biology,12(1),pp.10-19.

Eliezer,D.,Kutiuay,E.,Bussell Jr,R.and Browne,G.,2001.Conformational properties of a-Synuclein in its free and lipid-associated statesl.Journal of molecular biology,307(4),pp.1061-1073.

Emamzadeh,F.N.,2016.Alpha-Synuclein structure,functions,and interactions.Journal of research in medical sciences:the official journal of Isfahan University of Medical Sciences,21.

Ferreira,E.,Bignoux,M.J.,Otgaar,T.C.,Tagliatti,N.,Jovanovic,K.,Letsolo,B.T.and Weiss,S.F.,2018.LRP/LR specific antibody IgGl-iS18 impedes neurodegeneration in Alzheimer's disease mice.Oncotarget,9(43),p.27059.

Ford CL,Randal-Whitis L,Ellis SR.Yeast proteins related to the p40/laminin receptor precursor are required for 20S ribosomal RNA processing and the maturation of 40S ribosomal subunits.Cancer Res 1999;59(3):704-10

Fu,H.,Subramanian,R.R.and Masters,S.C.,2000.14-3-3 proteins:structure,function,and regulation.Annual review of pharmacology and toxicology,40(1),pp.617-647.

Gauczynski,S.,Nikles,D.,El-Gogo,S.,Papy-Garcia,D.,Rey,C.,Alban,S.,Barritault,D.,Lasmezas,C.I.and Weiss,S.,2006.The 37-kDa/67-kDa laminin receptor acts as a receptor for infectious prions and is inhibited by polysulfated glycanes.The Journal of infectious diseases,194(5),pp.702-709.

Gauczynski,S.,Nikles,D.,El-Gogo,S.,Papy-Garcia,D.,Rey,C.,Alban,S.,Barritault,D.,Lasmezas,C.I.and Weiss,S.,2006.The 37-kDa/67-kDa laminin receptor acts as a receptor for infectious prions and is inhibited by polysulfated glycanes.The Journal of infectious diseases,194(5),pp.702-709.

Guardia-Laguarta,C.,Area-Gomez,E.,Schon,E.A.and Przedborski,S.,2015.Novel subcellular localization for a-Synuclein:possible functional consequences.Frontiers in neuroanatomy,9,p.17.

Hong,D.P.,Xiong,W.,Chang,J.Y.and Jiang,C.,2011.The role of the C-terminus of human a-Synuclein:Intra-disulfide bonds between the C-terminus and other regions stabilize non-fibrillar monomeric isomers.FEBS letters,585(3),pp.561-566.

Jao,C.C.,Der-Sarkissian,A.,Chen,J.and Langen,R.,2004.Structure of membrane-bound a-Synuclein studied by site-directed spin labeling.Proceedings of the National Academy of Sciences,101(22),pp.8331-8336.

Jenner,P.,2003.Oxidative stress in Parkinson's disease.Annals of neurology,53(S3).Jones,D.R.,Moussaud,S.and McLean,P.,2014.Targeting heat shock proteins to modulate a-synuclein toxicity.Therapeutic advances in neurological disorders,7(1),pp.33-51.

Jovanovic,K.,Gonsalves,D.,Dias,B.D.C.,Moodley,K.,Reusch,U.,Knackmuss,S.,Penny,C.,Weinberg,M.S.,Little,M.and Weiss,S.F.,2013.Anti-LRP/LR specific antibodies and shRNAs impede amyloid beta shedding in Alzheimer's disease.Scientific reports,3,p.2699.

Jovanovic,K.,Chetty,C.J.,Khumalo,T.,Da Costa Dias,B.,Ferreira,E.,Malindisa,S.T.,Caveney,R.,Letsolo,B.T.and Weiss,S.F.,2015.Novel patented therapeutic approaches targeting the 37/67 kDa laminin receptor for treatment of cancer and Alzheimer’s disease.Expert opinion on therapeutic patents,25(5),pp.567-582.

Khumalo,T.,Ferreira,E.,Jovanovic,K.,Veale,R.B.and Weiss,S.F.,2015.Knockdown of LRP/LR induces apoptosis in breast and oesophageal cancer cells.PloS one,10(10),p.e0139584.

Khusal,R.,Dias,B.D.C.,Moodley,K.,Penny,C.,Reusch,U.,Knackmuss,S.,Little,M.and Weiss,S.F.,2013.In vitro inhibition of angiogenesis by antibodies directed against the 37 kDa/67 kDa laminin receptor.PloS one,8(3),p.e58888.

Kinoshita K,Kaneda Y,Sato M,et al.LBP-p40 binds DNA tightly through associations with histones H2A,H2B,and H4.BiochemBiophys Res Commun 1998;253(2):277-82.Kitada,T.,Asakawa,S.,Hattori,N.,Matsumine,H.,Yamamura,Y.,Minoshima,S.,Yokochi,M.,Mizuno,Y.and Shimizu,N.,1998.Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism.Nature,392(6676),p.605.

Klucken,J.,Shin,Y.,Masliah,E.,Hyman,B.T.and McLean,P.J.,2004.Hsp70 reduces a-synuclein aggregation and toxicity.Journal of Biological Chemistry,279(24),pp.25497-25502.

Lees,A.,2017.An essay on the shaking palsy.Brain,140(3),pp.843-848.

Leucht,C.,Simoneau,S.,Rey,C.,Vana,K.,Rieger,R.,Lasmezas,C.I.and Weiss,S.,2003.The 37 kDa/67 kDa laminin receptor is required for PrPSc propagation in scrapie-infected neuronal cells.EMBO reports,4(3),pp.290-295.

Lima,M.M.,Reksidler,A.B.and Vital,M.A.,2009.The neurobiology of the substantia nigra pars compacta:from motor to sleep regulation.In Birth,Life and Death of Dopaminergic Neurons in the Substantia Nigra(pp.135-145).Springer,Vienna.

Mandel,S.A.,Morelli,M.,Halperin,I.and Korczyn,A.D.,2010.Biomarkers for prediction and targeted prevention of Alzheimer’s and Parkinson’s diseases:evaluation of drug clinical efficacy.EPMA Journal,1(2),pp.273-292.

Marinus,J.,Zhu,K.,Marras,C.,Aarsland,D.and van Hilten,J.J.,2018.Risk factors for non-motor symptoms in Parkinson's disease.The Lancet Neurology.

Mbazima,V.,Da Costa Dias,B.,Omar,A.,Jovanovic,K.and Weiss,S.F.,2010.Interactions between PrP(c)and other ligands with the 37-kDa/67-kDa laminin receptor.Front Biosci,15(1150-1163),p.3667.

Muzerengi,S.and Clarke,C.E.,2015.Initial drug treatment in Parkinson’s disease.Bmj,351,p.h4669.

Oertel,W.and Schulz,J.B.,2016.Current and experimental treatments of Parkinson disease:A guide for neuroscientists.Journal of neurochemistry,139,pp.325-337.

Omar,A.,Reusch,U.,Knackmuss,S.,Little,M.and Weiss,S.F.,2012.Anti-LRP/LR-specific antibody IgGl-iS18 significantly reduces adhesion and invasion of metastatic lung,cervix,colon and prostate cancer cells.Journal of molecular biology,419(1-2),pp.102-109.

Otgaar,T.C.,Ferreira,E.,Malindisa,S.,Bemert,M.,Letsolo,B.T.and Weiss,S.F.,2017.37kDa LRP::FLAG enhances telomerase activity and reduces senescent markers in vitro.Oncotarget,8(49),p.86646.

Palacino,J.J.,Sagi,D.,Goldberg,M.S.,Krauss,S.,Motz,C.,Wacker,M.,Klose,J.and Shen,J.,2004.Mitochondrial dysfunction and oxidative damage in parkin-deficient mice.Journal of Biological Chemistry,279(18),pp.18614-18622.

Rao,N.C.,Barsky,S.H.,Terranova,V.P.and Liotta,L.A.,1983.Isolation of a tumor cell laminin receptor.Biochemical and biophysical research communications,111(3),pp.804-808.

Rieger,R.,Edenhofer,F.,Lasmezas,C.I.and Weiss,S.,1997.The human 37-kDa laminin receptor precursor interacts with the prion protein in eukaryotic cells.Nature medicine,3(12),pp.1383-1388.

Riss,T.L.,Moravec,R.A.,Niles,A.L.,Duellman,S.,Benink,H.A.,Worzella,T.J.and Minor,L.,2016.Cell viability assays.

Rocha,E.M.,De Miranda,B.and Sanders,L.H.,2018.Alpha-Synuclein:pathology,mitochondrial dysfunction and neuroinflammation in Parkinson’s disease.Neurobiology of disease,109,pp.249-257.

Rodriguez-Araujo,G.,Nakagami,H.,Takami,Y.,Katsuya,T.,Akasaka,H.,Saitoh,S.,Shimamoto,K.,Morishita,R.,Rakugi,H.and Kaneda,Y.,2015.Low alpha-Synuclein levels in the blood are associated with insulin resistance.Scientific reports,5,p.12081.

Savitt,J.M.,Dawson,V.L.and Dawson,T.M.,2006.Diagnosis and treatment of Parkinson disease:molecules to medicine.The Journal of clinical investigation,116(7),pp.1744-1754.Schlachetzki,J.,Saliba,S.W.and Oliveira,A.C.P.D.,2013.Studying neurodegenerative diseases in culture models.Revista brasileira de psiquiatria,35,pp.S92-S100.

Seirafi,M.,Kozlov,G.and Gehring,K.,2015.Parkin structure and function.The FEBSjoumal,282(11),pp.2076-2088.

Shimura,H.,Hattori,N.,Kubo,S.I.,Mizuno,Y.,Asakawa,S.,Minoshima,S.,Shimizu,N.,Iwai,K.,Chiba,T.,Tanaka,K.and Suzuki,T.,2000.Familial Parkinson disease gene product,parkin,is a ubiquitin-protein ligase.Nature genetics,25(3),p.302.

Sidransky,E.,Samaddar,T.and Tayebi,N.,2009.Mutations in GBAare associated with familial Parkinson disease susceptibility and age at onset.Neurology,73(17),pp.1424-1426.

Song,P.,Trajkovic,K.,Tsunemi,T.and Krainc,D.,2016.Parkin modulates endosomal organization and function of the endo-lysosomal pathway.Journal of Neuroscience,36(8),pp.2425-2437.

Stefanis,L.,2012.a-Synuclein in Parkinson's disease.Cold Spring Harbor perspectives in medicine,2(2),p.a009399.

Stockert,J.C.,Blazquez-Castro,A.,Canete,M.,Horobin,R.W.and Villanueva,A.,2012.MTT assay for cell viability:Intracellular localization of the formazan product is in lipid droplets.Acta histochemica,114(8),pp.785-796.

Tarakad,A.and Jankovic,J.,2017,April.Diagnosis and management of Parkinson's disease.In Seminars in neurology(Vol.37,No.02,pp.118-126).Thieme Medical Publishers.

Vamvaca K.,Voiles M.J.and Lansbury P.T.(2009)The first Nterminal amino acids of alpha-Synuclein are essential for alphahelical structure formation in vitro and membrane binding in yeast.J.Mol.Biol.389,413-424.

Vana,K.and Weiss,S.,2006.A trans-dominant negative 37 kDa/67 kDa laminin receptor mutant impairs PrPSc propagation in scrapie-infected neuronal cells.Journal of molecular biology,358(1),pp.57-66.

van der Wateren,I.M.,Knowles,T.,Buell,A.,Dobson,C.M.and Galvagnion,C.,2018.C-terminal truncation of a-Synuclein promotes amyloid fibril amplification at physiological pH.Chemical Science.

Wakabayashi,K.,Tanji,K.,Mori,F.and Takahashi,H.,2007.The Lewy body in Parkinson's disease:Molecules implicated in the formation and degradation of a-synuclein aggregates.Neuropathology,27(5),pp.494-506.

Weiss,S.F.,2017.Bad Boy with a Twist:Targeting the 37 kDa/67 kDa Laminin Receptor for Treatment of Cancer and Neurodegenerative Diseases and for Changing Telomere Dynamics.Cell Cell.Life Sci.J.,2,p.000114.

Xicoy,H.,Wieringa,B.and Martens,G.J.,2017.The SH-SY5Y cell line in Parkinson’s disease research:a systematic review.Molecular neurodegeneration,12(1),p.10.

Xu,B.,Liu,W.,Deng,Y.,Yang,T.Y.,Feng,S.and Xu,Z.F.,2015.Inhibition of calpain prevents manganese-induced cell injury and alpha-Synuclein oligomerization in organotypic brain slice cultures.PloS one,10(3),p.eOl 19205.

Xu,L.and Pu,J.,2016.Alpha-Synuclein in Parkinson’s disease:from pathogenetic dysfunction to potential clinical application.Parkinson’s Disease,2016.

Yang,Y.,Nishimura,I.,Imai,Y.,Takahashi,R.and Lu,B.,2003.Parkin suppresses dopaminergic neuron-selective neurotoxicity induced by Pael-R in Drosophila.Neuron,37(6),pp.911-924.

Yasuda,T.and Mochizuki,H.,2010.The regulatory role of a-Synuclein and parkin in neuronal cell apoptosis;possible implications forthe pathogenesis of Parkinson’s disease.Apoptosis,15(11),pp.1312-1321.

Applicants believe that the present invention provides at least new and inventive compounds, pharmaceutical compositions and/or methods for treating and/or preventing Parkinson's Disease (PD).

It is very surprising for the applicant that providing LRP/LR, i.e. up-regulating its expression, to the human or animal body increases the C-terminal domain (CTD) of alpha-synuclein. As the prior art teaches down-regulating LRP/LR in the treatment of another neurodegenerative disease, namely Alzheimer's Disease (AD), applicants do not anticipate that up-regulating LRP/LR (or providing to the human or animal body) would have any positive impact on neurodegenerative diseases such as PD.

Providing LRP/LR as part of a PD treatment regimen will significantly limit the chance of any unwanted side effects as it is a naturally occurring protein/peptide. This certainly improves at least one of the problems known in the prior art. While the invention has been described in detail with respect to specific embodiments and/or examples thereof, it will be appreciated that those skilled in the art, upon attaining an understanding of the foregoing, may readily conceive of alterations to, variations of, and equivalents to these embodiments. The scope of the invention should, therefore, be assessed as that of the appended claims and any equivalents thereto.

Sequence listing

<110> university of John Nernsberg Witwortland

(University of the Witwatersrand, Johannesburg)

<120> Compounds for the treatment of Parkinson's disease

<130> P1200PC

<140>

<141>

<150> 2018/08025

<151> 2018-11-28

<160> 5

<170> PatentIn version 3.5

<210> 1

<211> 295

<212> PRT

<213> human (Homo sapiens)

<400> 1

Met Ser Gly Ala Leu Asp Val Leu Gln Met Lys Glu Glu Asp Val Leu

1 5 10 15

Lys Phe Leu Ala Ala Gly Thr His Leu Gly Gly Thr Asn Leu Asp Phe

20 25 30

Gln Met Glu Gln Tyr Ile Tyr Lys Arg Lys Ser Asp Gly Ile Tyr Ile

35 40 45

Ile Asn Leu Lys Arg Thr Trp Glu Lys Leu Leu Leu Ala Ala Arg Ala

50 55 60

Ile Val Ala Ile Glu Asn Pro Ala Asp Val Ser Val Ile Ser Ser Arg

65 70 75 80

Asn Thr Gly Gln Arg Ala Val Leu Lys Phe Ala Ala Ala Thr Gly Ala

85 90 95

Thr Pro Ile Ala Gly Arg Phe Thr Pro Gly Thr Phe Thr Asn Gln Ile

100 105 110

Gln Ala Ala Phe Arg Glu Pro Arg Leu Leu Val Val Thr Asp Pro Arg

115 120 125

Ala Asp His Gln Pro Leu Thr Glu Ala Ser Tyr Val Asn Leu Pro Thr

130 135 140

Ile Ala Leu Cys Asn Thr Asp Ser Pro Leu Arg Tyr Val Asp Ile Ala

145 150 155 160

Ile Pro Cys Asn Asn Lys Gly Ala His Ser Val Gly Leu Met Trp Trp

165 170 175

Met Leu Ala Arg Glu Val Leu Arg Met Arg Gly Thr Ile Ser Arg Glu

180 185 190

His Pro Trp Glu Val Met Pro Asp Leu Tyr Phe Tyr Arg Asp Pro Glu

195 200 205

Glu Ile Glu Lys Glu Glu Gln Ala Ala Ala Glu Lys Ala Val Thr Lys

210 215 220

Glu Glu Phe Gln Gly Glu Trp Thr Ala Pro Ala Pro Glu Phe Thr Ala

225 230 235 240

Thr Gln Pro Glu Val Ala Asp Trp Ser Glu Gly Val Gln Val Pro Ser

245 250 255

Val Pro Ile Gln Gln Phe Pro Thr Glu Asp Trp Ser Ala Gln Pro Ala

260 265 270

Thr Glu Asp Trp Ser Ala Ala Pro Thr Ala Gln Ala Thr Glu Trp Val

275 280 285

Gly Ala Thr Thr Asp Trp Ser

290 295

<210> 2

<211> 295

<212> PRT

<213> little mouse (Mus musculus)

<400> 2

Met Ser Gly Ala Leu Asp Val Leu Gln Met Lys Glu Glu Asp Val Leu

1 5 10 15

Lys Phe Leu Ala Ala Gly Thr His Leu Gly Gly Thr Asn Leu Asp Phe

20 25 30

Gln Met Glu Gln Tyr Ile Tyr Lys Arg Lys Ser Asp Gly Ile Tyr Ile

35 40 45

Ile Asn Leu Lys Arg Thr Trp Glu Lys Leu Leu Leu Ala Ala Arg Ala

50 55 60

Ile Val Ala Ile Glu Asn Pro Ala Asp Val Ser Val Ile Ser Ser Arg

65 70 75 80

Asn Thr Gly Gln Arg Ala Val Leu Lys Phe Ala Ala Ala Thr Gly Ala

85 90 95

Thr Pro Ile Ala Gly Arg Phe Thr Pro Gly Thr Phe Thr Asn Gln Ile

100 105 110

Gln Ala Ala Phe Arg Glu Pro Arg Leu Leu Val Val Thr Asp Pro Arg

115 120 125

Ala Asp His Gln Pro Leu Thr Glu Ala Ser Tyr Val Asn Leu Pro Thr

130 135 140

Ile Ala Leu Cys Asn Thr Asp Ser Pro Leu Arg Tyr Val Asp Ile Ala

145 150 155 160

Ile Pro Cys Asn Asn Lys Gly Ala His Ser Val Gly Leu Met Trp Trp

165 170 175

Met Leu Ala Arg Glu Val Leu Arg Met Arg Gly Thr Ile Ser Arg Glu

180 185 190

His Pro Trp Glu Val Met Pro Asp Leu Tyr Phe Tyr Arg Asp Pro Glu

195 200 205

Glu Ile Glu Lys Glu Glu Gln Ala Ala Ala Glu Lys Ala Val Thr Lys

210 215 220

Glu Glu Phe Gln Gly Glu Trp Thr Ala Pro Ala Pro Glu Phe Thr Ala

225 230 235 240

Ala Gln Pro Glu Val Ala Asp Trp Ser Glu Gly Val Gln Val Pro Ser

245 250 255

Val Pro Ile Gln Gln Phe Pro Thr Glu Asp Trp Ser Ala Gln Pro Ala

260 265 270

Thr Glu Asp Trp Ser Ala Ala Pro Thr Ala Gln Ala Thr Glu Trp Val

275 280 285

Gly Ala Thr Thr Glu Trp Ser

290 295

<210> 3

<211> 8

<212> PRT

<213> Escherichia coli (Escherichia coli)

<400> 3

Asp Tyr Lys Asp Asp Asp Asp Lys

1 5

<210> 4

<211> 194

<212> PRT

<213> human (Homo sapiens)

<400> 4

Arg Phe Thr Pro Gly Thr Phe Thr Asn Gln Ile Gln Ala Ala Phe Arg

1 5 10 15

Glu Pro Arg Leu Leu Val Val Thr Asp Pro Arg Ala Asp His Gln Pro

20 25 30

Leu Thr Glu Ala Ser Tyr Val Asn Leu Pro Thr Ile Ala Leu Cys Asn

35 40 45

Thr Asp Ser Pro Leu Arg Tyr Val Asp Ile Ala Ile Pro Cys Asn Asn

50 55 60

Lys Gly Ala His Ser Val Gly Leu Met Trp Trp Met Leu Ala Arg Glu

65 70 75 80

Val Leu Arg Met Arg Gly Thr Ile Ser Arg Glu His Pro Trp Glu Val

85 90 95

Met Pro Asp Leu Tyr Phe Tyr Arg Asp Pro Glu Glu Ile Glu Lys Glu

100 105 110

Glu Gln Ala Ala Ala Glu Lys Ala Val Thr Lys Glu Glu Phe Gln Gly

115 120 125

Glu Trp Thr Ala Pro Ala Pro Glu Phe Thr Ala Thr Gln Pro Glu Val

130 135 140

Ala Asp Trp Ser Glu Gly Val Gln Val Pro Ser Val Pro Ile Gln Gln

145 150 155 160

Phe Pro Thr Glu Asp Trp Ser Ala Gln Pro Ala Thr Glu Asp Trp Ser

165 170 175

Ala Ala Pro Thr Ala Gln Ala Thr Glu Trp Val Gly Ala Thr Thr Asp

180 185 190

Trp Ser

<210> 5

<211> 194

<212> PRT

<213> little mouse (Mus musculus)

<400> 5

Arg Phe Thr Pro Gly Thr Phe Thr Asn Gln Ile Gln Ala Ala Phe Arg

1 5 10 15

Glu Pro Arg Leu Leu Val Val Thr Asp Pro Arg Ala Asp His Gln Pro

20 25 30

Leu Thr Glu Ala Ser Tyr Val Asn Leu Pro Thr Ile Ala Leu Cys Asn

35 40 45

Thr Asp Ser Pro Leu Arg Tyr Val Asp Ile Ala Ile Pro Cys Asn Asn

50 55 60

Lys Gly Ala His Ser Val Gly Leu Met Trp Trp Met Leu Ala Arg Glu

65 70 75 80

Val Leu Arg Met Arg Gly Thr Ile Ser Arg Glu His Pro Trp Glu Val

85 90 95

Met Pro Asp Leu Tyr Phe Tyr Arg Asp Pro Glu Glu Ile Glu Lys Glu

100 105 110

Glu Gln Ala Ala Ala Glu Lys Ala Val Thr Lys Glu Glu Phe Gln Gly

115 120 125

Glu Trp Thr Ala Pro Ala Pro Glu Phe Thr Ala Ala Gln Pro Glu Val

130 135 140

Ala Asp Trp Ser Glu Gly Val Gln Val Pro Ser Val Pro Ile Gln Gln

145 150 155 160

Phe Pro Thr Glu Asp Trp Ser Ala Gln Pro Ala Thr Glu Asp Trp Ser

165 170 175

Ala Ala Pro Thr Ala Gln Ala Thr Glu Trp Val Gly Ala Thr Thr Glu

180 185 190

Trp Ser

39页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:纤毛疾病的治疗

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!