Use of peptides inhibitory for thrombospondin dependent...

Drug – bio-affecting and body treating compositions – Designated organic active ingredient containing – Peptide containing doai

Reexamination Certificate

Rate now

  [ 0.00 ] – not rated yet Voters 0   Comments 0

Details

Reexamination Certificate

active

06458767

ABSTRACT:

FIELD OF THE INVENTION
The present invention generally relates to a method of regulating transforming growth factor TGF-&bgr; (TGF-&bgr;) activity. More particularly, the present invention relates to a method of interfering with the activation of TGF-&bgr; by thrombospondin through the administration of peptides in order to treat kidney diseases and/or conditions.
BACKGROUND OF THE INVENTION
Extracellular matrix accumulation is one of the hallmarks of inflammatory glomerular disease as a major cause of end-stage renal disease in man. Mesangial proliferative glomerulonephritis, the most common type of glomerulonephritis in the Western World (D'Amico (1987)
QJM
245, 709-727), is characterized by mesangial cell (MC) proliferation, activation, and extracellular matrix expansion (Johnson (1994)
Kidney Int
. 45 (6), 1769-82). In up to 50% of the patients with mesangial proliferative glomerulonephritis, the disease process eventually progresses to end-stage renal disease, since specific treatment is still lacking (Galla (1995)
Kidney Int
. 47, 377-387). Typical features of human mesangial proliferative glomerulonephritis are mimicked by an experimental model in the rat, induced by an antibody against the Thy1-antigen on MC ((Johnson (1994)
Kidney Int
. 45 (6), 1769-82). In this model, a single injection of anti-thymocyte antibody results in an acute, complement-dependent MC injury (days zero to two) with proteinuria, followed by a FGF-2- and PDGF-dependent MC proliferative response that is accompanied by a TGF-&bgr;-dependent overproduction of extracellular matrix proteins (days three to ten) (Johnson (1994)
Kidney Int
. 45 (6), 1769-82).
The role of TGF-&bgr; as a major profibrotic cytokine in the anti-Thy1 model has been well established (Border et al. (1994)
N. Engl. J. Med
. 331, 1286-1292). It has been demonstrated that TGF-&bgr;1 mRNA and protein are increased in the anti-Thy1 model (Okuda et al. (1990)
J. Clin. Invest
. 86, 453-462) and that blocking TGF-&bgr;1 by injections with a polyclonal anti-TGF-&bgr;1 antibody markedly reduced extracellular matrix accumulation (Border et al. (1990)
Nature
346, 371-374). Injections with the proteoglycan decorin, a TGF-&bgr;1, -2, and -3 binding protein, also suppressed TGF-&bgr;-dependent alterations such as extracellular matrix accumulation in the anti-Thy1 model (Border et al. (1992)
Nature
360, 361-364). The results of these studies by Border and colleagues were confirmed by studies using gene transfer techniques in the anti-Thy1 model. Transfer of antisense oligonucleotides against the TGF-&bgr;1 mRNA into the rat kidney suppressed upregulation of glomerular TGF-&bgr;1 mRNA and protein as well as extracellular matrix accumulation (Akaki et al. (1996)
Kidney Int
. 50, 148-155). Transfer of decorin cDNA into rat skeletal muscle increased the amount of decorin in skeletal muscle and in the kidney, and again ameliorated glomerular disease by decreasing matrix formation (Isaka et al. (1996)
Nat. Med
. 2, 418-423). In contrast, mice transgenic for an active form of TGF-&bgr;1 exhibit elevated plasma levels of TGF-&bgr;1 and develop progressive renal disease characterized by MC matrix accumulation, interstitial fibrosis, and proteinuria (Kopp et al. (1996)
Lab. Invest
. 74, 991-1003). Transfer of the TGF-&bgr;1 gene into glomeruli of normal rats caused an increase in glomerular TGF-&bgr;1 protein that was linked to extracellular matrix formation (Isaka et al (1993)
J. Clin. Invest
. 92, 2597-2601). The potential importance of TGF-&bgr; in mediating fibrosis also in human kidney disease has been supported by the widespread link of TGF-&bgr; upregulation and extracellular matrix excess in many different types of human kidney disease (Border et al. (1994)
N. Engl. J. Med
. 331, 1286-1292).
While these studies suggest great benefit from suppression of TGF-&bgr; function in fibrotic kidney disease, it has to be considered that TGF-&bgr; is a multifunctional cytokine that exhibits other essential functions in mammals. Mice lacking either the TGF-&bgr;1, or -2, or -3 gene do not survive beyond a few weeks after birth (Shull et al. (1992)
Nature
359, 693-699; Sanford et al. (1997)
Development
124, 2659-2670; Kaartinen et al. (1995)
Nature Genet
. 11, 415-421). TGF-&bgr;1 null mice die a few weeks after birth from a severe generalized inflammatory response demonstrating that complete suppression of TGF-&bgr; function must not be a therapeutic goal in treating inflammatory kidney disease (Shull et al. (1992)
Nature
359, 693-699). Therefore, accurate regulation of TGF-&bgr; function seems to be critical for the health of mammals and any anti-TGF-&bgr; therapeutic approach should try to target the local overproduction (-function) of TGF-&bgr; as specifically as possible.
One possibility to approach this goal could be by controlling (interfering with) the activation process of locally produced TGF-&bgr;. TGF-&bgr; is secreted by most cell types as a latent, inactive procytokine-complex that consists of the mature, active TGF-&bgr; protein, which is noncovalently bound to a dimer of its N-terminal propeptide, the so-called latency-associated protein (LAP), and variably to a latent TGF-&bgr; binding protein (LTBP) (Harpel et al. (1992)
Prog. Growth Factor Res
. 4, 321-335). The mature TGF-&bgr; protein has to be extracellularly released or unmasked from this procytokine-complex to be able to interact with its receptors. While various players/mechanisms such as pH changes, gamma irradiation, reactive oxygen species, plasmin, calpain, cathepsin, or thrombospondin 1 (TSP1) have been identified to activate TGF-&bgr; under in vitro conditions, it is still unknown how TGF-&bgr; is activated in an inflammatory process in vivo (Harpel et al. (1992)
Prog. Growth Factor Res
. 4, 321-335).
Recent data have suggested the homotrimeric extracellular matrix protein TSP1 as an activator of TGF-&bgr;1 in vitro in different cell systems including MC (Schultz-Cherry et al. (1993)
J. Cell Biol
. 122, 923-932; Tada et al. (1998)
Nephron
79, 38-43; Schultz-Cherry et al. (1994)
J. Biol. Chem
. 269, 26775-82) as well as in cell free systems. It has been demonstrated that TSP1 forms a trimolecular complex with the TGF-&bgr; procytokine-complex by interacting with the mature TGF-&bgr; protein as well as the LAP (Ribeiro et al. (1999)
J. Biol. Chem
. 274, 13586-13593). Hereby, the hexapeptide (AA or GG) WSHW (SEQ ID NO:22 or SEQ ID NO:3, respectively) from the type I repeats of the TSP1 molecule is required for TSP1-binding to the mature TGF-&bgr; protein facilitating interaction of the KRFK-amino acid sequence (SEQ. ID NO:5) of the TSP1 molecule with the N-terminal LSKL-sequence (SEQ ID NO:21) of the LAP (Schultz-Cherry et al. (1995)
J. Biol. Chem
. 270, 7304-7310; Ribeiro et al. (1999)
J. Biol. Chem
. 274, 13586-13593). This complex interaction is thought to induce a conformational change probably within the LAP that allows the mature TGF-&bgr; protein to bind to its receptors. It has been shown that both the hexapeptide AAWSHW (SEQ. ID NO:22) and the LSKL (SEQ. ID NO:21) peptides are able to block activation of TGF-&bgr; by TSP1. In addition, comparing TSP1 null mice with TGF-&bgr;1 null mice, Crawford et al. identified TSP1 as a major activator of TGF-&bgr;1 in vivo during mouse post-natal development (Crawford et al. (1998)
Cell
93, 1159-1170). Organ pathology of TGF-&bgr;1 null pups and TSP1 null pups were strikingly similar and could be induced in wild type pups by intraperitoneal (i.p.) treatment with the LSKL-peptide that specifically blocks activation of TGF-&bgr;1 by TSP1. Loss of TSP1 expression in TSP1 null mice spontaneously produced inflammatory lung disease (Lawler et al. (1998)
J. Clin. Invest
. 101, 982-992) and histological changes in TSP1 null mice reverted toward wild type by treatment with the TGF-&bgr; activating peptide KRFK (SEQ ID NO:5).
Interestingly, TSP1 expression in vitro is regulated by various cytokines such as PDGF, FGF-2, or TGF&bgr;, and is frequently expressed de novo at sites of inflammation and wound h

LandOfFree

Say what you really think

Search LandOfFree.com for the USA inventors and patents. Rate them and share your experience with other people.

Rating

Use of peptides inhibitory for thrombospondin dependent... does not yet have a rating. At this time, there are no reviews or comments for this patent.

If you have personal experience with Use of peptides inhibitory for thrombospondin dependent..., we encourage you to share that experience with our LandOfFree.com community. Your opinion is very important and Use of peptides inhibitory for thrombospondin dependent... will most certainly appreciate the feedback.

Rate now

     

Profile ID: LFUS-PAI-O-2941351

  Search
All data on this website is collected from public sources. Our data reflects the most accurate information available at the time of publication.