Tumor growth requires proteolytic activity. As a consequence, protein breakdown products are present in the circulation of patients with cancer. Within the past decade a large number of proteolytic fragments have been identified that inhibit angiogenesis and tumor growth. The mechanism of action of these inhibitors is still poorly understood. We recently found that the effects of the angiogenesis inhibitor endostatin on endothelial cells is critically dependent on the presence of cross-β structure, a structure also present in amyloidogenic polypeptides in plaques of patients with amyloidosis, such as Alzheimer disease. We also showed that cross-β structure containing endostatin is a ligand for tissue-type plasminogen activator (tPA). We noted that many angiogenesis inhibitors stimulate tPA-mediated plasminogen activation. Because the presence of cross-β structure is the common denominator in tPA-binding ligands, we hypothesize that these endogenous antiangiogenic proteolytic fragments share features with amyloidogenic polypeptides. We postulate that the cross-β structural fold is present in these antiangiogenic polypeptide fragments and that this structure mediates the inhibitory effects. The hypothesis provides new insights in the potential mechanisms of these angiogenesis inhibitors and offers opportunities to improve their use. (Blood. 2004;104:1601-1605)

Angiogenesis, the formation of new blood vessels from preexisting vasculature, occurs in physiologic and pathologic processes, including embryonic development, the menstrual cycle, wound healing, inflammation, and tumor growth (for a review, see Carmeliet and Jain1 ). In 1971, Folkman2  postulated that tumor growth is dependent on angiogenesis. Accumulating evidence indicates that drugs that target the growing vasculature are promising new therapeutics for the treatment of cancer. Based on the concept that a primary tumor produces inhibitors of angiogenesis that can inhibit the outgrowth of metastases, the first endogenous angiogenesis inhibitor, termed angiostatin, was purified in the laboratory of Folkman in 1994.3  The identification of angiostatin prompted the search for other angiogenesis inhibitors, resulting in the isolation of endostatin,4  prothrombin fragment 1 and 2,5  cleaved antithrombin III,6  fibrin(ogen) fragments (Brown et al7  and A.R., unpublished observations, September 1999) and many other inhibitory polypeptide fragments. Commonly, such antiangiogenic polypeptides are proteolytically cleaved or denatured derivatives of endogenous proteins. Endostatin, angiostatin, and a thrombospondin peptide are currently being tested in clinical trials.8-10 

In 1997, Folkman and colleagues reported that treatment of experimental tumors with endostatin induced complete regression.11  These promising results from animal studies fueled the great expectations that accompanied the proposed use of antiangiogenic therapy with endogenous inhibitors of angiogenesis. However, the initial results could not be reproduced by others and lessened the initial enthusiasm.12  Controversies regarding the antitumor effects of endostatin arose and initiated a heated discussion about its efficacy.12  The variability in endostatin bioactivity is still unexplained. Originally, endostatin was purified from tissue culture supernatant. In the in vivo experiments that showed dramatic tumor regression a recombinant denatured and insoluble form of endostatin produced in Escherichia coli was used.11  In a number of subsequent studies endostatin has been used from a number of different sources. These different endostatin preparations caused less dramatic, minor, or even no effects.13-16  For studies in patients, a soluble globular form is used, produced in Pichia pastoris.8  However, this globular form has no or very little effect on endothelial cells.17  We found that only denatured endostatin is toxic to cells.18  We wondered whether our findings might explain the observed variability in endostatin bioactivity and to what extent our observations could be extrapolated to other angiogenesis inhibitors.

Similar to certain antiangiogenic polypeptides, toxic polypeptides involved in conformational diseases such as Alzheimer disease (AD), light-chain amyloidosis, pancreatic islet amyloidosis, and spongiform encephalopathies are proteolytic fragments or conformationally changed forms of generally harmless proteins. Properly folded proteins form a stable 3-dimensional structure. However, protein fragments (such as endostatin) are often prone to (partial) denaturation and undergo sequential aggregation steps leading to the formation of highly ordered aggregates. Initially, (partly) unfolded proteins or protein fragments associate with each other and form small soluble aggregates, preceding the formation of protofibrils. Ultimately, mature fibrils accumulate and are deposited as plaques. Fibrillar aggregates are classified as amyloid fibrils based on the presence of cross-β structure. Cross-β structures in amyloid fibrils are stacked β sheets composed of flat and nontwisted β strands. This will result in a unique and flat 2-dimensional β-sheet surface, not seen in globular proteins (Figure 1). The list of amyloidogenic peptides arising from proteolytic processing of harmless proteins is long and still increasing (for a review, see Selkoe19 ). In addition to disease-associated amyloid formation, intriguing physiologic roles for amyloid fibril formation have been elucidated.20,21  These recent reports highlight such a behavior of specific proteins or protein fragments in microorganisms as well as in eukaryotes. It is expected that numerous other proteins will be found to form amyloid as a natural product.

Figure 1.

Illustration of amyloid formation and cross-β structure. (A) Initiation of amyloid fibrillization by a partially structured conformer. An unstable amyloidogenic intermediate is formed via partial unfolding of a compact globular protein (i) or via the gain of structure by a natively unfolded polypeptide (ii). The partially ordered intermediate, which contains some β structure (depicted as a green block arrow), is stabilized by ordered self-assembly (iii) to form a nucleus enriched with β sheet. Alternatively, self-association may lead to the formation of large amorphous aggregates (iv). The amorphous aggregates may facilitate nucleation by providing a high local concentration of amyloidogenic intermediates (v). Growth of the nucleus by β-sheet extension leads to the formation of higher order oligomers (vi) and fibrils (vii). The fibril may consist of antiparallel, as well as parallel, β sheet. For details, see Rochet and Lansbury.61  (B) Amyloid cross-β structure. In amyloid fibrils, cross-β structures are stacked β sheets likely composed of flat and nontwisted β strands. This will result in a unique and flat 2-dimensional β-sheet surface, not seen in globular proteins. The inter-β strand distance of 4.7 Å that is measured using x-ray fiber diffraction is an import characteristic not seen in globular proteins.

Figure 1.

Illustration of amyloid formation and cross-β structure. (A) Initiation of amyloid fibrillization by a partially structured conformer. An unstable amyloidogenic intermediate is formed via partial unfolding of a compact globular protein (i) or via the gain of structure by a natively unfolded polypeptide (ii). The partially ordered intermediate, which contains some β structure (depicted as a green block arrow), is stabilized by ordered self-assembly (iii) to form a nucleus enriched with β sheet. Alternatively, self-association may lead to the formation of large amorphous aggregates (iv). The amorphous aggregates may facilitate nucleation by providing a high local concentration of amyloidogenic intermediates (v). Growth of the nucleus by β-sheet extension leads to the formation of higher order oligomers (vi) and fibrils (vii). The fibril may consist of antiparallel, as well as parallel, β sheet. For details, see Rochet and Lansbury.61  (B) Amyloid cross-β structure. In amyloid fibrils, cross-β structures are stacked β sheets likely composed of flat and nontwisted β strands. This will result in a unique and flat 2-dimensional β-sheet surface, not seen in globular proteins. The inter-β strand distance of 4.7 Å that is measured using x-ray fiber diffraction is an import characteristic not seen in globular proteins.

Close modal

The formation of amyloid aggregates and their relation to disease are studied intensively (for reviews, see Selkoe19  and Dobson22 ). Just recently a common, although unknown, mechanism of cell toxicity has been implied for protein misfolding diseases. Toxicity is an inherent property of aggregates of denatured proteins and is not related to the amino acid sequence composition of the protein.23  Although the nature of the toxicity remains unclear, recent studies suggest that the toxicity of amyloid proteins is enclosed in soluble oligomers, rather than in fibrils.24-26  This notion is supported by the finding that antibodies that detect early amyloidogenic aggregates can inhibit their toxicity.27  Many studies have indicated a close relationship between the presence of amyloid depositions and vascular damage. Transmissible spongiform disease is associated with the presence of the abnormal form of prion protein that aggregates into amyloid fibrils. Prion deposits are toxic to a wide variety of cells, including cerebral endothelial cells.28  Damage to endothelial cells is also seen in AD. Patients with AD exhibit significant cerebrovascular pathology in addition to neuronal degeneration.29,30  Furthermore, it is known that patients with AD suffer from stroke and that hemorrhages are present.31  In vitro experiments revealed that amyloid β increases permeability of endothelial cell monolayers and induces apoptosis.32,33  Other studies have shown that smooth muscle cells and endothelial cells are damaged in cerebral blood vessels of those with AD.33-35  Finally, the diabetes-related amyloidogenic peptide amylin and the circulating amyloidogenic peptide β2-microglobulin damage blood vessels.36  We recently showed that insoluble endostatin forms amyloid fibrils, suggesting that endostatin may induce apoptosis as a consequence of its amyloid structure.18,37  In AD endostatin colocalized with amyloid β-positive plaques that were surrounded by focal gliosis.38  These studies indicate that the cross-β structure has direct effects on endothelial cells. Is the cross-β structure then a common denominator in polypeptide fragments with antiangiogenic activity?

In general, denatured proteins can stimulate plasminogen activation by tissue-type plasminogen activator (tPA).39  Amyloid fibrils such as amyloid β and prion protein markedly stimulate plasminogen activation by tPA.40, 41  We found that the presence of cross-β structure is a prerequisite for tPA binding and for activation of plasminogen.37  We noticed that many antiangiogenic compounds, generated through proteolytic cleavage or denaturation, stimulate tPA-mediated plasminogen activation (Table 1).

Table 1.

Overview of antitumorigenic proteins with tPA-stimulating activity


Protein

Effect on angiogenesis or tumor growth

Stimulation of tPA-mediated plasminogen activation
Endostatin   O'Reilly et al, 19974   Reijerkerk et al, 200317  
Thrombospondin*  Volpert et al, 199862   Silverstein et al, 1984, 1985, 198663-65  
Angiostatin  O'Reilly et al, 19943   Unknown  
Denatured antithrombin III   O'Reilly et al, 19996   Machovich and Owen, 199739  
Prothrombin fragments   Rhim et al, 19985   Machovic et al, 199966  
βpep25 (Anginex)   Griffioen et al, 200167   A.R., unpublished  
Maspin   Zou et al, 199468   Sheng et al, 199869  
Histidine-proline-rich glycoprotein   Juarez et al, 200270   Silverstein et al, 198563 ; Borza and Morgan, 199771  
Fibrin(ogen) degradation products   AR unpublished and Brown et al, 20027   Stewart et al, 199872  
Calreticulin (fragments), vasostatin   Pike et al, 199973   Binds tPA. Allen and Bulleid, 199774  
Amphoterin
 
Huttunen et al, 200255 
 
Parkinen and Rauvala, 199175 
 

Protein

Effect on angiogenesis or tumor growth

Stimulation of tPA-mediated plasminogen activation
Endostatin   O'Reilly et al, 19974   Reijerkerk et al, 200317  
Thrombospondin*  Volpert et al, 199862   Silverstein et al, 1984, 1985, 198663-65  
Angiostatin  O'Reilly et al, 19943   Unknown  
Denatured antithrombin III   O'Reilly et al, 19996   Machovich and Owen, 199739  
Prothrombin fragments   Rhim et al, 19985   Machovic et al, 199966  
βpep25 (Anginex)   Griffioen et al, 200167   A.R., unpublished  
Maspin   Zou et al, 199468   Sheng et al, 199869  
Histidine-proline-rich glycoprotein   Juarez et al, 200270   Silverstein et al, 198563 ; Borza and Morgan, 199771  
Fibrin(ogen) degradation products   AR unpublished and Brown et al, 20027   Stewart et al, 199872  
Calreticulin (fragments), vasostatin   Pike et al, 199973   Binds tPA. Allen and Bulleid, 199774  
Amphoterin
 
Huttunen et al, 200255 
 
Parkinen and Rauvala, 199175 
 
*

Thrombospondin also binds the multiligand receptor CD36.56,76 

Angiostatin that is tested in clinical trials is made in P pastoris. The original experiments, however, were performed with angiostatin prepared by proteolytic cleavage with elastase, followed by purification on a lysine-Sepharose column and subsequent dialysis against H2O.3  The latter method of purification leaves the possibility that fragments are present with amyloid properties. We, therefore, suggest that different forms of angiostatin may also yield differences in bioactivity.

Amphoterin also binds the multiligand receptor RAGE.55 

It is intriguing that unrelated proteins lacking sequence homology acquire antiangiogenic activity on proteolytic cleavage or denaturation and at the same time acquire the ability to stimulate tPA-mediated plasminogen activation. Toxicity of aggregates of denatured proteins is also independent of the amino acid sequence.23,27  Taken together, this suggests that a common antiangiogenic pathway may exist that is induced by tPA-binding proteins. We hypothesize that antiangiogenic polypeptide fragments contain the cross-β structure and that this structure is the common denominator responsible for their antiangiogenic and antitumor effects. If correct, the presence of cross-β structure in antiangiogenic proteins could represent yet another example of a physiologic function of protein aggregation as has been published recently for microbial and eukaryotic proteins (for a review, see Huff et al20 ).

Can tPA contribute to the antiangiogenic effects of these compounds?41  tPA is historically known for its role in blood clot lysis (fibrinolysis). tPA cleaves the zymogen plasminogen into the active serine proteinase plasmin. Pericellular plasmin subsequently degrades a fibrin clot. Apart from their well-established role in clot lysis, tPA and plasmin also function in other processes, including long-term potentiation42  and neuronal cell migration.43  tPA has also been identified as a mediator of neuronal cell death following ischemia or excitotoxic injury in the brain.44  Intracerebral injection of excitotoxins such as glutamate and focal cerebral ischemia provoke degradation of the extracellular matrix protein laminin and cause neuronal death.45-47  We found that tPA activation by amyloid endostatin induced plasmin formation, degradation of the extracellular matrix protein vitronectin, and cell detachment.17  Matrix proteins including vitronectin, fibronectin, and laminin play an important role in cell survival48  and their breakdown can result in apoptosis. Induction of plasminogen activation leads to endothelial cell detachment,49  inhibition of cell adhesion,50  endothelial cell destruction,51  or regression of capillary tubes.52  Most recently, Angles-Cano and coworkers showed that plasminogen activation induces detachment and apoptosis of Chinese hamster ovary (CHO) fibroblasts53  and smooth muscle cells.54  tPA is constitutively expressed by CHO fibroblasts and smooth muscle cells and appears to be responsible for pericellular plasminogen activation.53,54  Finally, a possible role for tPA and plasmin in amyloid-mediated toxicity is further supported by the finding that serpins (a family of protease inhibitors) can inhibit amyloid toxicity. Taken together, activation of tPA by amyloid polypeptides may induce excessive degradation of extracellular matrix with subsequent loss of cell attachment sites resulting in cell detachment and apoptosis.

If our hypothesis is correct, other cross-β structure receptors in addition to tPA may mediate the antiangiogenic response. Good evidence indicates that at least 2 other receptors that bind amyloid polypeptides, CD36 and the receptor for advanced glycation end products (RAGE), could be involved.55,56  Many potential mechanisms for amyloid-induced cell toxicity have already been proposed. It is well established that amyloid-induced neuronal cell toxicity involves the production of reactive oxygen species and disruption of calcium homeostasis. Endothelial cell toxicity caused by Aβ is also associated with the production of superoxide free radicals, causing oxidative lipid and protein damage, and increased intracellular calcium concentration.32,57,58 

Implications

If correct, our hypothesis implies that (1) it is relevant to look at structural properties of angiogenesis inhibitors, rather than at their primary amino acid sequence and to consider the possibility that an active drug requires a cross-β structure; (2) the inhibitory effect of the drugs listed in Table 1 may be improved by adding factors that are known to increase amyloid-mediated toxicity, such as lipopolysaccharide (LPS), tumor necrosis factor α (TNF-α), and interferon γ (IFN-γ)59,60 ; and (3) it is necessary to use these polypeptide fragments with caution because they may form toxic aggregates.

Moreover, tumor-derived endogenous circulating inhibitors may also possess cross-β structure. Besides their antiangiogenic activity, these circulating proteolytic fragments may, however, then also contribute to some of the clinical complications of cancer, such as amyloidosis or bleeding events.

Note added in proof. After this article was accepted for publication, we became aware of new results obtained by Paris et al.77  The authors showed that amyloid β peptides inhibit angiogenesis. Their results add amyloid β peptides to the list of angiogenesis inhibitors that interact with tPA, and support our hypothesis that antiangiogenic fragments in general may have amyloid properties.

Prepublished online as Blood First Edition Paper, May 27, 2004; DOI 10.1182/blood-2004-02-0433.

Supported by the Dutch Cancer Society (UU1999-2114).

1
Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases.
Nature
.
2000
;
407
:
249
-257.
2
Folkman J. Tumor angiogenesis: therapeutic implications.
N Engl J Med
.
1971
;
285
:
1182
-1186.
3
O'Reilly MS, Holmgren L, Shing Y, et al. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma.
Cell
.
1994
;
79
:
315
-328.
4
O'Reilly MS, Boehm T, Shing Y, et al. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth.
Cell
.
1997
;
88
:
277
-285.
5
Rhim TY, Park CS, Kim E, Kim SS. Human prothrombin fragment 1 and 2 inhibit bFGF-induced BCE cell growth.
Biochem Biophys Res Commun
.
1998
;
252
:
513
-516.
6
O'Reilly MS, Pirie-Shepherd S, Lane WS, Folkman J. Antiangiogenic activity of the cleaved conformation of the serpin antithrombin.
Science
.
1999
;
285
:
1926
-1928.
7
Brown NJ, Staton CA, Rodgers GR, et al. Fibrinogen E fragment selectively disrupts the vasculature and inhibits the growth of tumours in a syngeneic murine model.
Br J Cancer
.
2002
;
86
:
1813
-1816.
8
Herbst RS, Hess KR, Tran HT, et al. Phase I study of recombinant human endostatin in patients with advanced solid tumors.
J Clin Oncol
.
2002
;
20
:
3792
-3803.
9
Beerepoot LV, Witteveen PO, Groenewegen G, et al. Recombinant human angiostatin by twice-daily subcutaneous injection in advanced cancer: a pharmacokinetic and long-term safety study.
Clin Cancer Res
.
2003
;
9
:
4025
-4033.
10
Gordon MS, Mendelson D, Guirguis MS, et al. ABT-510, an antiangiogenic, thrombospondin-1 (TSP-1) mimetic peptide, exhibits favorable safety profile and early signals of activity in a randomized phase IB trial [abstract].
Proceedings of the Thirty-ninth Annual Meeting of the American Society of Clinical Oncology, May 31-June 3, 2003
; Chicago, IL.
2003
:
195
. Abstract 780.
11
Boehm T, Folkman J, Browder T, O'Reilly MS. Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance.
Nature
.
1997
;
390
:
404
-407.
12
Marshall E. Cancer therapy. Setbacks for endostatin.
Science
.
2002
;
295
:
2198
-2199.
13
Steele FR. Can “negative” be positive?
Mol Ther
.
2002
;
5
:
338
-339.
14
Eisterer W, Jiang X, Bachelot T, et al. Unfulfilled promise of endostatin in a gene therapy-xenotransplant model of human acute lymphocytic leukemia.
Mol Ther
.
2002
;
5
:
352
-359.
15
Pawliuk R, Bachelot T, Zurkiya O, et al. Continuous intravascular secretion of endostatin in mice from transduced hematopoietic stem cells.
Mol Ther
.
2002
;
5
:
345
-351.
16
Jouanneau E, Alberti L, Nejjari M, et al. Lack of antitumor activity of recombinant endostatin in a human neuroblastoma xenograft model.
J Neurooncol
.
2001
;
51
:
11
-18.
17
Reijerkerk A, Mosnier LO, Kranenburg O, et al. Amyloid endostatin induces endothelial cell detachment by stimulation of the plasminogen activation system.
Mol Cancer Res
.
2003
;
1
:
561
-568.
18
Kranenburg O, Kroon-Batenburg LM, Reijerkerk A, et al. Recombinant endostatin forms amyloid fibrils that bind and are cytotoxic to murine neuroblastoma cells in vitro.
FEBS Lett
.
2003
;
539
:
149
-155.
19
Selkoe DJ. Folding proteins in fatal ways.
Nature
.
2003
;
426
:
900
-904.
20
Huff ME, Balch WE, Kelly JW. Pathological and functional amyloid formation orchestrated by the secretory pathway.
Curr Opin Struct Biol
.
2003
;
13
:
674
-682.
21
Kelly JW, Balch WE. Amyloid as a natural product.
J Cell Biol
.
2003
;
161
:
461
-462.
22
Dobson CM. Protein folding and misfolding.
Nature
.
2003
;
426
:
884
-890.
23
Bucciantini M, Giannoni E, Chiti F, et al. Inherent toxicity of aggregates implies a common mechanism for protein misfolding diseases.
Nature
.
2002
;
416
:
507
-511.
24
Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics.
Science
.
2002
;
297
:
353
-356.
25
Kirkitadze MD, Bitan G, Teplow DB. Paradigm shifts in Alzheimer's disease and other neurode-generative disorders: the emerging role of oligomeric assemblies.
J Neurosci Res
.
2002
;
69
:
567
-577.
26
Walsh DM, Klyubin I, Fadeeva JV, et al. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo.
Nature
.
2002
;
416
:
535
-539.
27
Kayed R, Head E, Thompson JL, et al. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis.
Science
.
2003
;
300
:
486
-489.
28
Deli MA, Sakaguchi S, Nakaoke R, et al. PrP fragment 106-126 is toxic to cerebral endothelial cells expressing PrP(C).
Neuroreport
.
2000
;
11
:
3931
-3936.
29
Vinters HV, Wang ZZ, Secor DL. Brain parenchymal and microvascular amyloid in Alzheimer's disease.
Brain Pathol
.
1996
;
6
:
179
-195.
30
Moody DM, Brown WR, Challa VR, et al. Cerebral microvascular alterations in aging, leukoaraiosis, and Alzheimer's disease.
Ann N Y Acad Sci
.
1997
;
826
:
103
-16.:103-116.
31
Mandybur TI, Bates SR. Fatal massive intracerebral hemorrhage complicating cerebral amyloid angiopathy.
Arch Neurol
.
1978
;
35
:
246
-248.
32
Blanc EM, Toborek M, Mark RJ, et al. Amyloid beta-peptide induces cell monolayer albumin permeability, impairs glucose transport, and induces apoptosis in vascular endothelial cells.
J Neurochem
.
1997
;
68
:
1870
-1881.
33
Kawai M, Kalaria RN, Cras P, et al. Degeneration of vascular muscle cells in cerebral amyloid angiopathy of Alzheimer disease.
Brain Res
.
1993
;
623
:
142
-146.
34
Kalaria RN, Grahovac I. Serum amyloid P immunoreactivity in hippocampal tangles, plaques and vessels: implications for leakage across the blood-brain barrier in Alzheimer's disease.
Brain Res
.
1990
;
516
:
349
-353.
35
Kalaria RN, Hedera P. Differential degeneration of the cerebral microvasculature in Alzheimer's disease.
Neuroreport
.
1995
;
6
:
477
-480.
36
Castillo MJ, Scheen AJ, Lefebvre PJ. Amylin/islet amyloid polypeptide: biochemistry, physiology, patho-physiology.
Diabetes Metab
.
1995
;
21
:
3
-25.
37
Kranenburg O, Bouma B, Kroon-Batenburg LM, et al. Tissue-type plasminogen activator is a multiligand cross-beta structure receptor.
Curr Biol
.
2002
;
12
:
1833
-1839.
38
Deininger MH, Fimmen BA, Thal DR, et al. Aberrant neuronal and paracellular deposition of endostatin in brains of patients with Alzheimer's disease.
J Neurosci
.
2002
;
22
:
10621
-10626.
39
Machovich R, Owen WG. Denatured proteins as cofactors for plasminogen activation.
Arch Biochem Biophys
.
1997
;
344
:
343
-349.
40
Kingston IB, Castro MJ, Anderson S. In vitro stimulation of tissue-type plasminogen activator by Alzheimer amyloid beta-peptide analogues.
Nat Med
.
1995
;
1
:
138
-142.
41
Ellis V, Daniels M, Misra R, Brown DR. Plasminogen activation is stimulated by prion protein and regulated in a copper-dependent manner.
Biochemistry
.
2002
;
41
:
6891
-6896.
42
Huang YY, Bach ME, Lipp HP, et al. Mice lacking the gene encoding tissue-type plasminogen activator show a selective interference with late-phase long-term potentiation in both Schaffer collateral and mossy fiber pathways.
Proc Natl Acad Sci U S A
.
1996
;
93
:
8699
-8704.
43
Seeds NW, Basham ME, Haffke SP. Neuronal migration is retarded in mice lacking the tissue plasminogen activator gene.
Proc Natl Acad Sci U S A
.
1999
;
96
:
14118
-14123.
44
Tsirka SE. Tissue plasminogen activator as a modulator of neuronal survival and function.
Biochem Soc Trans
.
2002
;
30
:
222
-225.
45
Chen ZL, Strickland S. Neuronal death in the hippocampus is promoted by plasmin-catalyzed degradation of laminin.
Cell
.
1997
;
91
:
917
-925.
46
Tsirka SE, Gualandris A, Amaral DG, Strickland S. Excitotoxin-induced neuronal degeneration and seizure are mediated by tissue plasminogen activator.
Nature
.
1995
;
377
:
340
-344.
47
Wang YF, Tsirka SE, Strickland S, et al. Tissue plasminogen activator (tPA) increases neuronal damage after focal cerebral ischemia in wild-type and tPA-deficient mice.
Nat Med
.
1998
;
4
:
228
-231.
48
Isik FF, Gibran NS, Jang YC, et al. Vitronectin decreases microvascular endothelial cell apoptosis.
J Cell Physiol
.
1998
;
175
:
149
-155.
49
Ge M, Tang G, Ryan TJ, Malik AB. Fibrinogen degradation product fragment D induces endothelial cell detachment by activation of cell-mediated fibrinolysis.
J Clin Invest
.
1992
;
90
:
2508
-2516.
50
Reinartz J, Schafer B, Batrla R, et al. Plasmin abrogates alpha v beta 5-mediated adhesion of a human keratinocyte cell line (HaCaT) to vitronectin.
Exp Cell Res
.
1995
;
220
:
274
-282.
51
Sugimura M, Kobayashi H, Terao T. Plasmin modulators, aprotinin and anti-catalytic plasmin antibody, efficiently inhibit destruction of bovine vascular endothelial cells by choriocarcinoma cells.
Gynecol Oncol
.
1994
;
52
:
337
-346.
52
Davis GE, Pintar Allen KA, Salazar R, Maxwell SA. Matrix metalloproteinase-1 and -9 activation by plasmin regulates a novel endothelial cell-mediated mechanism of collagen gel contraction and capillary tube regression in three-dimensional collagen matrices.
J Cell Sci
.
2001
;
114
:
917
-930.
53
Rossignol P, Ho-Tin-Noe B, Vranckx R, et al. Protease nexin-1 inhibits plasminogen activation-induced apoptosis of adherent cells.
J Biol Chem
.
2004
;
279
:
10346
-10356.
54
Meilhac O, Ho-Tin-Noe B, Houard X, et al. Pericellular plasmin induces smooth muscle cell anoikis.
FASEB J
.
2003
;
17
:
1301
-1303.
55
Huttunen HJ, Fages C, Kuja-Panula J, et al. Receptor for advanced glycation end products-binding COOH-terminal motif of amphoterin inhibits invasive migration and metastasis.
Cancer Res
.
2002
;
62
:
4805
-4811.
56
Dawson DW, Pearce SF, Zhong R, et al. CD36 mediates the In vitro inhibitory effects of thrombospondin-1 on endothelial cells.
J Cell Biol
.
1997
;
138
:
707
-717.
57
Suo Z, Fang C, Crawford F, Mullan M. Superoxide free radical and intracellular calcium mediate A beta(1-42) induced endothelial toxicity.
Brain Res
.
1997
;
762
:
144
-152.
58
Hase M, Araki S, Hayashi H. Fragments of amyloid beta induce apoptosis in vascular endothelial cells.
Endothelium
.
1997
;
5
:
221
-229.
59
Stepanichev MY, Zdobnova IM, Yakovlev AA, et al. Effects of tumor necrosis factor-alpha central administration on hippocampal damage in rat induced by amyloid beta-peptide (25-35).
J Neurosci Res
.
2003
;
71
:
110
-120.
60
Gasic-Milenkovic J, Dukic-Stefanovic S, Deuther-Conrad W, et al. beta-Amyloid peptide potentiates inflammatory responses induced by lipopolysaccharide, interferon-gamma and “advanced glycation endproducts” in a murine microglia cell line.
Eur J Neurosci
.
2003
;
17
:
813
-821.
61
Rochet JC, Lansbury PT Jr. Amyloid fibrillogenesis: themes and variations.
Curr Opin Struct Biol
.
2000
;
10
:
60
-68.
62
Volpert OV, Lawler J, Bouck NP. A human fibrosarcoma inhibits systemic angiogenesis and the growth of experimental metastases via thrombospondin-1.
Proc Natl Acad Sci U S A
.
1998
;
95
:
6343
-6348.
63
Silverstein RL, Nachman RL, Leung LL, Harpel PC. Activation of immobilized plasminogen by tissue activator. Multimolecular complex formation.
J Biol Chem
.
1985
;
260
:
10346
-10352.
64
Silverstein RL, Leung LL, Harpel PC, Nachman RL. Complex formation of platelet thrombospondin with plasminogen. Modulation of activation by tissue activator.
J Clin Invest
.
1984
;
74
:
1625
-1633.
65
Silverstein RL, Harpel PC, Nachman RL. Tissue plasminogen activator and urokinase enhance the binding of plasminogen to thrombospondin.
J Biol Chem
.
1986
;
261
:
9959
-9965.
66
Machovich R, Komorowicz E, Kolev K, Owen WG. Facilitation of plasminogen activation by denatured prothrombin.
Thromb Res
.
1999
;
94
:
389
-394.
67
Griffioen AW, van der Schaft DW, Barendsz-Janson AF, et al. Anginex, a designed peptide that inhibits angiogenesis.
Biochem J
.
2001
;
354
:
233
-242.
68
Zou Z, Anisowicz A, Hendrix MJ, et al. Maspin, a serpin with tumor-suppressing activity in human mammary epithelial cells.
Science
.
1994
;
263
:
526
-529.
69
Sheng S, Truong B, Fredrickson D, et al. Tissue-type plasminogen activator is a target of the tumor suppressor gene maspin.
Proc Natl Acad Sci U S A
.
1998
;
95
:
499
-504.
70
Juarez JC, Guan X, Shipulina NV, et al. Histidineproline-rich glycoprotein has potent antiangiogenic activity mediated through the histidine-proline-rich domain.
Cancer Res
.
2002
;
62
:
5344
-5350.
71
Borza DB, Morgan WT. Acceleration of plasminogen activation by tissue plasminogen activator on surface-bound histidine-proline-rich glycoprotein.
J Biol Chem
.
1997
;
272
:
5718
-5726.
72
Stewart RJ, Fredenburgh JC, Weitz JI. Characterization of the interactions of plasminogen and tissue and vampire bat plasminogen activators with fibrinogen, fibrin, and the complex of D-dimer noncovalently linked to fragment E.
J Biol Chem
.
1998
;
273
:
18292
-18299.
73
Pike SE, Yao L, Setsuda J, et al. Calreticulin and calreticulin fragments are endothelial cell inhibitors that suppress tumor growth.
Blood
.
1999
;
94
:
2461
-2468.
74
Allen S, Bulleid NJ. Calnexin and calreticulin bind to enzymically active tissue-type plasminogen activator during biosynthesis and are not required for folding to the native conformation.
Biochem J
.
1997
;
328
:
113
-119.
75
Parkkinen J, Rauvala H. Interactions of plasminogen and tissue plasminogen activator (t-PA) with amphoterin. Enhancement of t-PA-catalyzed plasminogen activation by amphoterin.
J Biol Chem
.
1991
;
266
:
16730
-16735.
76
Reiher FK, Volpert OV, Jimenez B, et al. Inhibition of tumor growth by systemic treatment with thrombospondin-1 peptide mimetics.
Int J Cancer
.
2002
;
98
:
682
-689.
77
Paris D, Townsend K, Quadros A, et al. Inhibition of angiogenesis by Aβ peptides.
Angiogenesis
. In press.
Sign in via your Institution