You are on page 1of 5

From www.bloodjournal.org at NATIONAL INSTITUTES OF HEALTH LIB on February 12, 2009.

For personal
use only.

1991 78: 547-550

Dr Ham's test revisited [editorial]


WF Rosse

Information about reproducing this article in parts or in its entirety may be found online at:
http://bloodjournal.hematologylibrary.org/misc/rights.dtl#repub_requests

Information about ordering reprints may be found online at:


http://bloodjournal.hematologylibrary.org/misc/rights.dtl#reprints

Information about subscriptions and ASH membership may be found online at:
http://bloodjournal.hematologylibrary.org/subscriptions/index.dtl

Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published


semimonthly by the American Society of Hematology, 1900 M St, NW,
Suite 200, Washington DC 20036.
Copyright 2007 by The American Society of Hematology; all rights
reserved .
From www.bloodjournal.org at NATIONAL INSTITUTES OF HEALTH LIB on February 12, 2009. For personal
use only.

The Jouml of
BLOOD The American SocieQ of Hematology

VOL 78, NO 3 AUGUST 1, 1991

EDITORIAL
Dr Ham’s Test Revisited
C3 to C3b, thus forming evermore convertase complexes on
I N 1937, Thomas Hale Ham made a remarkable observa-
tion: the cells of patients with paroxysmal nocturnal
hemoglobinuria (PNH) are hemolyzed when incubated
the membrane.
This process appears to be aided by two factors. Acidifi-
with acidified normal serum.’ This lysis appeared to be due cation in some way not entirely delineated increases the
to the activation of complement because procedures that activation of the pathway. It is not clear whether this is due
inactivated complement abolished the reaction.’ Because to optimization of reaction conditions for one of the
the red blood cells in no other disease appeared to share enzymes or for the fixation of C3b to membrane compo-
this peculiarity, this test became the sine qua non for the nents. The optimum pH is about 6.2, the pH recommended
diagnosis of the d i ~ e a s e . ~ in the performance of the acidified serum lysis test.
In fact, the phenomenon had been observed some years Furthermore, May et a1” showed that the hemolysis of
previously by Hijmans van den Bergh, who had acidified PNH erythrocytes in acidified serum could be increased by
serum with “carbonic acid” and had noted the “striking optimization of the concentration of Mg2’ to 0.015 mol/L If
result” that the cells of the patient with “ictere hemolytique this was not done, not all of the abnormal cells were lysed
avec crises hemoglobinuriques” were lysed, whereas nor- and small populations of abnormal cells could not be
mal cells were not.4 Although he suspected that comple- detected. With the optimization of these factors, the test
ment was the cause of the hemolysis, he could not prove it became both specific (with the exception noted below) and
because the amount of fresh serum he added after inactiva- sensitive.
tion of complement was insufficient to carry the reaction. Even more intriguing is the question of what is different
Both Hijmans van den Bergh and Ham noted that about PNH cells that results in their lysis in this reaction.
acidification of the patient’s own serum could affect the We showed that the reaction resulted in the deposition of
lysis of his own cells. For many years, this was puzzling large amounts of C3b on the cell membrane” and that this
because antibody was the only known means by which was due to a greater efficiency of the convertase complex on
complement could be activated and no evidence of autoanti- the PNH membrane.” This proved to be due to the fact that
bodies could be detected in the autologous serum of most PNH cells lack a membrane protein that regulates the
patients. The explanation of this paradox became apparent activity of the “convertase” complexes, C3bBb and C4b2a.I3
when Pillemer et a15 delineated the alternative pathway of This protein accelerates the disassembly of the bimolecular
complement activation that did not depend on antibody. It complexes and was called “decay accelerating factor” or
was soon shown that the activated complement responsible DAF; more recently, it has been assigned the designation
for the lysis in acidified serum arose by activation of this CD55. It was thought at first that its absence explained the
alternative pathway, and this reaction remained one of the sensitivity of these cells to activated ~omplement,’~ but it
strongest arguments in favor of the alternative pathway at a was soon realized that another factor or factors must also
time when its very existence was under considerable attack.6 be missing to account for the reactions that were ~bserved.’~
Two questions were asked by the acidified serum test: (1) That factor is the “membrane inhibitor of reactive lysis”
How is complement activated? and (2) Why are PNH or MIRL (now assigned the designation CD59).I6 Holguin
erythrocytes lysed whereas normal cells are not? et all7showed that this protein, missing totally or in part on
The reactions of the alternative pathway have been PNH red blood cells, regulates the activation of the
carefully worked out.’ The primary step is the fixation to the membrane attack complex, which consists of C5b-9. Two
membrane of C3; this is accomplished either through the groups have shown that at least one of the actions of the
spontaneous “tick over” activation of plasma C3 through protein is to diminish the rate of assembly of the polymer-
spontaneous hydrolysis of a thiolester bridge characteristic ized C9 complex’X.”through an interaction with both CS
of the molecule or through the enzymatic activation of the and C9.”
natural molecule, again with the resultant rupture of the A third protein has been postulated, the CS binding
thiolester bond and covalent fixation to membrane enti- protein” or homologous restriction factor.22The character-
ties.’.’ Once C3b is atfixed, it binds factor B, which is then ization of this protein has been very difficult and it is now
activated to a serine protease after cleavage by factor D. suspected that, in fact, it is simply a polymeric or otherwise
This “convertase” complex, C3bBb, can then act as an altered form of CD59.
“autocatalytic” amplification step in enzymatically cleaving How CD55 and CD59 interact to bring about the protec-

Blood, Val 78, No 3 (August 1). 1991: pp 547-550 547


From www.bloodjournal.org at NATIONAL INSTITUTES OF HEALTH LIB on February 12, 2009. For personal
use only.

548 EDITORIAL

tion of normal cells is made more clear by the work of If the lack of GPI-linked proteins is the fundamental
Wilcox et al. They demonstrate in the accompanying defect in PNH, then we should be able to explain all the
reportz3that CD59 is the more important and that it may clinical manifestations by their absence. As yet, we cannot.
have some effect on the assembly or stability of the The marked tendency to thrombosis that these patients
convertase complex as well as on the formation of poly- have may be in part due to the absence of CD59. Ando et
meric C9 complexes. Whatever its actions, the replacement a14' have shown that the insertion of polymeric C9 into the
of CD59 to the deficient cells in PNH restores most of the membrane of the platelet results in a process by which they
protection of normal cells against the attack by comple- are removed in external vesicles without lysis of the platelet.
ment. These vesicles do not keep the acidic phospholipids internal-
The lack on the cells of PNH patients of several proteins ized, thus, they become the sites of prothrombinase forma-
in addition to CD55 and CD59 (erythrocyte acetylcholinest- tion and are strongly p r o c ~ a g u l a n tIf. ~the
~ CD59 of normal
e ~ a s e , 'leukocyte
~ alkaline phosphatase," CD16,'6 CD14,z7 platelets is inhibited by antibody, more of these vesicles are
CD71,28etc) suggested a common posttranslational defect formed because more nascent sites of polymerized C9 are
rather than genetic deletion. It was found that all of these built up. We have recently shown in preliminary data that
proteins are attached to the membrane by a glycosyl PNH platelets form three to five times as many vesicles as
phosphatidylinositol (GPI) anchor that consists of phos- normal platelets when limited activation of complement
phatidylinositol, N-glucosamine, three mannose residues, occurs. This might account for at least part of the throm-
and e t h a n ~ l a m i n eThe
. ~ ~nascent proteins were attached by botic tendency in these patients.
their carboxyl terminus to the ethanolamine by a transami- The explanation of other symptoms is even more obscure.
dation reaction in the endoplasmic reticulum. The defect in Most patients have a deficiency of hematopoiesis that is
PNH has been thought to be due to some defect in these greater or lesser in magnitude. A few patients become
processes. leukemic after having the PNH for several years. These
Most recent evidence has suggested that the defect is in manifestations are not readily explained by what we now
the synthesis of the anchor. Stafford et aI3' have shown that know about the GPI proteins and their absence in these
PNH granulocytes have normal mRNA and synthesize a cells.
protein of aberrant size.3' In a series of 10 patients, we have
found that PNH granulocytes are unable to synthesize the
complete anchor." Although the exact step or steps at HEMPAS
which the defect occurs has not been delineated, it is In only one other syndrome have the red blood cells been
probably in the addition of the mannose or ethanolamine shown to be lysed in acidified serum: hereditay erythroblas-
residues because these cells appear to be able to add the tic multinuclearity with an acidified serum lysis test (some-
N-acetyl-glucosamine to phosphatidylinositol and to deacet- times acronymically HEMPAS) or congenital dyserythropoi-
ylate it.33In GPI-deficient murine lymphocyte cell lines, etic anemia type II.45This syndrome is so clearly different
defects at each of the steps in the synthesis of the anchor from PNH that no difficulty in distinguishing them is
have been defined,34and the same may be true of PNH. apparent. It is a congenital disorder and is characterized by
What ever the defect or defects, it is not likely to be ineffective erythropoiesis with marked multinuclearity of
absolute in most cases. Most patients with PNH have the erythroblast^.^^ The red blood cells, once circulating,
erythrocytes that are intermediate in their sensitivity to have a relatively normal survival.
complement?' These cells uniformly bear small amounts of The reactions that bring about the lysis of the red blood
CD55 and CD59,34336 again, even small amounts of comple- cells in acidified normal serum are very different from those
ment-protective proteins largely spare the cells premature that cause lysis of PNH cells. In this case, the lysis is
destruction in vivo because the survival of these PNH I1 mediated by an IgM antibody that is present in most normal
cells is only slightly ~hortened.~' serum that reacts with an antigen present only on the cells
Recent studies have suggested that PNH granulocytes of patients with this syndrome; this antibody can be
deficient in CD55 and CD59 may possess CD16, the absorbed from normal serum by HEMPAS cells but not by
Fc,,,,,,,III receptor, another GPI-linked p r ~ t e i n . ~Al-
' normal cells.47The serum of patients with the disorder
though proof is lacking, it may be that this molecule is always lack the antibody and thus, unlike the case in PNH,
preferentially transamidated to the anchor because of its lysis does not occur in autologous serum.
amino acid structure. Some investigators have examined These cells are more readily lysed by complement when it
the conditions for this transamidation and have found that is activated by cold agglutinin antibodies. They exhibit
a hydrophobic sequence of at least 13 amino acids is extraordinarily large quantities of the i antigen on the
req~ired.~'~~'' Six amino acids can serve as acceptors of the surface but often react the same as normal cells with anti-I
amide and these varied in the degree to which they were antibodies. The increased lysis occurs despite the fact that
transamidated to the anchor; the terminal residue that was both HEMPAS cells and normal cells may bind the same
most readily transamidated was ~ e r i n e , ~which ' is the amount of antib~dy.~' Careful analysis showed that this was
terminal residue of the mature GPI-linked form of CD16." due to the fact that much more C4 was fixed for a given
It may be that, when limited quantities of anchor are amount of antibody and C1 on HEMPAS cells than on
synthesized, a disproportionate amount is attached to this normal cells, thus accounting for the greater activation of
molecule. complement. This contrasts with PNH cells on which the
From www.bloodjournal.org at NATIONAL INSTITUTES OF HEALTH LIB on February 12, 2009. For personal
use only.

EDITORIAL 549

fixation of C4 is normal but, because of the defects defective glycosylation of surface membrane proteins, result-
described previously, the fixation of C3 is vastly greater. ing in the loss of sialic acid from the surface.
This defect in HEMPAS cells closely resembles the effect of How these defects result in the erythroblastic multinucle-
removal of sialic acid by the action of sialidase on the arity characteristic of this syndrome is not clear, and this
reactions of has led to the suggestion that these defects are secondary to
The defect in HEMPAS has been ascribed to the inability other dyshematopoietic defects. However, no other specific
to synthesize certain biantennary lactosaminylglycansof the defects have been found. Furthermore, the fact that three
hematopoietic cells. These molecules are constructed on a enzyme defects that lead to the same biochemical result
lead to a similar cellular abnormality suggests that these
complex polymannose core and have N-acetylglucosamine
structures are important in membrane biosynthesis.
as the first sugar of a long series of repeated aminyllactose
The somewhat serendipitous observation of Dr Ham has
group^.^" The molecules terminate in a sialic acid residue. turned out to be one of great interest indeed. It has allowed
Three enzyme defects involved in the generation of these the simple and accurate diagnosis of a disease of some
moieties have been found to result in the abnormalities importance. Equally important, the investigation of its basis
characteristic of HEMPAS. In one, one of the enzymes has led to a number of seminal observations in immunology
responsible for the first N-acetylglucosamine is missing, and cell biology. Rarely has so much come from such simple
resulting in the absence of the entire sialylated antenna?l In beginnings.
the second, there is a a deficiency of the membrane-bound
form of galactosyltransfera~e,~~ which results in markedly WENDELL F. ROSSE, MD
attenuated antennae. In the third, the metabolism of the Departments of Medicine and Microbiology and Immunology
mannose-containing core is defective because of a defect in Duke UniversityMedical Center
the gene encoding a-mannosidase II.53 In each, there is Durham, NC

REFERENCES
1. Ham TH: Chronic hemolytic anemia with paroxysmal noctur- activity in the alternative pathway convertase when bound to the
nal hemoglobinuria. A study of the mechanism of hemolysis in erythrocytes of paroxysmal nocturnal hemoglobinuria. J Clin Invest
relation to acid-base equilibrium N Engl J Med 217:915,1937 69:337, 1982
2. Ham TH, Dingle JH: Studies on destruction of red blood 13. Nicholson-Weller A, March JP, Rosenfeld SI, Austen K F
cells. 11. Chronic hemolytic anemia with paroxysmal nocturnal Affected erythrocytes of patients with paroxysmal nocturnal hemo-
hemoglobinuria: Certain immunological aspects of the hemolytic globinuria are deficient in the complement regulatory protein,
mechanism with special reference to serum complement. J Clin decay accelerating factor. Proc Natl Acad Sci USA 80:5430,1983
Invest 18:657,1939 14. Medof ME, Kinoshita T, Silber R, Nussenzweig V: Amelio-
3. Ham TH: Studies on the destruction of red blood cells. I. ration of the lytic abnormalities of paroxysmal nocturnal hemoglo-
Chronic hemolytic anemia with paroxysmal nocturnal hemoglobin- binuria with decay accelerating factor. Proc Natl Acad Sci USA
uria: An investigation of the mechanism of hemolysis with observa- 82:2980,1985
tions on five cases. Arch Intern Med 64:1271,1939 15. Medof ME, Gottleib A, Kinoshita T, Hall S, Silber R,
4. Hijmans van den Bergh AA: Ictere hemolytique avec crises Nussenzweig V, Rosse WF: Relationship between decay accelerat-
hemoglobinuriques. Fragilite globulaire. Rev Medecine 31:63, ing factor deficiency, diminished acetylcholinesterase activity, and
1911 defective terminal complement pathway restriction in paroxysmal
5. Pillemer L, Blum L, Lepow IH, Ross OA, Todd EA, Wardlaw nocturnal hemoglobinuria erythrocytes. J Clin Invest 80:165, 1987
AC: The properdin system and immunity: I. Demonstration and 16. Holguin MH, Frederick LR, Bernshaw NJ, Wilcox LA,
isolation of a new serum protein, properdin, and its role in immune Parker CJ: Isolation and characterization of a membrane protein
phenomena. Science 120:279,1954 from normal human erythrocytes that inhibits reactive lysis of the
6. Hinz CJ Jr, Jordan WS Jr, Pillemer L The properdin system erythrocytes of paroxysmal nocturnal hemoglobinuria. J Clin Invest
and immunity. IV. The hemolysis of erythrocytes from patients 84:7, 1989
with paroxysmal nocturnal hemoglobinuria. J Clin Invest 35:453, 17. Holguin MH, Wilcox LA, Bernshaw NJ, Rosse, WF, Parker
1956 CJ: Relationship between the membrane inhibitor of reactive lysis
7. Pangburn MK, Muller-Eberhard HJ: The alternative pathway and the erythrocyte phenotypes of paroxysmal nocturnal hemoglo-
of complement. Springer Semin Immunopathol7:163,1984 binuria. J Clin Invest 84:1387.1989
8. Pangburn MK, Muller-Eberhard HJ: Relation of a putative 18. Rollins SA, Sims PJ: The complement-inhibitory activity of
thioester bond in C3 to activation of the alternative pathway and CD59 resides in its capacity to block incorporation of C9 into
the binding of C3b to biological targets of complement. J Exp Med membrane C5b-9. J Immunol144:3478,1990
152:1102,1980 19. Meri S, Morgan BP, Davies A, Daniels RH, Olavesen MG,
9. Lachmann PJ, Hughes-Jones NC: Initiation of complement Waldmann H, Lachmann PJ: Human protectin (CD59), an 18,000-
activation. Springer Semin Immunopathol7:143,1984 20,000 MW complement lysis restricting factor, inhibits C5b-8
10. May JE, Frank MM, Rosse WF: Alternate complement- catalyzed insertion of C9 into lipid bilayers. Immunology 71:1,1990
pathway-mediated lysis induced by magnesium. N Engl J Med 20. Rollins SA, Zhao JI, Ninomiya H, Sims PJ: Inhibition of
298:705,1973 homologous complement by CD59 is mediated by a species-
11. Logue GL, Rosse WF, A d a m JP: Mechanism of immune selective recognition conferred through binding to C8 within C5b-8
lysis of red blood cells in vitro.I. Paroxysmal nocturnal hemoglobin- or C9 within C5b-9. J Immunol 146:2345, 1991
uria cells. J Clin Invest 42:1129, 1973 21. Schonermark S, Rauterberg EW, Shin ML, Loke S, Roelcke
12. Parker CJ, Baker PJ, Rosse W F Increased enzymatic D, Hansch GM: Homologous species restriction in lysis of human
From www.bloodjournal.org at NATIONAL INSTITUTES OF HEALTH LIB on February 12, 2009. For personal
use only.

550 EDITORIAL

erythrocytes: A membrane-derived protein with C8-binding capac- nal hemoglobinuria. Discordant expression of glycosyl phosphati-
ity functions as an inhibitor. J Immunol 136:1772, 1986 dylinositol-linked proteins. J Clin Invest 8758, 1991
22. Zalman LS, Wood LM, Muller-Eberhard HJ: Isolation of a 39. Caras IW, Weddell GN, Davitz MA, Nussenzweig V, Martin
human erythrocyte membrane protein capable of inhibition expres- DW Jr: Signal for attachment of a phospholipid membrane anchor
sion of homologous complement transmembrane channels. Proc in decay accelerating factor. Science 238:1280,1987
Natl Acad Sci USA 83:6975,1986 40. Micanovic R, Bailey CA, Brink L, Gerber L, Pan YC,
23. Wilcox LA, Ezzell JL, Bernshaw NJ, Parker CJ: Molecular Hulmes JD, Udenfriend S: Aspartic acid-484 of nascent placental
basis of the enhanced susceptibility of the erythrocytes of paroxys- alkaline phosphatase condenses with a phosphatidylinositol glycan
mal nocturnal hemoglobinuria to hemolysis in acidified serum. to become the carboxyl terminus of the mature enzyme. Proc Natl
Blood 1991 (in press) Acad Sci USA 851389,1988
24. Auditore JV, Harmann RC, Flexner JM, Balchum OJ: The 41. Micanovic R, Gerber LD, Berger J, Kodulkula K, Uden-
erythrocyte acetylcholinesterase enzyme in paroxysmal nocturnal friend S: Selectivity of the cleavageiattachment site of phosphatidy-
hemoglobinuria. Arch Pathol69534, 1960 linositol-glycan-anchoredmembrane proteins determined by site-
25. Beck WS, Valentine W N Biochemical studies on leucocytes. specific mutagenesis at Asp-484 of placental alkaline phosphatase.
11. Phosphatase activity in chronic lymphatic leukemia, acute Proc Natl Acad Sci USA 87:157,1990
leukemia, and miscellaneous hematologic conditions. J Lab Clin 42. Kurosaki T, Ravetch JV: A single amino acid in the glycosyl
Med 38:245,1951 phosphatidylinositol attachment domain determines the mem-
26. Selvaraj P, Rosse WF, Silber R, Springer TA: The major Fc brane topology of Fc gamma RIII. Nature 342:805,1989
receptor in blood has a phosphotidylinositol anchor and is deficient 43. Ando B, Wiedmer T, Sims PJ: The secretoly release reaction
in paroxysmal nocturnal hemoglobinuria. Nature 333565, 1988 initiated by complement proteins C5b-9 occurs without platelet
27. Simmons DL, Tan S, Tenen DG, Nicholson-Weller A, Seed aggregation through glycoprotein IIb-IIIa. Blood 73:462, 1989
B: Monocyte antigen CD14 is a phospholipid anchored membrane 44. Sims PJ, Faioni EM, Wiedmer T, Shattil SJ: Complement
protein. Blood 73:284,1989 proteins C5b-9 cause release of membrane vesicles from the
28. Selvaraj P, Dustin ML, Silber R, Low MG, Springer TA: platelet surface that are enriched in the membrane receptor for
Deficiency of lymphocyte function-associated antigens 3 (LFA-3) coagulation factor Va and express prothrombinase activity. J Biol
in paroxysmal nocturnal hemoglobinuria. Functional correlates Chem 263:18205,1988
and evidence for a phosphatidylinositol membrane anchor. J Exp
45. Crookston JH, Crookston MC, Burnie KL, Francombe WH,
Med 166:1011,1987
Hereditary erythroblastic multinuclearity
29. Cross GAM: Glycolipid anchoring of plasma membrane
ve acidified-serum test: A type of congenital
proteins. Annu Rev Cell Biol6:1,1990
dyserythropoietic anaemia. Br J Haematol 17:11,1969
30. Stafford HA, Tykocinski ML, Lublin DM, Holers VM, Rosse
46. Venvilghen RL, Lewis SM, Dacie JV, Crookston JH, Crook-
WF, Atkinson JP, Medof ME: Normal polymorphic variations and
ston M: HEMPAS: Congenital dyserythropoietic anemia (Type 11).
transcription of the decay accelerating factor gene in paroxysmal
Q J Med 42:257,1973
nocturnal hemoglobinuria cells. Proc Natl Acad Sci 85:880, 1988
31. Carothers DJ, Hazra SV, Andreson SW, Medof ME: Synthe- 47. Crookston JH, Crookston MC, Rosse WF: Red cell abnor-
sis of aberrant decay-accelerating factor proteins by affected malities in HEMPAS (hereditary erythroblastic multinuclearity
paroxysmal nocturnal hemoglobinuria leucocytes. J Clin Invest with a positive acidified-serum test). Br J Haematol 23:83, 1983
8547,1990 (SUPPI)
32. Mahoney JF, Urakaze M, Hall S, DeGasperi R, Chang H-M, 48. Rosse WF, Logue GL, A d a m J, Crookston JH: Mechanisms
Warren CD, Nicholson-Weller A, Rosse WF, Yeh ETH: Defective of immune lysis of the red cells in hereditary evthroblastic
glycosylphosphatidylinositol anchor synthesis in paroxysmal noctur- multinuclearity with a positive acidified serum test and paroxysmal
nal hemoglobinuria. Clin Res 39:210A, 1991 (abstr) nocturnal hemoglobinuria. J Clin Invest 53:31, 1974
33. Hirose S, Ravie L, Hazra SV, Medof ME: Assembly and 49. Rosse WF: HEMPAS, in Clinical Immunohematology: Basic
deacetylation of N-acetylglucosaminyl-plamanylinositolin normal Concepts and Clinical Applications. New York, NY, Blackwell
and affected paroxysmal nocturnal hemoglobinuria cells. Proc Natl Scientific, 1990
Acad Sci USA 88:3762,1991 50. Jarnefelt J, Ruksh J, Li Y-T, Laine RA: Erythroglycan, a
34. Sugiyama E, DeGasperi R, Urakaze M, Chang H-M, Tho- high molecular weight glycopeptide with the repeating structure
mas LT, Hymans R, Warren CD, Yeh ETH: Identification of [galactosyl (1 4)-2-deoxy-2-acetaminoglucosyl(1 3)] comprising
defects in GPI anchor biosynthesis in the Thy-1 expression mu- more than one-third of the protein-bound carbohydrate of the
tants. J Biol Chem (in press) human erythrocyte stroma. J Biol Chem 253:8006,1978
35. Rosse WF, Hall S, Campbell M, Borowitz M, Moore JO, 51. Fukuda MN, Dell A, Scartezzini P: Primary defect of
Parker CJ: The erythrocytes in paroxysmal nocturnal hemoglobin- congenital dyserythropoietic anemia type 11. Failure in glycosyla-
uria of intermediate sensitivity to complement lysis. Br J Haematol tion of erythrocyte lactoaminoglycan proteins caused by lowered
(in press) N-acetylaminylglucosetransferase11. J Biol Chem 262:7195,1987
36. Holguin MH, Wilcox LA, Bernshaw NJ, Rosse, WF, Parker 52. Fukuda MN, Masri KA, Dell A, Thonar EJ, Klier G,
CJ: Relationship between the membrane inhibitor of reactive lysis Lowenthal RM: Defective glycosylation of erythrocyte membrane
and the erythrocyte phenotypes of paroxysmal nocturnal hemoglo- glycoconjugates in a variant of congenital dyserythropoietic anemia
binuria. J Clin Invest 84:1387,1989 type 11: Association of low level of membrane-bound form of
37. Rosse WF: The life-span of complement-sensitive and -insen- galactosyltransferase. Blood 73: 1331,1989
sitive red cells in paroxysmal nocturnal hemoglobinuria. Blood 53. Fukuda MN, Masri KA, Dell A, Luzzatto L, Morement KW:
37:556, 1971 Incomplete systhesis of N-glycans in congenital dyserythropoietic
38. Edberg JC, Salmon JE, Whitlow M, Kimberly RP: Preferen- anemia type I1 caused by a defect in the gene encoding alpha-
tial expression of human Fc RIIIpMN (CD16) is paroxysmal noctur- mannosidase 11. Proc Natl Acad Sci USA 87:7443,1990

You might also like