Detection of GPI anchored proteins

Chemistry: molecular biology and microbiology – Measuring or testing process involving enzymes or... – Involving antigen-antibody binding – specific binding protein...

Reexamination Certificate

Rate now

  [ 0.00 ] – not rated yet Voters 0   Comments 0

Details

C435S004000, C435S007700, C435S007900, C435S040520, C435S069100, C435S252300, C435S254110, C435S320100, C435S325000, C530S350000, C536S023100, C536S023500, C436S117000, C436S503000, C436S906000

Reexamination Certificate

active

06593095

ABSTRACT:

FIELD OF THE INVENTION
The invention relates to the discovery that certain biological toxins specifically bind to the glycosylphosphatidylinositol (GPI) anchor component of certain cell-surface proteins. Applications of this discovery include the detection and diagnosis of paroxysmal nocturnal hemoglobinuria.
BACKGROUND AND SUMMARY OF THE INVENTION
Aerolysin is a channel-forming cytolytic protein produced by virulent Aeromonas species, such as
Aeromonas hydrophila
. Aerolysin is one of the best studied of all of the bacterial cytolytic toxins. It is known to be secreted as a 52 kDa precursor called proaerolysin; this precursor form is converted to the active form by proteolytic removal of a C-terminal peptide. Many eucaryotic proteases can activate proaerolysin, as can proteases secreted by
A. hydrophila
itself. Once bound to a susceptible cell, aerolysin is transformed into an insertion-competent state by oligomerization. The oligomers, which are heptameric, bridge the lipid bilayer, producing discrete 1 nm channels which result in cell lysis.
It was previously believed that aerolysin bound specifically to certain proteins found on the cell surface, such as the Thy-1 antigen (see U.S. Pat. No. 5,798,218). The present invention, however, is founded on the discovery that the target for aerolysin binding is not any particular cell surface protein, but is actually the glycosylphosphatidylinositol (GPI) anchor that is a component of many cell surface proteins. As their name suggests, GPI anchors function to anchor proteins into the cell membrane; typically the GPI anchor component is at least partly embedded in the membrane, permitting the extracellular component of the protein to be presented to the surrounding environment.
Since GPI anchors are components of many important cell surface proteins, this discovery permits the detection of such proteins (or the determination of their absence) through the use of specific binding assays. For example, since aerolysin typically lyses cells following binding to the GPI anchored protein, one such assay is based on the differential rates of lysis observed when aerolysin is mixed with cells that either have, or do not have GPI anchored surface proteins. While such assays may be performed using aerolysin, other toxins that are related to aerolysin may also be employed for such methods. Such toxins include
Clostridium septicum
alpha toxin (Parker et al., 1996), and enterlobin (a cytolytic protein produced by the Brazilian tree Enterolobium, Sousa et al., 1994). The binding specificity of these toxins and the use of this specificity to detect GPI anchored proteins is of particular clinical relevance for the disease paroxysmal nocturnal hemoglobinuria (PNH).
PNH is an acquired hematopoietic stem cell disorder manifested by abnormal hematopoiesis, complement-mediated intravascular hemolysis and a propensity toward thrombosis (Rosse, 1997). The disease usually results from a somatic mutation in the X-linked gene, PIGA (Miyata et al, 1993; Takeda et al., 1993; Miyata et al., 1994;
Bessler et al., 1994) The product of the PIGA gene is necessary for the first step in the biosynthesis of GPI anchors. Hence, cells harboring PIGA mutations are characterized by a deficiency, absolute or partial, of all proteins affixed to the cell membrane by GPI anchors. The cells affected by this mutation include erythrocytes, granulocytes, monocytes and lymphocytes. PNH is closely associated with a range of hematological disorders, including aplastic anemia, certain leukemias and myelodysplasic syndrome, and assays for the disease are routinely performed for patients manifesting hematological disorders.
GPI anchored proteins have been shown to be involved in a wide range of important cell functions, including signal transduction (Robinson, 1991; Stefanova et al., 1991 ), and trafficking of apically expressed epithelial proteins (Brown et al., 1989; Powell et al., 1991). They may also play a role in regulating apoptosis (Brodsky, 1997). Two GPI anchored proteins, CD55 (decay accelerating factor) and CD59 (membrane inhibitor of reactive lysis), normally protect cells from the action of homologous complement, and it is their absence that leads to the hemolytic anemia associated with PNH (Rosse, 1982; Rosse, 1995).
The initial mutation of the PIGA gene occurs in a pluripotent hematopoietic stem cell. This cell subsequently divides and gives rise to multiple hematopoietic lineages. These various lineages generate lymphocytes, such as B cells and T cells, leukocytes and erythrocytes, all of which may be GPI anchor deficient (Rosse, 1997). Therefore, in a blood sample from a PNH patient there will be both GPI anchor deficient lymphocytes (those derived from the affected stem cell), as well as normal lymphocytes (those derived from an unaffected stem cell). Furthermore, cells derived from the affected stem cell generally fall into one of three categories. These categories are defined by the cells sensitivity to complement (I=normally sensitive cells, II=cells of intermediate sensitivity, and III=very sensitive cells).
Another characteristic of PNH is a decrease in erythrocyte survival (Rosse, 1971), and a normal or even increased survival of granulocytes (Brodsky, 1997: Horikawa et al., 1997; Brubaker et al., 1977). This decrease in erythrocyte survival poses a problem to clinicians because the two most popular methods of diagnosing PNH, the Ham's test and the sucrose hemolysis test, involve isolating erythrocytes and measuring their sensitivity to homologous complement. Because of decreased erythrocyte survival in PNH patients, the results from these assays do not provide accurate information about the percentage of affected blood cells. Furthermore, these assays are relatively insensitive and may not detect small populations of PNH cells, such as may be present at the early stages of the disease.
In addition to the Ham's test and the sucrose hemolysis test, flow cytometry is sometimes used to diagnose PNH. Flow cytometry offers the clinician the ability to use monoclonal antibodies to a variety of different GPI anchored proteins. These antibodies can be used to detect GPI anchored proteins on a variety of different cell types. For example, monoclonal antibodies to CD59 can be used to detect GPI anchors on granulocytes and other cells, thus providing a more accurate assessment of the number of PNH affected cells. Unfortunately, however, flow cytometry requires expensive equipment and significant technical expertise that is not available in many laboratories. Additionally, while flow cytometry can be significantly more sensitive than the sucrose hemolysis and Ham's tests, it cannot be routinely used to detect PNH cells in populations of less than 1-2% of total cells (Schubert et al., 1991; Hall et al., 1996). Therefore, there is a need for an assay that is inexpensive, accurate and specific for the detection of small populations of PNH affected cells, and the present invention provides such an assay.
The invention provides a number of ways of detecting the presence of PNH cells in biological samples. Generally, these methods comprise contacting a biological sample containing blood cells with a toxin that specifically binds to GPI anchored proteins, and monitoring binding of the toxin to the blood cells. In view of the specificity of the toxin, decreased binding of the toxin to the blood cells compared to binding observed with a control blood sample indicates decreased GPI anchored proteins, and thus the presence of PNH cells.
The biological sample will typically be taken from an individual who is to be screened for PNH, and may be, for example, whole blood, granulocytes or erythrocytes. The toxin may be aerolysin, proaerolysin, Clostridium alpha toxin, enterolobin, or any other toxin that specifically binds to GPI anchored proteins. While the toxin will generally be in its naturally occurring form, forms of the toxin having an altered amino acid sequence may also be employed. For example, forms of aerolysin that bind to GPI anchored p

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

Detection of GPI anchored proteins does not yet have a rating. At this time, there are no reviews or comments for this patent.

If you have personal experience with Detection of GPI anchored proteins, we encourage you to share that experience with our LandOfFree.com community. Your opinion is very important and Detection of GPI anchored proteins will most certainly appreciate the feedback.

Rate now

     

Profile ID: LFUS-PAI-O-3004786

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