13
233 © 2001 Schattauer GmbH, Stuttgart Thromb Haemost 2001; 86: 233 – 45 Key words Albinism, platelet dense bodies, giant lysosomes, LYST, intracellular trafficking Summary The rare autosomal recessive metabolic disorders Hermanky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS) share the clini- cal findings of oculocutaneous albinism and a platelet storage pool deficiency. In addition, HPS exhibits ceroid lipofuscinosis and CHS is characterized by infections and an accelerated phase. The two dis- orders result from defects in vesicles of lysosomal lineage. Of the two known HPS-causing genes, HPS1 has no recognizable function, while ADTB3A codes for a subunit of an adaptor complex responsible for new vesicle formation from the trans-Golgi network. Other HPS- causing genes are likely to exist. The only known CHS-causing gene, LYST, codes for a large protein of unknown function. In general, HPS appears to be a disorder of vesicle formation and CHS a defect in vesicle trafficking. These diseases and their variants mirror a group of mouse hypopigmentation mutants. The gene products involved will reveal how the melanosome, platelet dense body, and lysosome are formed and trafficked within cells. Introduction Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued the inter- est of cell biologists focusing on the genesis and movement of intra- cellular vesicles (1-6). In this review, we describe these two diseases and demonstrate why they provide insight into cell biological proces- ses. In fact, we propose that HPS and CHS represent extremes of a spectrum of disease entities that correspond to discrete steps in the genesis and movement of vesicles of lysosomal lineage. Hermansky-Pudlak Syndrome HPS, first reported in 1959 by two Czechoslovakian pathologists (7), consists of a collection of genetically distinct defects having com- mon clinical and laboratory findings (3, 4). All patients have some degree of oculocutaneous albinism and platelet storage pool deficiency. Many also demonstrate ceroid lipofuscinosis, although the presence of this intracellular lipid/protein complex is no longer necessary to make the diagnosis of HPS. In our own studies, we have also arbitrarily required that dense bodies be absent from platelets for the diagnosis of HPS to be confirmed, although future clinical/molecular correlations may mandate modification of this requirement. What constitutes HPS will be discussed below, after description of the disease as we currently know it. Clinical and Laboratory Findings The clinical manifestations of HPS are variable (3, 4) because the physiologic processes involved are modulated by a panoply of genetic and environmental influences, and because the genetic basis of HPS is itself heterogeneous. Despite these caveats, some generalizations can be made regarding the signs and symptoms of HPS. Oculocutaneous albinism. In HPS, tyrosinase activity remains present within melanocytes (2), but melanosome function is impaired, and pigment dilution results. This affects skin, hair, and eye color, but to variable extents. Hair color ranges from nearly normal to completely white (Fig. 1A, B). Even among the approximately 400 Puerto Rican patients with exactly the same mutation in the HPS1 gene (see below), the spectrum of hair color varies enormously (8). Typically, the hair is dirty blond or tan. HPS skin also varies from pale to brown, and often must be compared with that of an unaffected sibling to verify hypo- pigmentation. HPS patients are susceptible to solar damage, including actinic keratoses, nevi, and skin malignancies (9). Ophthalmic involvement in HPS patients (10-12) includes increased crossing over of nerve fibers at the optic chiasm, typical of albinism. Iris transillumination (Fig. 1C, D) occurs because cells in the iris have reduced amounts of pigment. Loss of retinal pigment epithelium causes a pale fundus and decreased visual acuity, usually 20/200 or worse (legally blind), but sometimes as high as 20/50 (8, 12). Vision in HPS is stable, uncorrectable, and accompanied by variable degrees of con- genital nystagmus. Hematologic findings. In HPS, platelet counts are normal or ele- vated, but the platelets are dysfunctional because they lack dense bodies (Fig. 1E, F). These electron dense intracellular organelles, which contain ADP, ATP, serotonin, calcium, and polyphosphates (13), disgorge their contents upon activation and promote clot formation. Absence of dense bodies attenuates the secondary aggregation res- ponse and can prolong the bleeding time. HPS patients exhibit findings typical for a platelet storage pool defi- ciency, i. e., bleeding of mucosal membranes and spontaneous soft tissue bruising. Major bleeds into the joints or brain are not seen, and coagulation factors, prothrombin times, and partial thromboplastin times are normal (1, 8). Excess bruising usually begins at ambulation, Correspondence to: William A. Gahl, MD, PhD, 10 Center Drive, MSC 1830, Building 10, Room 9S-241, NICHD, NIH, Bethesda, Maryland 20892-1830, USA – Tel.: 3 01-4 02-27 39; Fax: 3 01-4 02-27 40; E-mail: [email protected] Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome: Disorders of Vesicle Formation and Trafficking Marjan Huizing, Yair Anikster, William A. Gahl Section on Human Biochemical Genetics, Heritable Disorders Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA For personal or educational use only. No other uses without permission. All rights reserved. Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

  • Upload
    others

  • View
    11

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

233

© 2001 Schattauer GmbH, Stuttgart Thromb Haemost 2001; 86: 233–45

Key words

Albinism, platelet dense bodies, giant lysosomes, LYST, intracellulartrafficking

Summary

The rare autosomal recessive metabolic disorders Hermanky-Pudlaksyndrome (HPS) and Chediak-Higashi syndrome (CHS) share the clini-cal findings of oculocutaneous albinism and a platelet storage pooldeficiency. In addition, HPS exhibits ceroid lipofuscinosis and CHSis characterized by infections and an accelerated phase. The two dis-orders result from defects in vesicles of lysosomal lineage. Of the twoknown HPS-causing genes, HPS1 has no recognizable function, whileADTB3A codes for a subunit of an adaptor complex responsible for new vesicle formation from the trans-Golgi network. Other HPS-causing genes are likely to exist. The only known CHS-causing gene,LYST, codes for a large protein of unknown function. In general, HPSappears to be a disorder of vesicle formation and CHS a defect invesicle trafficking. These diseases and their variants mirror a group ofmouse hypopigmentation mutants. The gene products involved willreveal how the melanosome, platelet dense body, and lysosome areformed and trafficked within cells.

Introduction

Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome(CHS), share several clinical characteristics and have piqued the inter-est of cell biologists focusing on the genesis and movement of intra-cellular vesicles (1-6). In this review, we describe these two diseasesand demonstrate why they provide insight into cell biological proces-ses. In fact, we propose that HPS and CHS represent extremes of aspectrum of disease entities that correspond to discrete steps in thegenesis and movement of vesicles of lysosomal lineage.

Hermansky-Pudlak Syndrome

HPS, first reported in 1959 by two Czechoslovakian pathologists(7), consists of a collection of genetically distinct defects having com-mon clinical and laboratory findings (3, 4). All patients have somedegree of oculocutaneous albinism and platelet storage pool deficiency.

Many also demonstrate ceroid lipofuscinosis, although the presence ofthis intracellular lipid/protein complex is no longer necessary to makethe diagnosis of HPS. In our own studies, we have also arbitrarilyrequired that dense bodies be absent from platelets for the diagnosis ofHPS to be confirmed, although future clinical/molecular correlationsmay mandate modification of this requirement. What constitutes HPSwill be discussed below, after description of the disease as we currentlyknow it.

Clinical and Laboratory Findings

The clinical manifestations of HPS are variable (3, 4) because thephysiologic processes involved are modulated by a panoply of geneticand environmental influences, and because the genetic basis of HPS isitself heterogeneous. Despite these caveats, some generalizations canbe made regarding the signs and symptoms of HPS.

Oculocutaneous albinism. In HPS, tyrosinase activity remainspresent within melanocytes (2), but melanosome function is impaired,and pigment dilution results. This affects skin, hair, and eye color, butto variable extents. Hair color ranges from nearly normal to completelywhite (Fig. 1A, B). Even among the approximately 400 Puerto Ricanpatients with exactly the same mutation in the HPS1 gene (see below),the spectrum of hair color varies enormously (8). Typically, the hair isdirty blond or tan. HPS skin also varies from pale to brown, and oftenmust be compared with that of an unaffected sibling to verify hypo-pigmentation. HPS patients are susceptible to solar damage, includingactinic keratoses, nevi, and skin malignancies (9).

Ophthalmic involvement in HPS patients (10-12) includes increasedcrossing over of nerve fibers at the optic chiasm, typical of albinism.Iris transillumination (Fig. 1C, D) occurs because cells in the iris havereduced amounts of pigment. Loss of retinal pigment epithelium causesa pale fundus and decreased visual acuity, usually 20/200 or worse(legally blind), but sometimes as high as 20/50 (8, 12). Vision in HPSis stable, uncorrectable, and accompanied by variable degrees of con-genital nystagmus.

Hematologic findings. In HPS, platelet counts are normal or ele-vated, but the platelets are dysfunctional because they lack dense bodies (Fig. 1E, F). These electron dense intracellular organelles, whichcontain ADP, ATP, serotonin, calcium, and polyphosphates (13), disgorge their contents upon activation and promote clot formation.Absence of dense bodies attenuates the secondary aggregation res-ponse and can prolong the bleeding time.

HPS patients exhibit findings typical for a platelet storage pool defi-ciency, i. e., bleeding of mucosal membranes and spontaneous softtissue bruising. Major bleeds into the joints or brain are not seen, andcoagulation factors, prothrombin times, and partial thromboplastintimes are normal (1, 8). Excess bruising usually begins at ambulation,

Correspondence to: William A. Gahl, MD, PhD, 10 Center Drive,MSC 1830, Building 10, Room 9S-241, NICHD, NIH, Bethesda, Maryland20892-1830, USA – Tel.: 301-402-2739; Fax: 301-402-2740; E-mail:[email protected]

Hermansky-Pudlak Syndrome and Chediak-HigashiSyndrome: Disorders of Vesicle Formation and Trafficking

Marjan Huizing, Yair Anikster, William A. Gahl

Section on Human Biochemical Genetics, Heritable Disorders Branch,National Institute of Child Health and Human Development,

National Institutes of Health, Bethesda, Maryland, USA

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Page 2: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

234

Thromb Haemost 2001; 86: 233–45

and epistaxis begins in childhood. Bleeding episodes can occur duringdental extractions, surgeries, acute colitis, menstrual periods, and childbirth. Fatal bleeds are rare but a significant number of patients have re-ceived transfusions of packed red cells or platelets.

Interestingly, von Willebrand factor, which is stored within plateletalpha granules, is decreased in many HPS patients (14, 15). Lympho-cyte and neutrophil function appear normal in HPS (16).

Pulmonary fibrosis. Many HPS patients suffer a progressive restric-tive lung disease beginning in their late twenties or early thirties (1, 8,17, 18), perhaps related to accumulation of ceroid in HPS alveolarmacrophages (Fig. 1G, H). Affected patients typically exhibit extensivefibrosis by the end of the fourth or fifth decade of life (Fig. 1I, J).Infection, smoking or exposure to pulmonary toxins can lead to deathwithin two or three years. The pulmonary involvement of HPS variesconsiderably, and occasional patients are over 60 years old with normallung function. Some subgroups of HPS, specifically, patients bearingmutations in the HPS1 gene (see below), are more susceptible to pul-monary fibrosis than others (8, 18). Pulmonary fibrosis has been notedin HPS patients from Czechoslovakia, Japan, Belgium, and England(3), prior to the availability of molecular genotyping.

Granulomatous colitis. This complication affects at least 15% of allHPS patients, whether or not they are of Puerto Rican heritage (8).Although the distal colon is most often involved, the entire gastro-intestinal tract may be affected, with granulomatous gingivitis occuringin rare cases (1). Patients present with blood and mucus in their stools,and the colitis resembles Crohn’s disease histologically (19, 20).

Ceroid lipofuscin. This electron dense, autofluorescent aging pig-ment is increased in some HPS patients, who accumulate it within thekidneys, urinary sediment, lung alveolar macrophages, bone marrow,spleen, liver, and large intestine (1). Smaller amounts can be found inthe heart, lymph nodes and other tissues (1). The ceroid lipofuscin ofHPS appears to reside within lysosomes. Some theories blame ceroidlipofuscin accumulation for the pulmonary fibrosis, granulomatouscolitis, and occasional renal impairment (1, 8) and cardiomyopathy (1)of HPS. However, no convincing evidence has been put forth to supportthis.

Subtypes of HPS

Two patients have been reported with mutations in the ADTB3Agene (21, 22) (see below), and we have recently identified a third.All three had persistent neutropenia and childhood infections (22).Balance problems in the two reported cases may be related to decreasedvisual acuity. In other respects, the clinical findings are typical for HPS,i. e., moderate visual impairment, nystagmus, hypopigmentation, andabsence of platelet dense bodies.

Another subtype of HPS exists in central Puerto Rico, where affect-ed individuals have absent dense bodies but mild hypopigmentationand visual acuity deficits. The HPS1 gene has been eliminated as acause for the disease in this group of patients (23). As other HPS-causing genes are identified, additional clinical subtypes should emerge.

Diagnosis

The diagnosis of HPS relies on the clinical suspicion of the physician.Dirty blond hair color, excessive bruising in toddlers, and epistaxis inmiddle childhoood provide hints, but bleeding times may remain nor-mal. Nevertheless, virtually every patient has some degree of horizon-tal nystagmus present from birth (12), and every patient has a visualacuity of 20/50 or poorer.

Fig. 1 Clinical Findings in HPS. A. Typical light brown/dirty brown haircolor in a 19-year old Puerto Rican patient. B. Extreme lack of hair pigmentin a 9-year old Puerto Rican boy with HPS. C. Mild iris transillumination ina 4-year old girl from central Puerto Rico with no mutation in HPS1. Note resid-ual, scattered pigment. D. Nearly complete iris transillumination in a 40-yearold patient from northwest Puerto Rico with the classic 16-bp duplicationin HPS1. E. Wet mount electron microscopy showing normal platelet densebodies in an unaffected individual. F. Platelet of an HPS patient showing absentdense bodies. G. Normal pulmonary alveolar macrophage. H. Alveolar macro-phage from a 34-year old northwest Puerto Rican patient showing engorgedvesicles typical of ceroid lipofuscinosis. I. Normal chest CT scan of a 53-yearold Puerto Rican HPS patient with normal pulmonary function. J. Chest CTscan from a 38-year old Puerto Rican HPS patient with severe pulmonaryfibrosis. (C, D courtesy of Dr. M. I. Kaiser-Kupfer and Mr. E. Kuehl, Natio-nal Eye Institute, NIH; E, F courtesy of Dr. James White, University of Minne-sota)

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Page 3: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

235

Huizing: Hermansky-Pudlak and Chediak-Higashi Syndromes

The sine qua non of the HPS diagnosis remains demonstration ofabsent platelet dense bodies using the method of wet-mount electronmicroscopy (24). This procedure is currently performed by Dr. JamesWhite of the University of Minnesota. Studies showing reduced orabsent serotonin uptake or an absent secondary aggregation responsecan also be helpful.

Many physicians, recognizing that HPS occurs with increasedfrequency among Puerto Ricans, mistakenly discount the diagnosiswhen caring for non-Puerto Rican patients. One HPS patient carriedthe diagnosis of isolated ocular albinism until her mid-twenties, whenher excessive bleeding during menstrual periods prompted an examina-tion for platelet dense bodies, which were absent by electron microscopy.We recommend consideration of platelet dense body analysis in albinopatients without a molecular basis for their disorder.

Treatment

For HPS patients, sun avoidance and sunscreens are critical to pre-vent solar damage, and skin cancer surveillance can detect early malig-nancies (9). For the bleeding diathesis of HPS, thrombin and gelfoamcan be used for minor wounds, and some, but not all, patients respondto 1-desamino-8-D-arginine vasopressin (DDAVP), 0.2 �g/kg in 50 mlof normal saline given intravenously over 30 minutes (25). Platelets can be either on hand or transfused prior to major procedures. The mostcrucial issue, however, is recognition of the functional platelet defect,which cannot be appreciated from a simple platelet count. Hence, theuse of a medical alert bracelet can be extemely helpful.

Only supportive care is available for the pulmonary fibrosis of HPS, al-though a double-blind, placebo-controlled trial of the investigational anti-inflammatory drug, pirfenidone, is currently underway. Lung transplan-tation, although never performed on an HPS patient, should be feasiblewith close attention to coagulation, and a transplanted lung should last thenatural lifetime of a patient. The granulomatous colitis of HPS generallyresponds to steroids and other treatments appropriate for Crohn’s disease.

Genetics

The population frequency of HPS in northwest Puerto Rico is 1in 1800, with a gene frequency of 1 in 21 (26). All 400 affected individ-uals in northwest Puerto Rico are homozygous for a specific mutation,reflecting a founder effect. However, HPS has been reported through-out the globe (1), with different mutations and different genes involved(27-30). Of the approximately 60 non-Puerto Rican HPS patientsexamined molecularly, only approximately 25-40% have their diseasedue to mutations in a known gene (28, 29). In addition to the knownHPS-causing genes HPS1 and ADTB3A, there is certainly anothergene causing the disease in central Puerto Rico (23). The many animalmodels of HPS (31) also indicate locus heterogeneity. All subtypes ofHPS appear to be inherited in an autosomal recessive fashion (1).

The HPS1 gene. The first HPS-causing gene was mapped to chromo-some 10q23.1-23.3 in 1995 using Puerto Rican and Swiss families (32).Subsequently, the gene (GenBank accession #1654350), called HPSand later HPS1, was sequenced and shown to contain an open readingframe of 2100 bp (27). The genomic structure was then determined (33).HPS1 consists of 20 exons spanning approximately 30.5 kb. Itsstandard transcript, 3.0 kb in size, is expressed in most tissues (27).Minor 3.9-kb, 4.4-kb, and 1.5-kb mRNAs also occur (34). Four alter-native splices of HPS1 have been described (27, 33). A pseudogeneof HPS1 (GenBank accession #6707403) exists on chromosome22q12.2-12.3 and contains complete exons 3, 4, and 6 (35).

The 16-bp duplication in exon 15 of HPS1 is present in the homo-zygous state in all northwest Puerto Rican patients but not in any otherpatient group (27-29). This frameshift mutation produces no mRNA,and is easily detected by PCR amplification (27). Other mutations inHPS1 include T322insC and T322delC, indicating that the region ofcodons 321-324 represents a mutation hot spot (28). Codon 396 maybe another area subject to recurrent mutation, since S396delC hasappeared in several other patients (28, 29). In all, 12 different mutations,including deletions, insertions, nonsense mutations, and splice junctionmutations, have been reported for HPS1 (3, 5, 30).

Most of the HPS1 gene mutations, as well as the frameshift mutationof the mouse homologue of HPS1, pale ear (36, 37) (see below), arepredicted to result in a protein with a truncated carboxy terminus and,presumably, no residual function. This points to the critical nature ofthe terminal portion of the HPS1 protein. To date, no missense mutationsin HPS1 have been found, although at least 24 nonpathologic DNAsequence polymorphisms have been reported (29, 30, 33).

The ADTB3A gene. ADTB3A (GenBank accession #1923267),whose coding sequence has 3281 bp, produces a 4.2-kb mRNA presentin all tissues and cell lines examined (38). The product is �3A, a subunitof adaptor complex-3 (AP3), which functions to form vesicles fromexisting membranes such as the trans-Golgi network, or TGN (39). Theonly reported cases of mutations in ADTB3A are those found in thecompound heterozygous state in two brothers affected with HPS (22).The mutations are a 21-amino acid deletion (∆390-410) and a singleamino acid substitution, L580R (21). Mutations in the pearl mouse, whichis the murine equivalent of the humans with ADTB3A mutations, inclu-de a 793-bp internal tandem duplication and a 107-bp deletion (40).

Cell Biology

HPS has long been considered a defect of vesicular membranes, inpart because the intracellular compartments causing the clinical mani-festations of the disease share integral membrane proteins. Specifically,the melanosome’s ME491 protein is the same as the platelet’s CD63or granulophysin which is the same as the lysosome’s LAMP-3 orLIMP-1 (3). In addition, two lysosomal membrane proteins, LAMP-1and LAMP-2, are also found on platelet dense body membranes (41).The hypothesis that HPS results from dysfunctional membrane traf-ficking has been strongly supported by the finding that AP3 deficiencycan cause HPS, and by the fact that several genes responsible for HPSin murine and Drosophila models are involved in vesicle formation andtrafficking (see below).

The HPS1 protein. The HPS1 gene product has 700 amino acids anda predicted molecular weight of 79.3 kDa (27), with no apparent glyco-sylation (42) and no homology to proteins with a known function.It does contain the sequence DKF(L/V)KNRG, which resembles aregion of the CHS protein (27, 43) (see below), and the carboxy termi-nus of the HPS1 protein contains a putative melanosomal localizationsignal, PLL (44). The amino acid sequence of the HPS1 protein is 81%conserved between human and mouse (36), with similar conservationbetween human and rat (5). A predicted Drosophila protein of un-known function has high sequence identity to the carboxy terminal portion of HPS1, but no homologues exist in lower species.

A role for the HPS1 protein in vesicle formation is suggested by theHis-Leu-Leu sequence near its carboxy teminus. This recognition mar-ker could serve as a sorting signal to target the protein to compartmentsof lysosomal lineage, such as the melanosome and dense granule(45, 46), although the HPS1 protein does not appear to be directly asso-ciated with lysosomes (42).

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Page 4: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

236

Thromb Haemost 2001; 86: 233–45

The HPS1 protein is a component of two high molecular weightcomplexes, a cytosolic complex of approximately 200 kDa in nonmela-notic cells and a larger, 500-kDa complex in melanotic cells (42). Thelarge complex is perinuclear and associated with tubulovesicular struc-tures, small noncoated vesicles, and nascent and early-stage melanoso-mes but not with later stage melanosomes (42). Other studies place theHPS1 protein in the perinuclear region of normal melanocytes, possiblyassociated with a cisternal network outside of the Golgi zone (47). Thissuggests that the protein is a part of the premelanosome as it forms fromthe smooth endoplasmic reticulum.

The HPS1 gene product appears to influence trafficking of melano-cyte-specific proteins from the TGN to preformed premelanosomes(48). Specifically, the pigment-forming proteins TRP-1 (for tyrosinaserelated protein-1) and granulophysin displayed a large granular patternof expression in HPS-1 melanocytes, consistent with localization tolarge membrane complexes visible ultrastructurally in the mutant cells(48). The appropriate location for these proteins would be within mela-nosomes.

The HPS1 protein is present on normal platelets but not on plateletsfrom patients with null mutations in HPS1. Other studies have shownthat fibroblasts do not require the HPS1 protein to grow, and that HPS1-deficient cells display a normal distribution and trafficking of the lyso-somal membrane proteins CD63 and LAMP-1. HPS1 does not appearto interact with AP3 (46).

The �3A subunit of AP3. The �3A protein is a subunit of AP3, oneof four known adaptor complexes (AP1, AP2, AP3, and AP4).Adaptors are components of cytosolic protein coats that mediate vesicleformation and incorporation of cargo proteins into the nascent vesicle’smembranes (39). AP3 apparently produces vesicles from the TGNand/or endosomes. It interacts with clathrin (49), which provides struc-ture to the newly formed vesicle. AP3 consists of a 160 kDA � subunit,a 47-kDA � subunit, a 23-kDa � subunit, and the 140 kDa �3A subunit(38, 50-52). The �3A subunit is phosphorylated on serine residues (38).The � subunit of AP3 recognizes tyrosine-based (50, 53) and dileucine-based (54) sorting signals of cargo proteins targeted for incorporationinto the membranes of new vesicles.

AP3-deficient fibroblasts display enhanced trafficking of lysosomalmembrane proteins through the plasma membrane (21), a defaultpathway employed when direct movement from the TGN to the endo-some is impaired. Similar findings were reported for fibroblasts of thepearl mouse bearing ADTB3A mutations (40) and the mocha mousecarrying mutations in the � subunit of AP3 (55). However, not allintegral membrane proteins use AP3 for targeting to lysosome-relatedorganelles. Trafficking of MHC class II molecules and the associatedinvariant chains to their intracellular compartments appeared normal inAP3-deficient cells (56).

In �3A-deficient human melanocytes, TRP-1 localization is normal,but tyrosinase expression is restricted to the perinuclear region (Fig. 2)and localized in large vesicles resembling late endosomes (57). More-over, the abnormal tyrosinase distribution is corrected by transfectionwith the normal �3A gene, supporting the concept that tyrosinase traf-ficking is regulated by AP3 (57).

Fibroblasts from patients with ADTB3A mutations show reduction ofthe �, �, and � subunits of AP3 as well; apparently the �3A subunitstabilizes the entire AP3 complex against degradation (21).

Models of Pigment Dilution in Other Organisms

The earliest and closest models for HPS consist of inbred micehaving variable degrees of pigment dilution combined with a plateletstorage pool deficiency (31) (Table 1). This group of mutants actuallyrepresents a spectrum of disease ranging from HPS to CHS and reflect-ing various human conditions (see below). Several of the model miceexhibit increased urinary excretion of lysosomal enzymes. All the HPSmice inherit their disease in an autosomal recessive fashion.

Six mouse models have had their causative genes isolated. Two ofthese, pale ear and pearl, represent the murine counterparts of humanswith mutations in HPS1 and ADTB3A, respectively. In fact, the humanADTB3A mutations were discovered because the murine pearl genewas found to code for the �3A subunit of AP3 (40). The mocha geneencodes the � subunit of AP3 (55) and pallid encodes a 25-kDa protein,pallidin (58), that interacts with syntaxin 13, part of the membrane

Table 1 Murine models of hypopigmentation due to impaired vesicle formation or trafficking

Chromosome HumanModel Mouse Human Gene Disease Comment

1. pale ear 19 10q23.1-23.3 HPS1 HPS-1 Small eumelanin granules in hair2. pearl 13 5q11-14 ADTB3A HPS-2 Night blindness in mouse; neutropenia in human3. mocha 10 19p13.3 AP3D1 – Small melanosomes in hair, hyperactivity, balance problem4. pallid 2 15q11.2-22.3 pallidin – Emphysema, balance defect, enhanced response to morphine5. gunmetal 14 14q11.2 RABGGTA – Both � and � granule defect, thrombocytopenia, large platelets6. cappuccino 5 4p15-16 – – Elevated liver lysosomal enzymes7. cocoa 3 8q13-22 – – Normal lysosomal function8. subtle grey 3 3q24-28 – – No lysosomal dysfunction, mild bleeding/pigment dilution9. light ear 5 4p16 – – Few, large melanin granules in choroid, small in hair

10. reduced 7 19q13 – – Increased anesthetic risk, small pigment granules in hairpigment

11. ruby eye 19 10q24.1-25.1 – – Reduced number of melanocytes12. ruby eye-2 7 11p15/15q11 – – –13. muted 13 6p21-23/5p15 – – Balance defect, ears lack otoliths14. sandy 13 6p21-23/5p15 – – –15. ashen 9 15q21 rab27a Griscelli Large pigment clumps on hairshafts, hemophagocytic syndrome

syndrome II16. dilute 9 15q21 myo5a Griscelli Neurologic involvement

syndrome I17. leaden 1 – – – Large pigment clumps on hairshafts18. beige 13 1q42.1-42.2 lyst CHS Giant intracellular granules

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Page 5: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

237

Huizing: Hermansky-Pudlak and Chediak-Higashi Syndromes

fusion machinery (59). The gunmetal gene codes for the � subunit ofrab geranylgeranyltransferase, an enzyme that adds 20-carbon prenylgroups to cysteine residues on the carboxy termini of rab proteins tomake them membrane-bound (60). Rab proteins are key to vesicletransport and organelle dynamics (61), and the gunmetal mutationresults in decreased prenylation and decreased membrane associationof rab27a (60). The ashen mouse, which recently became a model ofHPS based upon the finding of absent platelet dense bodies, codes forrab27a itself (62). Mutations in rab27a result in Griscelli syndrome(63), which falls within the HPS-CHS spectrum of disorders (64)(see below).

There also exist at least 11 Drosophila melanogaster models of HPSand related disorders. These flies have mutations at eye color loci andare members of the “granule group” of mutants (65). The 7 granulegroup genes isolated to date are identical to genes involved in lyso-somal trafficking in other organisms. For example, all four subunits ofAP3 have a Drosophila mutant model. The garnet gene produces a �subunit of AP3 (52, 66), while ruby codes for a �3 subunit (67), car-mine codes for �3 (68), and orange for �3 (69). Three other genes of thegranule group encode Drosophila homologues of the yeast VPS(Vesicle Proteins for Sorting) genes. The light fly has mutations inVPS41, whose protein product interacts with the � subunit of AP3 (70).Deep orange and carnation have mutations in VPS18 and VPS33,respectively (71, 72). The products of these genes interact in a largecomplex, and mutations in deep orange cause accumulation of multi-vesicular bodies (72).

In yeast, the VPS18 and VPS33 proteins form a multisubunit com-plex with VPS11 and VPS16 (73). Moreover, VPS33 interacts withthe t-SNARE VAM3, indicating that these proteins might act togetherto direct docking and/or fusion of transport intermediates with the yeastvacuole (74). VAM3 is transported via an AP3-dependent sorting

pathway (75). There are more than 40 different yeast VPS genes whichinfluence trafficking of proteins destined for the lysosome-like vacuole(76). Mutants in any of these genes could be primitive models forhuman HPS.

Chediak-Higashi Syndrome

CHS (1, 2, 77, 78) was first described by Beguez-Cesar in 1943 (79),but acquired its eponym a decade later from Moises Chediak (80) andOtotaka Higashi (81). It is characterized by an infectious diathesis,giant intracellular granules, variable degrees of oculocutaneousalbinism, and a platelet storage pool deficiency. The infections aregenerally fatal in the first decade of life, but CHS patients can alsosuccumb to a chronic lymphohistiocytic infiltration known as the accel-erated phase during the second or third decades of life.

Clinical and Laboratory Findings

As for HPS, the clinical findings in CHS are variable but includeseveral characteristic manifestations. Heterozygotes for CHS are al-ways completely normal.

Oculocutaneous albinism. Hair color in CHS patients can be blondto light brown, but the truly characteristic color is metallic silver. Gianthypomelanized melanosomes in CHS melanocytes cannot be trans-ferred to surrounding keratinocytes (82), resulting in skin color thatvaries from white to gray. Sun-exposed areas may pigment, and neviand lentigines do arise with some frequency. The eyes of CHS patientshave reduced pigment in the retina, iris, choroid and ciliary epithelium.On electron microscopy, giant aggregates of melanin appear in themelanosomes of these tissues (83). Irides can be gray, blue, or evenbrown. Patients can also have nystagmus, photophobia, and reduced

Fig. 2 Distribution of �3A, LAMP-3, TRP-1 (tyro-sinase-related protein-1) and tyrosinase in normal andHPS-2 melanocytes, fixed in formaldehyde and stainedwith monoclonal antibodies. A. In both normal andHPS-2 melanocytes, LAMP-3 (green) showed a similarmelanosomal distribution pattern. In normal melano-cytes, �3A (red) was expressed throughout the cell, par-tially colocalizing with LAMP-3. In HPS-2 melano-cytes, the �3A signal was dramatically decreased, asexpected. B. In normal melanocytes, TRP-1 (green) andtyrosinase (red) exhibited a similar distribution through-out the cells. In HPS-2 cells, TRP-1 had a normal distri-bution, but tyrosinase was localized mainly in theperinuclear area and was absent from the cell periphery.This finding indicates that tyrosinase, but not TRP-1,is carried by an AP3-dependent pathway

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Page 6: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

238

Thromb Haemost 2001; 86: 233–45

visual acuity. Electroretinograms and visual evoked potentials showprogressive abnormalities (84).

Bleeding. CHS patients have the same diathesis toward easy bruis-ability, mucosal bleeding, and epistaxis that HPS patients exhibit, butsome CHS patients have reduced or irregular platelet dense bodiesrather than total absence of these intracellular organelles. CHS plateletsdo have low levels of ATP, ADP, calcium, and serotonin, and secreteddense body constituents (i. e., nucleotides and calcium) are lacking aswell (85). Aggregation studies performed on CHS patients (85, 86)reveal an absent or minimal second wave of aggregation followingstimulation with epinephrine or other agonists. Alpha granules havebeen normal in number and morphology in CHS patients.

Infections. Children with CHS suffer from recurrent infections be-ginning in infancy. Typical organs affected include the skin andrespiratory systems. Periorbital cellulitis, otitis media, pneumonias,pyoderma, abscesses, sinus infections, and dental caries are prominenttypes of infections, and Staphylococcus aureus and �-hemolyticStreptococcus are the primary organisms, although gram negative orga-nisms, Candida, and Aspergillus are also important (87). Response toantibiotics is slower than expected.

Although the bone marrow appears normal to hypercellular in CHS,neutropenia is common. CHS neutrophils, as well as lymphocytes,eosinophils, and platelets, contain giant granules, up to 4 �m in diam-eter (Fig. 3). The granules contain acid hydrolases, myeloperoxidase,and other proteins necessary for bactericidal activity (88), and existalongside normal-sized primary lysosomes (89). The giant granulesimpair leucocyte migration, perhaps by inhibiting cell deformability.Phagocytosis appears normal in CHS cells (90), with intracellularbactericidal acivity delayed but improving to near normal over time.This suggests that the primary defect in bacterial killing resides in theearly stage of the process, while later functions remain intact. In CHSpatients, natural killer (NK) cells are present in normal or slightlyelevated numbers, but their function is impaired (91), contributing tothe susceptibility to defective cellular immunity and to development ofthe accelerated phase.

A number of parameters are normal in CHS, including immuno-globulins, antibody production, delayed hypersensitivity, reticuloendo-thelial clearance, monocyte number and phagocytic function (90),levels of complement (92), suppressor cells, response to interferon,target cell recognition, and antibody-dependent, cell-mediated cyto-lysis (93, 94).

Accelerated phase. Approximately 85% of CHS patients experiencethe accelerated phase sometime between a few months and severalyears of age. The accelerated phase involves fever, anemia, neutro-penia, and occasionally thrombocytopenia, hepatosplenomegaly,lymphadenopathy, and jaundice (87). Liver function tests are elevated,cellular immunity is decreased, and pancytopenia presents due tosplenomegaly and hemolysis. A coagulopathy may also develop due toliver dysfunction and thrombocytopenia (87). The accelerated phaserepresents a reactive process with perivascular lymphohistiocytic in-filtrates that are benign histologically (87, 95, 96) and resemble infec-tious mononucleosis (95). Lack of NK cell function has been theorizedto allow the accelerated phase to become fulminant.

Neurologic manifestations. CHS patients can experience a peri-pheral or cranial neuropathy, autonomic dysfunction, weakness andsensory deficits, loss of deep tendon reflexes, clumsiness, a wide-basedgait, seizures, abnormal electromyograms, and decreased motor nerveconduction velocites or spinocerebellar degeneration (77, 97). Onpathological examination of four CHS patients, lymphohistiocytic infil-trates were evident throughout the nervous system (98). Cytoplasmic

inclusions, resembling lysosomes or large, irregular lipofuscin granu-les, were present in astrocytes, choroid plexus epithelial cells, Schwanncells, and satellite cells of the dorsal spinal ganglia.

Diagnosis

The hallmark of CHS is the presence of giant peroxidase-positivegranules in polymorphonuclear leucocytes, megakaryocytes, neurons,conjunctival fibroblasts, and cultured lymphoblasts (1, 2, 77, 78). Otherclinical characteristics, such as mild oculocutaneous albinism, silveryhair, bleeding problems, provide clues to seek a definitive diagnosis.Prenatal diagnosis has been achieved by examination for large, acidphosphatase-positive lysosomes in amniocytes and chorionic villuscells (99). The molecular diagnosis of CHS remains difficult because ofthe large size of the LYST gene and because several different mutationshave been identified. Molecular diagnosis is not commercially available.

Treatment

The only curative therapy for CHS is bone marrow transplantation,which has also reversed the leucocyte defect in the mouse model ofCHS, beige (100). Seven of 10 children from two separate institutionswho received related donor bone marrows had a successful transplanta-tion (101). None of the children had recurrence of the acceleratedphase, and all had improved NK cell activity. However, this therapydoes not prevent the progressive neuropathy of CHS.

Other therapeutic modalities for CHS are symptomatic (77). Child-hood immunizations are well-tolerated and should be provided (87).Antibiotics will treat the infections, although with a slow response.Aspirin-containing products should be avoided. Desmopressin and�-amino-caproic acid can be effective for treatment and prophylaxis,while platelet transfusions may be needed for major procedures orbleeds. Corticosteroids, chemotherapeutics, intravenous immunoglobu-lin, and splenectomy may occasionally induce a temporary remission ofthe accelerated phase. Ascorbic acid treatment has not proven clinicallyeffective in preventing either infections or the accelerated phase in CHSpatients (102, 103).

Genetics

CHS is an extremely rare, autosomal recessively inherited disorderthat has been found in countries all over the world. Although more than200 cases have been reported over time (1, 77), many fewer are likelyto be alive today.

Unlike HPS, CHS does not display locus heterogeneity. However, itremains possible that some CHS patients, particularly clinical variants,might have their disease due to mutations in genes not yet discovered.The clinical variability observed in the animal models of CHS, i. e., theAleutian mink, cattle, cats, killer whales, and rats (77), supportsthe possibility of locus heterogeneity. Presently, the one gene provento cause CHS is LYST, which was discovered with the assistance ofextensive investigations into the best animal model of CHS, the beigemouse (77).

The LYST gene. The lyst gene in the beige mouse, on chromo-some 13, is homologous to the human LYST (for lysosome traffickingregulator), on chromosome 1q42.1-42.2 (104-106). LYST (107-109) has a coding region of 11,403 bp and is expressed in all cell types.The 13449-bp mRNA (GenBank accession #4502838) is present inlow levels and is difficult to detect by Northern blot analysis. A large,11.4 kb transcript appears to be responsible for gene function, while the

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Page 7: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

239

Huizing: Hermansky-Pudlak and Chediak-Higashi Syndromes

function of a smaller, 5.8 kb transcript derived from the 5’ terminus ofLYST is unknown (107, 108). The human LYST gene has homologues inall mammalian species (109). The human and mouse proteins are 82%identical and 88% homologous. High degrees of identity are also seenbetween the human and the rat and cow CHS genes.

To date, LYST mutations have been identified in 13 CHS patients(107-110). All human mutations, as well as those in the mouse (111)and rat (112) beige models, have been nonsense or frameshift mutationswith premature termination. There is no obvious “hot spot”, and nocorrelation between severity of disease and residual length of the trun-cated LYST protein.

The LYST protein. The LYST gene encodes a large cytosolic proteinwith 3801 amino acids and a molecular mass of 429 kDa (109). TheLYST protein contains several phosphorylation sites and myristylationsites, indicating a capacity for regulation and interaction with otherproteins or membranes. In addition, the amino-terminus of the LYSTprotein contains a series of hydrophobic helices resembling HEAT andARM domains (109). HEAT repeat proteins are involved in vesicletransport, and ARM domains are thought to mediate membrane asso-ciations (113). The N-terminus of LYST also shows low homologyto stathmin, a phosphoprotein involved in regulation of microtubulepolymerization (109, 114).

The central region of LYST contains two leucine zippers and an8-amino-acid domain homologous to a sequence in the HPS1 protein(27). HPS1 and LYST may interact through this homologous domain,possibly via another linker protein (115, 116).

The carboxy-terminus of LYST contains seven consecutive WD40motifs that form beta sheets (109). WD40 motifs are considered tomediate protein-protein interactions (117). Another motif in the C-terminus, conserved among LYST homologues in other species, is a345-amino-acid domain called BEACH, for “beige” and “CHS” (109).The BEACH domain contains a consensus “WIDL” amino acid stretchas well as other conserved amino acids. The combination of BEACH

and WD-40 domains are found in several mammalian proteins includ-ing FAN, CDC4L, and neurobeachin (78, 118). FAN links TNF� toneutral sphingomyelinase (119), and neurobeachin anchors proteinkinase A in the trafficking of neuronal post-Golgi membranes (118).The function of CDC4L (120) is unknown. The exact function of LYSTcannot be predicted from homology searches, but these structuralstudies do suggest that it functions in vesicule trafficking.

CHS Cell Biology

Mutations in LYST result in giant granular inclusions (78, 88) inmany different cell types, including platelets (Fig. 3), which are alsodeficient in dense bodies (121). To understand why giant granules formand some normal vesicles do not, several aspects of CHS cell biologymust be addressed.

Endosomal transport in CHS. Secretory lysosomes are synthesizednormally in CHS cells (122), and both early (recycling) and late (multi-vesicular) endosomes display normal morphology and accessibility toendocytic tracers (123). However, some downstream functions areimpaired. For example, peptide loading and antigen presentation, partsof the immunologic process, are delayed in CHS B cells (123), andmelanosome, secretory granule, and lysosome release are functionallyimpaired. Other endocytic functions, such as protein degradation andrecycling of transferrin receptors, are not affected.

The characteristic macrolysosomes of CHS cells are acidic, acquireendocytic tracers such as bovine serum albumin-conjugated colloidalgold with normal kinetics (123, 124), and occasionally fuse with theplasma membrane. The lysosomal membrane markers HLA-DM,LAMP-1, LAMP-2, CD63, CD82, and �-hexosaminidase all accumu-late in the macrolysosomes, as expected, but these proteins also accu-mulate at the cell surface in CHS cells (123, 125). The mannose-6-phosphate receptor (MPR), normally excluded from lysosomes, is veryabundant in the macrolysosomes in CHS. In contrast, late endosomes

Fig. 3 Giant granules (arrows) in CHS cells. A. Neu-trophil with several giant lysosomes. (�3300) B. Lym-phocyte with a single giant granule. (�3300) C. Eosino-phil with several large granules of different electrondensity. (�2600). D. Platelet showing many normalsized lysosomes and one giant lysosome. (�5500).All micrographs courtesy of Dr. James White, Univer-sity of Minnesota

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Page 8: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

240

Thromb Haemost 2001; 86: 233–45

Fig. 4 Hypothetical model of LYST protein function in protein sorting and vesicle formation and trafficking. A. Schematic drawing of the motifs found in theLYST protein. B. Late Golgi processes. Proteins destined to be transported to lysosomes bind to receptor molecules in the trans-Golgi membrane (1). This resultsin activation of the “VPS15 like” C-terminal portion of LYST (2). VPS15 activation provides a binding site for the VPS34-like PI3K (3),which then translocatesto the membrane (4). Since the PI3K can be replaced by another specific PI-kinases in this LYST-mediated process, it is termed PI(x)K. Inset: PI(x)K generatesPtdIns(x)Pn (phosphatidylinositol [x]-phosphate) from PtdIns and free phosphate. PtdIns(x)Pn is a source for the supply of DAG (diacylglycerol) to the Golgi com-plex. DAG is formed by phospholipase C, and activates PKC. C. Early vesicle formation. The C-terminus of LYST activates the specific local phosphorylation ofmembrane phospholipids, resulting in recruitment of coat proteins (5) and specific guanine nucleotide exchange factors (GEF) required for vesicle formation andtransport. The GEFs stimulate activation of members of the rab family of small GTPases (6). Meanwhile, the N-terminal portion of LYST connects to microtubu-le structures (7). D. Early vesicle movement. The N-terminal portion of LYST “pulls” the vesicle out of the late Golgi membrane towards the microtubule struc-tures (8). Once the vesicle is formed and starts moving along the microtubule, the LYST protein is released from the vesicle (9), probably recycling to the late Golgi for a new round of vesicle formation

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Page 9: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

241

Huizing: Hermansky-Pudlak and Chediak-Higashi Syndromes

contain a paucity of the normally abundant membrane markers HLA-DR,CD63, and CD82 (116).

Faigle et al. (116) concluded that defective lysosomal fusion withthe plasma membrane is not the primary defect in CHS cells. Further-more, the lack of T-cell and NK cell toxicity in CHS occurs becausecytolytic enzymes are missorted and thus absent from the granules,which release these enzymes upon fusion with the plasma membrane.Finally, the giant granules of CHS result from endosomal missorting ofproteins involved in the regulation of lysosomal homeostasis.

Association of LYST with microtubules. The macrolysosomes ofCHS cells are located in the perinuclear region, consistent with a defectin the microtubular network. Furthermore, the LYST protein hashomology to the microtubule polymerization protein, stathmin (119, 114),although CHS cells have normal numbers and distribution of micro-tubules themselves (126-128). We propose that the N-terminus ofLYST interacts with microtubules, but truncation of the C-terminus ofthe LYST protein, which occurs in all human LYST mutations, causesmalfunctioning of the microtubular moving system. In this case, themicrotubule-associated portion of LYST will remain present in amicrotubular distribution.

PKC activity and granule formation. Evidence indicates a correla-tion between decreased protein kinase C (PKC) activity and increasedlysosome size. Specific PKC inhibitors such as chelerythrin and cal-phostin C induce giant granule formation in normal fibroblasts (129),and inhibition of calpain, which inactivates PKC, prevents giant granuleformation in beige fibroblasts (129). These findings are important be-cause PKC activity is down-regulated in CHS cells (129-131). Moreover,decreased PKC activity in polymorphonuclear leucocytes (PMNs) andnatural killer (NK) cells from beige mice appears responsible for theimpaired immune functions in CHS. The down-regulation of PKC ac-tivity in CHS cells can be corrected by calpain inhibition (129-130),which also corrects the NK cell dysfunction of beige mice (131).

Phosphoinositol kinases. Phosphoinositol-3 kinase (PI3K) plays animportant role in the regulation of protein sorting (132). The localizedgeneration of PI-lipids by PI kinases (PI[x]K) acts to target proteinscritical for cellular trafficking, such as FYVE proteins (133, 134) tothe appropriate location on a membrane that will become a vesicle.PI-lipids also relocate specific guanine nucleotide exchange factors tothe vicinity of new vesicle formation. These exchange factors areneeded to activate members of the rab family of small GTPases whichfacilitate the docking of vesicles with target membranes. Inhibition ofPI3K with wortmannin impairs transport of membrane proteins fromendosomes to lysosomes (135), and causes accumulation of mannose-phosphate receptors in swollen lysosomal compartments of normalfibroblasts (136), an effect similar to that seen in CHS cells.

Structural characteristics of the LYST protein. By virtue of itsHEAT repeats, ARM repeats, and WD40 motifs, the LYST proteinresembles a p150 yeast serine/threonine protein kinase, VPS15 (109).The VPS15 yeast mutant presents as a defect in sorting to the vacuole(137), causing secretion of vacuolar hydrolases. VPS15 regulatesVPS34, a PI3 kinase, and in yeast and mammalian cells, VPS34/PI3Kis involved in endosomal transport and sorting into late endosomesand lysosomes (138, 139). The human LYST sequence does not containa bona fide kinase domain (140), but its carboxy terminus does resembleVPS15.

The Function and Dysfunction of LYST

We hypothesize that the LYST protein acts as an intermediate thataffects lipid-related protein trafficking. In this model, which is based

upon the various domains found in LYST (Fig. 4A), the C-terminus fun-ctions at the TGN to sort specific proteins pertinent for endosomes orlysosomes (Fig. 4B). The protein sorting is supported by lipid phos-phorylation carried out by a VPS34-like PI kinase activated by theVPS15-like domain of the C-terminal region of LYST. The PI kinasestimulates PKC activity indirectly, i. e., via DAG (Fig. 4 inset). Phos-phorylation of specific phospholipids in the TGN results in localrecruitment of coat proteins required for vesicle formation and trans-port. The localized generation of PI lipids by PI(x)K also attractsexchange factors which activate members of the rab family of smallGTPases (Fig. 4C). The GTP/GDP exchange factors interact with theleucine zippers and the HPS1 homology domain of the central part ofthe LYST protein. Rab GTP hydrolysis energizes vesicle formation orvesicle attachment to LYST.

Meanwhile, the N-terminal portion of LYST binds to microtubulesvia its stathmin-like domain (Fig. 4C). Once vesicles form, they contin-ue to be transported peripherally along the microtubules, but the verylarge and sterically hindering LYST protein is released for recyclingwell before the dendrite is reached (Fig. 4D).

When LYST is mutated, PI(x)K is not activated, and decreased DAGproduction leads to down regulation of PKC. Pigment forming vesiclesand dense bodies fail to form and do not move to the periphery.

The model is supported by the finding of a microtubule associationof LYST, by the down regulation of PKC activity, and by the knownstructural domains of LYST. There is also evidence that rabs play arole in the process. Antisense expression of rab7 or rab9 in HeLa cellsresults in the formation of large vacuoles, resembling those of CHSfibroblasts (141, 142), and rab7 interacts with VPS34 (143). Finally,the large size of LYST makes it a good candidate to bind to both theTGN and microtubules.

The model does not explicitly explain the formation of giant vesiclesin CHS cells. It may be that, absent the controlled, directed formationand peripheral movement of nascent vesicles, proteins accumulatewithin the TGN until a critical, massive size is reached, whereupon agiant vesicle breaks away. Its size would preclude movement from theperinuclear region. Alternatively, the giant vesicles may representendosomes formed from the plasma membrane which were intendedto fuse with LYST-dependent vesicles derived from the TGN. In theabsence of LYST, vital proteins, crucial for maintaining the size andcourse of the fused vesicles, never reach the endosomes, resulting intheir growth to giant granules.

Where Does HPS Stop and CHS Begin?

Studies of the clinical spectrum of HPS and the mouse models ofcombined hypopigmentation and storage pool deficiency provide in-sight into the relationship between HPS and CHS. HPS-2 disease,which clearly fits into the category of HPS based upon absence of pla-telet dense bodies and other clinical findings, manifests with neutro-penia and a diathesis toward infections. The ashen mouse, now con-sidered a model for HPS because of the recent finding of absent plate-let dense bodies (62), remains a model also for Griscelli syndrome, adisorder of pigment dilution and silvery hair plus other clinical findings(64). Griscelli syndrome patients who manifest immunological defectsand hemophagocytic syndrome, an uncontrolled T-lymphocyte andmacrophage activation syndrome, have their disease due to deficiencyof rab27a (63), as for ashen (62). Griscelli syndrome patients whoexhibit neurologic impairment instead, have mutations in myosin 5a(144), the gene affected in the mouse dilute (145). (Leaden resembleashen and dilute but its molecular defect has not been resolved.)

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Page 10: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

242

Thromb Haemost 2001; 86: 233–45

Clearly, the neurologic and immunological symptoms of thesepatients and mice bring them closer to CHS, and confirm the existenceof a continuum of disease. The concept of a gradation of pathologyapplies not only to clinical manifestations, but to cell biological pheno-mena as well. HPS defects appear more involved with vesicle forma-tion, while CHS appears due to vesicle transport defects. The Griscellisyndromes fall in between, with rab27a defects (vesicle formation/fusion)resembling HPS and myosin 5a defects (microtubular transport) resem-bling CHS.

The complete spectrum of disease between HPS and CHS requiressome shading. This will be accomplished by the molecular descriptionof HPS patients whose mutations have not yet been defined, and by theisolation of genes responsible for clinical variants. These genes arebeing sought among those responsible for animal models of theHPS/CHS spectrum, as well as genes involved in the pathways ofvesicle formation and trafficking. From our model of the mechanismof LYST action, we predict that specific rabs, exchange factors, andvesicle docking and fusion proteins are involved. As the gamut ofhuman disease becomes defined, so will the pathways for the formationof vesicles of lysosomal lineage.

Acknowledgement

Dr. Yair Anikster is a Howard Hughes Medical Institute Physician Post-doctoral Fellow.

References

1. Witkop CJ, Quevedo WC, Fitzpatrick TB, King RA. Albinism. In: TheMetabolic Basis of Inherited Disease. Scriver CR, Beaudet AL, Sly WS,Valle DL, eds. 6th ed., vol. 2. New York: McGraw-Hill 1989; 2905-47.

2. King RA, Hearing VJ, Creel DJ, Oetting WS. Albinism. In: The Meta-bolic and Molecular Bases of Inherited Disease. Scriver CR, Beaudet AL,Sly WS, Valle DL, eds. 7th ed., vol. 3. New York: McGraw-Hill 1995;4353-92.

3. Shotelersuk V, Gahl WA. Hermansky-Pudlak syndrome: Models for intra-cellular vesicle formation. Mol Genet Metab 1998; 65: 85-96.

4. Huizing M, Anikster Y, Gahl WA. Hermansky-Pudlak syndrome and re-lated disorders of organelle formation. Traffic 2000; 1: 823-35.

5. Spritz RA. Hermansky-Pudlak syndrome and pale ear: Melanosome-making for the mellenium. Pigment Cell Res 2000; 13: 15-20.

6. Spritz RA. Chediak-Higashi syndrome. In: Primary ImmunodeficiencyDiseases: A Molecular and Genetic Approach. Ochs HD, Smith CIE, PuckJM, eds. New York: Oxford University Press 1999; 389-96.

7. Hermansky F, Pudlak P. Albinism associated with hemorrhagic diathesisand unusual pigmented reticular cells in the bone marrow: Report of twocases with histochemical studies. Blood 1959; 14: 162-9.

8. Gahl WA, Brantly M, Kaiser-Kupfer MI, Iwata F, Hazelwood S, Shoteler-suk V, Duffy LF, Kuehl EM, Troendle J, Bernardini I. Genetic defects andclinical characteristics of patients with a form of oculocutaneous albinism(Hermansky-Pudlak syndrome). N Engl J Med 1998; 338: 1258-64.

9. Toro J, Turner M, Gahl WA. Dermatologic manifestations of Hermansky-Pudlak syndrome in patients with and without a 16-base pair duplication inthe HPS1 gene. Arch Dermatol 1999; 135: 774-80.

10. Simon JW, Adams RJ, Calhoun JH, Shapiro SS, Ingerman CM. Ophthal-mic manifestations of the Hermansky-Pudlak syndrome (oculocutaneousalbinism and hemorrhagic diathesis). Am J Ophthalmol 1982; 93: 71-7.

11. Summers CG, Knobloch WH, Witkop CJ, King RA. Hermansky-PudlakSyndrome: Ophthalmic findings. Ophthalmol 1988; 95: 545-54.

12. Iwata F, Reed GF, Caruso RC, Kuehl EM, Gahl WA, Kaiser-Kupfer MI.Correlation of visual acuity and ocular pigmentation with the 16-bp dupli-cation in the HPS-1 gene of Hermansky-Pudlak syndrome, a form of albi-nism. Ophthalmology 2000; 107: 783-9.

13. McNicol A, Israels SJ. Platelet dense granules: Structure, function andimplications for haemostasis. Thromb Res 1999; 95: 1-18.

14. Witkop CJ Jr, Bowie EJ, Krumwiede MD, Swanson JL, Plumhoff EA,White JG. Synergistic effect of storage pool deficient platelets and lowplasma von Willebrand factor on the severity of the hemorrhagic diathesisin Hermansky-Pudlak Syndrome. Am J Hematol 1993; 44: 256-9.

15. McKeown LP, Hansmann KE, Wilson O, Gahl WA, Gralnick HR, Rosen-feld KE, Rosenfeld SJ, Horne MK, Rick ME. Platelet von Willebrand fac-tor in Hermansky-Pudlak Syndrome. Am J Hematol 1998; 59: 115-20.

16. Shanahan F, Randolph L, King R, Oseas R, Brogan M, Witkop C, RotterJ, Targan. Hermansky-Pudlak syndrome: An immunologic assessment of15 cases. Am J Med 1988; 85: 823-8.

17. Garay SM, Gardella JE, Fazzini EP, Goldring RM. Hermansky-PudlakSyndrome: Pulmonary manifestations of a ceroid storage disorder. Am JMed 1979; 66: 737-47.

18. Brantly M, Avila NA, Shotelersuk V, Lucero C, Huizing M, Gahl WA.Pulmonary function and high-resolution CT findings in patients with aninherited form of pulmonary fibrosis, Hermansky-Pudlak syndrome, dueto mutations in HPS-1. Chest 2000; 117: 129-36.

19. Schinella RA, Greco MA, Cobert BL, Denmark LW, Cox RP. Hermansky-Pudlak syndrome with granulomatous colitis. Ann Intern Med 1980; 92:20-3.

20. Mahadeo R, Markowitz J, Fisher S, Daum F. Hermansky-Pudlak syn-drome with granulomatous colitis in children. J Pediatr 1991; 118: 904-6.

21. Dell’Angelica EC, Shotelersuk V, Aguilar RC, Gahl WA, Bonifacino JS.Altered trafficking of lysosomal proteins in Hermansky-Pudlak syndromedue to mutations in the �3A subunit of the AP-3 adaptor. Mol Cell 1999;3: 11-21.

22. Shotelersuk V, Dell’Angelica EC, Hartnell L, Bonifacino JS, Gahl WA.A new variant of Hermansky-Pudlak syndrome due to mutations in a generesponsible for vesicle formation. Am J Med 2000; 108: 423-7.

23. Hazelwood S, Shotelersuk V, Wildenberg SC, Chen D, Iwata F, Kaiser-Kupfer MI, White JG, King RA, Gahl WA. Evidence for locus hetero-geneity in Puerto Ricans with Hermansky-Pudlak syndrome. Am J HumGenet 1997; 61: 1088-94.

24. Witkop CJ, Krumwiede M, Sedano H, White JG. Reliability of absentplatelet dense bodies as a diagnostic criterion for Hermansky-Pudlaksyndrome. Am J Hematol 1987; 26: 305-11.

25. Van Dorp DB, Wijermans PW, Meire F, Vrensen G. The Hermansky-Pudlak syndrome: Variable reaction to 1-desamino-8D-arginine vasopres-sin for correction of the bleeding time. Ophthal Paediatr Genet 1990;11237-44.

26. Witkop CJ, Babcock MN, Rao GHR, Gaudier F, Summers CG, ShanahanF, Harmon KR, Townsend DW, Sedano HO, King RA, Cal SX, White JG.Albinism and Hermansky-Pudlak syndrome in Puerto Rico. Bol Asoc MedP Rico-Agosto 1990; 82: 333-9.

27. Oh J, Bailin T, Fukai K, Feng GH, Ho L, Mao J-i, Frenk E, Tamura N,Spritz RA. Positional cloning of a gene for Hermansky-Pudlak syndrome,a disorder of cytoplasmic organelles. Nat Genet 1996; 14: 300-6.

28. Oh J, Ho L, Ala-Mello S, Amato D, Armstrong L, Bellucci S, Caraku-shansky G, Ellis JP, Fong C-T, Green JS, Heon E, Legius E, Levin AV,Nieuwenhuis HK, Pinckers A, Tamura N, Whiteford ML, Yamasaki H,Spritz RA. Mutation analysis of patients with Hermansky-Pudlak syn-drome: A frameshift hot spot in the HPS gene and apparent locus hetero-geneity. Am J Hum Genet 1998; 62: 593-8.

29. Shotelersuk V, Hazelwood S, Larson D, Iwata F, Kaiser-Kupfer MI,Kuehl E, Bernardini I, Gahl WA. Three new mutations in a gene causingHermansky-Pudlak syndrome: Clinical correlations. Mol Gen Metab1998; 64: 99-107.

30. Oetting WS, King RA. Molecular basis of albinism: Mutations and poly-morphisms of pigmentation genes associated with albinism. Hum Mutat1999; 13: 99-115.

31. Swank RT, Novak EK, McGarry MP, Rusiniak ME, Feng L. Mousemodels of Hermansky Pudlak syndrome: A review. Pigment Cell Res1998; 11: 60-80.

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Page 11: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

243

Huizing: Hermansky-Pudlak and Chediak-Higashi Syndromes

32. Wildenberg SC, Oetting WS, Almodovar C, Krumwiede M, White JG,King RA. A gene causing Hermansky-Pudlak syndrome in a Puerto Ricanpopulation maps to chromosome 10q2. Am J Hum Genet 1995; 57: 755-65.

33. Bailin T, Oh J, Feng GH, Fukai K, Spritz RA. Organization and nucleo-tide sequence of the human Hermansky-Pudlak syndrome (HPS) gene.J Invest Dermatol 1997; 108: 923-7.

34. Wildenberg SC, Fryer JP, Gardner JM, Oetting WS, Brilliant MH, KingRA. Identification of a novel transcript produced by the gene responsiblefor the Hermansky-Pudlak syndrome in Puerto Rico. J Invest Dermatol1998; 110: 777-81.

35. Huizing M, Anikster Y, Gahl WA. Characterization of a partial pseudo-gene homologous to the Hermansky-Pudlak syndrome gene HPS-1; rele-vance for mutation detection. Hum Genet 2000; 106: 370-3.

36. Feng GH, Bailin T, Oh J, Spritz RA. Mouse pale ear (ep) is homologous tohuman Hermansky-Pudlak syndrome and contains a rare ‘AT-AC’ intron.Hum Mol Genet 1997; 6: 793-9.

37. Gardner JM, Wildenberg SC, Keiper NM, Novak EK, Rusiniak ME,Swank RT, Puri N, Finger JN, Hagiwara N, Lehman AL, Gales TL, BayerME, King RA, Brilliant MH. The mouse pale ear (ep) mutation is thehomologue of human Hermansky-Pudlak syndrome. Proc Natl Acad SciUSA 1997; 94: 9238-43.

38. Dell’Angelica EC, Ooi CE, Bonifacino JS. �3A-adaptin, a subunit of theadaptor-like complex AP-3. J Biol Chem 1997; 272: 15078-84.

39. Schekman R, Orci L. Coat proteins and vesicle budding. Science (WashDC) 1996; 271: 1526-33.

40. Feng L, Seymour AB, Jiang S, To A, Peden AA, Novak EK, Zhen L,Rusiniak ME, Eicher EM, Robinson MS, Gorin MB, Swank RT. The �3Asubunit gene (Ap3b1) of the AP-3 adaptor complex is altered in themouse hypopigmentation mutant pearl, a model for Hermansky-Pudlaksyndrome and night blindness. Hum Mol Genet 1999; 8: 323-30.

41. Israels SJ, McMillan EM, Robertson C, Singhory S, McNicol A. The lyso-somal granule membrane protein, LAMP-2, is also present in plateletdense granule membranes. Thromb Haemost 1996; 75: 623-9.

42. Oh J, Liu ZX, Feng GH, Raposo G, Spritz RA. The Hermansky-Pudlaksyndrome (HPS) protein is part of a high molecular weight complex in-volved in biogenesis of early melanosomes. Hum Mol Genet. 2000; 9:375-85.

43. Barbosa MDFS, Nguyen QA, Tchemev VT, Ashley JA, Detter JC,Blaydes SM, Brandt SJ, Chotai D, Hodgman C, Solari RCE, Lovett M,Kingsmore SF. Identification of the homologous beige and Chediak-Higashi syndrome genes. Nature 1996; 382: 262-5.

44. Jimbow K, Park JS, Kato F, Hirosaki K, Toyofuku K, Hua C, YamashitaT. Assembly, target-signaling and intracellular transport of tyrosinasegene family proteins in the initial stage of melanosome biogenesis.Pigment Cell Res 2000; 13: 222-9.

45. Johnson KF, Kornfeld S. A His-Leu-Leu sequence near the carboxylterminus of the cytoplasmic domain of the cation-dependent mannose6-phosphate receptor is necessary for the lysosomal enzyme sorting func-tion. J Biol Chem 1992; 267: 17110-5.

46. Dell’Angelica E, Aguilar R, Wolins N, Hazelwood S, Gahl WA, BonifacinoJS. Molecular characterization of the protein encoded by the Hermansky-Pudlak Syndrome type 1 gene. J Biol Chem 2000; 275: 1300-6.

47. Boissy RE, Zhao Y. The role of the Hermansky-Pudlak gene product inintracellular trafficking of melanogenic proteins. J Invest Dermatol 1999;112: 629 (Abstr 637).

48. Boissy RE, Zhao Y, Gahl WA. Altered protein localization in melanocytesfrom Hermansky-Pudlak syndrome: Support for the role of the HPS geneproduct in intracellular trafficking. Lab Invest 1998; 78: 1037-48.

49, Dell’Angelica EC, Klumperman J, Stoorvogel W, Bonifacino JS. Associa-tion of the AP-3 adaptor complex with clathrin. Science 1998; 280: 431-4.

50. Dell’Angelica EC, Ohno H, Ooi CE, Rabinovich E, Roche KW, BonifacinoJS. AP-3: an adaptor-like protein complex with ubiquitous expression.EMBO J 1997; 15: 917-28.

51. Simpson F, Bright NA, West MA, Newman LS, Darnell RB, RobinsonMS. A novel adaptor-related protein complex. J Cell Biol 1996; 133: 749-60.

52. Simpson F, Peden AA, Christopoulou L, Robinson MS. Characterizationof the adaptor-related protein complex, AP-3. J Cell Biol 1997; 137: 835-45.

53. Ohno H, Fournier MC, Poy G, Bonifacino JS. Structural determinants ofinteraction of tyrosine-based sorting signals with the adaptor mediumchains. J Biol Chem 1996; 271: 29009-15.

54. Honig S, Sandoval IV, von Figura K. A di-leucine-based motif in the cyto-plasmic tail of LIMP-II and tyrosine mediates selective binding of AP-3.EMBO J 1998; 17:1304-14.

55. Kantheti P, Qiao X, Diaz ME, Peden AA, Meyer GE, Carskadon SL, Kapf-hamer D, Sufalko D, Robinson MS, Noebels JL, Burmeister M.Mutation in AP-3 � in the mocha mouse links endosomal transport tostorage deficiency in platelets, melanosomes, and synaptic vesicles.Neuron 1998; 21: 111-22.

56. Caplan S, Dell’Angelica EC, Gahl WA, Bonifacino JS. Trafficking ofMHC class II molecules in human B-lymphoblasts deficient in the AP-3adaptor complex. Immunol Lett 2000; 72: 113-7.

57. Huizing M, Boissy RE, Gahl WA. Hermansky-Pudlak syndrome (HPS):A model for intracellular vesicle formation and trafficking. J Inherit MetabDis 2000 (Suppl. 1); 23: 284a (Abstr).

58. Huang L, Kuo Y-M, Gitschier J. The pallid gene encodes a novel, syntaxin13-interacting protein involved in platelet storage pool deficiency. NatGenet 1999; 23: 329-32.

59. Prekeris R, Klumperman J, Chen YA, Scheller RH. Syntaxin 13 mediatescycling of plasma membrane proteins via tubulovesicular recycling endo-somes. J Cell Biol 1998; 143: 957-71.

60. Detter JC, Zhang Q, Mules EH, Novak EK, Mishra VS, Li W, McMurtrieEB, Tchernev VT, Wallace MR, Seabra MC, Swank RT, Kingsmore SF.Rab geranylgeranyl transferase � mutation in the gunmetal mouse reducesRab27 prenylation and platelet synthesis. Proc Natl Acad Sci USA 2000;97: 4144-9.

61. Novick P, Zerial M. The diversity of Rab proteins in vesicle transport.Curr Opin Cell Biol 1997; 9: 496-504.

62. Wilson SM, Yip R, Swing DA, O’Sullivan TN, Zhang Y, Novak E, SwankRT, Russell LB, Copeland NG, Jenkins NA. A mutation in Rab27a causesthe vesicle transport defects observed in ashen mice. Proc Natl Acad SciUSA 2000; 97: 7933-8.

63. Menasche G, Pastural E, Feldmann J, Certain S, Ersoy F, Dupuis S, Wulf-fraat N, Bianchi D, Fischer A, Le Deist F, de Saint Basile G. Mutations inRAB27A cause Griscelli syndrome associated with haemophagocyticsyndrome. Nat Genet 2000; 25: 173-6.

64. Griscelli C, Durandy A, Guy-Grand D, Daguillard F, Herzog C, PrunicrasM. A syndrome associating partial albinism and immunodeficiency. Am JMed 1978; 65: 691-702.

65. Lloyd V, Ramaswami M, Kramer H. Not just pretty eyes: Drosophila eye-colour mutations and lysosomal delivery. Trends Cell Biol 1998; 8: 257-9.

66. Ooi CE, Moreira JE, Dell’Angelica EC, Poy G, Wassarman DA, BonifacinoJS. Altered expression of a novel adaptin leads to defective pigment gra-nule biogenesis in the Drosophila eye color mutant garnet. EMBO J 1997;16: 4508-18.

67. Kretzschmar D, Poeck B, Roth H, Ernst R, Keller A, Porsch M, Strauss R,Pflugfelder GO. Defective pigment granule biogenesis and aberrant be-havior caused by mutations in the Drosophila AP-3beta adaptin gene ruby.Genetics 2000; 155: 213-23.

68. Mullins C, Hartnell LM, Wassarman DA, Bonifacino JS. Defective ex-pression of the �3 subunit of the AP-3 adaptor complex in the Drosophilapigmentation mutant carmine. Mol Gen Genet 1999; 262: 401-12.

69. Mullins C, Hartnell LM, Wassarman DA, Bonifacino JS. Mutations insubunits of the AP-3 adaptor complex result in defective pigment granulebiogenesis in Drosophila melanogaster. Mol Biol Cell 1999; 10: 223a(Abstr).

70. Rehling P, Darsow T, Katzmann DJ, Emr SD. Formation of AP-3 transportintermediates requires VPS41 function. Nat Cell Biol 1999; 1: 346-53.

71. Shestopal SA, Makunin IV, Belyaeva ES, Ashburner M, Zhimulev IF.Molecular characterization of the deep orange (dor) gene of Drosophilamelanogaster. Mol Gen Genet 1997; 253: 642-8.

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Page 12: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

244

Thromb Haemost 2001; 86: 233–45

72. Sevrioukov EA, He JP, Moghtabi N, Sunio A, Kramer H. A role for thedeep orange and carnation eye color genes in lysosomal delivery inDrosophila. Mol Cell 1997; 4: 479-86.

73. Rieder SE, Emr SD. A novel RING finger protein complex essential for alate step in protein transport to the yeast vacuole. Mol Biol Cell 1997; 8:2307-27.

74. Dorsow T, Rieder SE, Emr SD. A multispecificity syntaxin homologue,Vam3p, essential for autophagic and biosynthetic protein transport to thevacuole. J Cell Biol 1997; 138: 517-29.

75. Dorsow T, Burd CG, Emr SD. Acidic di-leucine motif essential for AP3-dependent sorting and restriction of the functional specificity of theVam3p vacuolar t-SNARE. J Cell Biol 1998; 142: 913-22.

76. Lemmon SK, Traub LM. Sorting in the endosomal system in yeast andanimal cells. Curr Opin Cell Biol 2000; 12: 457-66.

77. Introne W, Boissy RE, Gahl WA. Clinical, molecular, and cell biologicalaspects of Chediak-Higashi syndrome. Mol Genet Metab 1999; 68:283-303.

78. McVey Ward D, Griffiths GM, Stinchcombe JC, Kaplan J. Analysis of thelysosomal storage disease Chediak-Higashi syndrome. Traffic 2000; 1:816-22.

79. Beguez-Cesar AB. Neutropenia cronica maligna familiar con granulacio-nes atipicas de los leucocitos. Bol Soc Cubana Pediatr 1943; 15: 900-22.

80. Chediak M. Nouvelle anomalie leukocytaire de caractere constitutionnel etfamiliel. Rev Hematol 1952; 7: 362-7.

81. Higashi O. Congenital gigantism of peroxidase granules. Tohoku J ExpMed 1954; 59: 315-32.

82. Zhao H, Boissy YL, Abdel-Malek Z, King RA, Nordlund JJ, Boissy RE.On the analysis of the pathophysiology of Chediak-Higashi syndrome.Defects expressed by cultured melanocytes. Lab Invest 1994; 71: 25-34.

83. Valenzuela R, Morningstar WA. The ocular pigmentary disturbance ofhuman Chediak-Higashi syndrome. A comparative light- and electron-microscopic study and review of the literature. Am J Clin Pathol 1981; 75:591-6.

84. BenEzra D, Mengistu F, Cividalli G, Weizman Z, Merin S, Auerbach E.Chediak-Higashi Syndrome: Ocular findings. J Pediatr Ophthalmol Stra-bis 1980; 17: 68-74.

85. Apitz-Castro R, Cruz MR, Ledezma E, Merino F, Ramirez-Duque P,Dangelmeier C, Holmsen H. The storage pool deficiency in platelets fromhumans with the Chediak-Higashi syndrome: study of six patients. Br JHaematol 1985; 59: 471-83.

86. Buchanan GR, Handin RI. Platelet function in the Chediak-Higashisyndrome. Blood 1976; 47: 941-8.

87. Blume R.S, Wolff SM. The Chediak-Higashi syndrome: studies in fourpatients and a review of the literature. Medicine 1972; 51: 247-80.

88. Kjeldsen L, Calafat J, Borregaard N. Giant granules of neutrophils inChediak-Higashi syndrome are derived from azurophil granules but notfrom specific and gelatinase granules. J Leukocyte Biol 1998; 64: 72-7.

89. White JG, Krumwiede M. Normal-sized primary lysosomes are present inChediak-Higashi syndrome neutrophils. Pediatr Res 1987; 22: 208-15.

90. Root RK, Rosenthal AS, Balestra DJ. Abnormal bactericidal, metabolic,and lysosomal functions of Chediak-Higashi syndrome leukocytes. J ClinInvest 1972; 51: 649-65.

91. Abo T, Roder JC, Abo W, Cooper MD, Balch CM. Natural killer (HNK-1+) cells in Chediak-Higashi patients are present in normal numbers butare abnormal in function and morphology. J Clin Invest 1982; 70: 193-7.

92. Wolff SM, Dale DC, Clark RA, Root RK, Kimball HR. The Chediak-Higashi syndrome: studies of host defenses. Ann Intern Med 1972; 76:293-306.

93. Haliotis T, Roder J, Klein M, Ortaldo J, Fauci AS, Herberman RB.Chediak-Higashi gene in humans. Impairment of natural-killer function.J Exp Med 1980; 151: 1039-48.

94. Klein M, Roder J, Haliotis T, Korec S, Jett JR, Herberman RB, Katz P,Fauci AS. Chediak-Higashi gene in humans. The selectivity of the defectin natural-killer and antibody-dependent cell-mediated cytotoxicity func-tion. J Exp Med 1980; 151: 1049-58.

95. Padgett, GA, Reiquam CW, Gorham JR, Henson JB, O’Mary CC. Compa-rative studies of the Chediak-Higashi syndrome. Am J Pathol 1967; 51:553-71.

96. Rubin CM, Burke BA, McKenna RW, McClain KL, White JG, NesbitME, Filipovich AH. The accelerated phase of Chediak-Higashi syndrome.An expression of the virus-associated hemophagocytic syndrome? Cancer1985; 56: 524-30.

97. Sheramata W, Kott HS, Cyr DP. The Chediak-Higashi-Steinbrincksyndrome. Presentation of three cases with features resembling spinocere-bellar degeneration. Arch Neurol 1971; 25: 289-94.

98. Sung JH, Meyers JP, Stadlan EM, Cowen D, Wolf A. Neuropathologicalchanges in Chediak-Higashi disease. J Neuropathol Exp Neurol 1968; 28:86-118.

99 Diukman R, Tanigawara S, Cowan MJ, Golbus, MS. Prenatal diagnosis ofChediak-Higashi syndrome. Prenat Diagn 1992; 12: 877-85.

100. Kazmierowski J, Elin R, Reynolds H, Durbin W, Wolff S. Chediak-Higashi syndrome: reversal of increased susceptibility to infection bybone marrow transplantation. Blood 1976; 47: 555-9.

101. Haddad E, Le Deist F, Blanche S, Benkerrou M, Rohrlich P, Vilmer E,Griscelli C, Fischer A. Treatment of Chediak-Higashi syndrome by allo-genic bone marrow transplantation: report of 10 cases. Blood 1995; 85:3328-33.

102. Gallin JI, Elin RJ, Hubert RT, Fauci AS, Kaliner MA, Wolff SM. Efficacyof ascorbic acid in Chediak-Higashi syndrome (CHS): studies in humansand mice. Blood 1979; 53: 226-34.

103. Griscelli C, Virelizier JL. Bone marrow transplantation in a patient withChediak-Higashi syndrome. Birth Defects Orig Artic Ser 1983; 19: 333-4.

104. Fukai K, Oh J, Karim MA, Moore KJ, Kandil HH, Ito H, Burger J, SpritzRA. Homozygosity mapping of the gene for Chediak-Higashi syndrome tochromosome 1q42-44 in a segment of conserved synteny that includes themouse beige locus (bg). Am J Hum Genet 1996; 59: 620-4.

105. Barrat FJ, Auloge L, Pastural E, Lagelouse RD, Vilmer E, Cant AJ, Weis-senbach J, Le Paslier D, Fischer A, de Saint Basile G. Genetic and physi-cal mapping of the Chediak-Higashi syndrome on Chr 1q42-43. Am JHum Genet 1996; 59: 625-32.

106. Perou CM, Moore KJ, Nagle DL, Misumi DJ, Woolf EA, McGrail SH,Holmgren L, Brody TH, Dussault BJ, Monroe CA, Duyk GM, PryorRJ, Li L, Lustice MJ, Kaplan J. Identification of the murine beige geneby YAC complementation and positional cloning. Nat Genet 1996; 13:303-8.

107. Barbosa MD, Barrat FJ, Tchemev VT, Nguyen QA, Mishra VS, ColmanSD, Pastural E, Dufourcq-Lagelouse R, Fischer A, Holcombe RF, Wallace MR, Brandt SJ, de Saint Basile G, Kingsmore SF. Identifica-tion of mutations in two major mRNA isoforms of the Chediak-Higashisyndrome gene in human and mouse. Hum Mol Genet 1997; 6: 1091-8.

108. Karim MA Nagle DL, Kandil HH, Burger J, Moore KJ, Spritz RA. Muta-tions in the Chediak-Higashi syndrome gene (CHS 1) indicate requirementfor the complete 3801 amino acid CHS protein. Hum Mol Genet 1997; 6:1087-9.

109. Nagle DL, Karim MA, Woolf EA, Holmgren L, Bork P, Misumi Dj,McGrail SH, Dussault BJ, Perou CM, Boissy RE, Duyk GM, Spritz RA,Moore KJ. Identification and mutation analysis of the complete gene forChediak-Higashi syndrome. Nat Genet 1996; 14: 307-11.

110. Certain S, Barrat F, Pastural E, LeDeist F, Goyo-Rivas J, Jabado N,Benkerrou M, Seger R, Vilmer E, Beullier G, Schwartz K, Fischer A,de Saint Basile G. Protein truncation test of LYST reveals heterogeneousmutations in patients with Chediak-Higashi syndrome. Blood 2000; 95:979-83.

111. Perou CM, Pryor RJ, Naas TP, Kaplan J. The bg allele mutation is due toa LINE1 element retrotranspositon. Genomics 1997; 42: 366-8.

112. Mori M, Nishikawa T, Higushi K, Nashimura A. Deletion in the beigegene of the beige rat owing to recombination between LINE1s. MammGenome 1999; 10: 692-5.

113. Pfeifer M, Berg S, Reynolds AB. A repeating amino acid motif shared byproteins with diverse cellular roles. Cell 1994; 76: 789-91.

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11

Page 13: Hermansky-Pudlak Syndrome and Chediak-Higashi Syndrome ......Hermansky-Pudlak syndrome (HPS) and Chediak-Higashi syndrome (CHS), share several clinical characteristics and have piqued

245

Huizing: Hermansky-Pudlak and Chediak-Higashi Syndromes

114. Belmont LD, Mitchinson TJ. Identification of a protein that interacts withtubulin dimers and increases the catastrophe rate of microtubules. Cell1996; 84: 623-31.

115. Ramsay M. Protein trafficking violations. Nat Genet 1996; 14: 242-5.116. Faigle W, Raposo G, Amigorena S. Antigen presentation and lysosomal

membrane traffick in the Chediak-Higashi syndrome. Protoplasma 2000;210: 117-22.

117. Neer FJ, Schmidt CJ, Nambudripad R, Smith TF. The ancient regulatoryprotein family of WD-repeat proteins. Nature 1994; 371: 297-300.

118. Wang X, Herberg FW, Laue MM, Wullner C, Hu B, Petrasch-Parwez E,Kilimann MW. Neurobeachin: A protein kinase A-anchoring, beige/Chediak-Higashi protein homolog implicated in neuronal membranetraffick. J Neurosci 2000; 20: 8551-65.

119. Adam-Klages S, Adam D, Wiegmann K, Struve S, Kolanus W, Schneider-Mergener J, Kronke M. FAN, a novel WD-repeat protein, couples the p55TNF receptor to neutral sphingomyelinase. Cell 1996; 86: 937-47.

120. Feuchter AE, Freeman JD, Mager DL. Strategy for detecting cellulartranscripts promoted by human endogenous long terminal repeats: identi-fication of a novel gene (CDC4C) with homology to yeast CDC4. Geno-mics 1992; 13: 1237-46.

121. Rendu F, Breton-Gorius J, Lebret M, Klebanoff C, Buriot D, Griscelli C,Levy-Toledano S, Caen JP. Evidence that abnormal platelet functions inhuman Chediak-Higashi syndrome are the result of a lack of dense bodies.Am J Pathol 1983; 111: 307-14.

122. Stinchcombe JC, Page LJ, Griffiths GM. Secretory lysosome biogenesis incytotoxic T lymphocytes from normal and Chediak-Higashi syndromepatients. Traffic 2000; 1: 435-44.

123. Faigle W, Raposo G, Tenza D, Pinet V, Vogt AB, Kropshofer H, FischerA, de Saint-Basile G, Amigorena S. Deficient peptide loading and MHCclass II endosomal sorting in a human genetic immunodeficiency disease:the Chediak-Higashi syndrome. J Cell Biol 1998; 141: 1121-34.

124. Buckhardt, JK, Wiebel FA, Hester S, Argon Y. The giant organelles inBeige and Chediak-Higashi fibroblasts are derived from late endosomesand mature lysosomes. J Exp Med 1993; 178: 1845-56.

125. Jones KL, Stweart RM, Fowler M, Fukuda M, Holcombe RF. Chediak-Higashi lymphoblastoid cell line: granule characteristics and expressionof lysosome-associated membrane proteins. Clin Immunol Immunopathol1992; 65: 219-26.

126. Frankel FR, Tucker RW, Bruce J, Stenberg R. Fibroblasts and macropha-ges of mice with the Chediak-Higashi syndrome have microtubules andactin cables. J Cell Biol 1978; 79: 401-8.

127. White JG. Platelet microtubules and giant granules in Chediak-Higashisyndrome. Am J Med Tech 1978; 44: 273-8.

128. Pryzwansky KB, Schliwa M, Boxer LA. Microtubule organization of un-stimulated and stimulated adherent human neutrophils in Chediak-Higashisyndrome. Blood 1985; 66: 1398-403.

129. Tanabe F, Cui S-H, Ito M. Abnormal down-regulation of PKC is responsi-ble for giant granule formation in fibroblasts from CHS (beige) mice-a thiol proteinase inhibitor, E-64-d, prevents giant granule formation inbeige fibroblasts. J Leukocyte Biol 2000; 67: 749-55.

130. Ito M, Tanabe F, Takami Y, Sato A, Shigeta S. Rapid down-regulation ofprotein kinase C in (Chediak-Higashi syndrome) beige mouse by phorbolester. Biochem Biophys Res Commun 1988; 153: 648-56.

131. Ito M, Sato A, Tanabe F, Ishida E, Takami Y, Shigeta S. The thiol pro-teinase inhibitors improve the abnormal rapid down-regulation of proteinkinase C and the impaired natural killer cell activity in (Chediak-Higashisyndrome) beige mouse. Biochem Biphys Res Commun 1998; 160:433-40.

132. Coffer PJ. Phosphatidylinositol 3-kinase signalling: A tale of two kinaseactivities. In: Protein Kinase functions. Woodgett J, ed. Oxford UniversityPress 2000; 1-39.

133. Corvera S, D’Arrigo A, Stenmark H. Phosphoinositides in membranetraffic. Curr Opin Cell Biol 1999; 11: 460-5.

134. Wurmser AE, Gary JD, Emr SD. Phosphoinositide 3-kinase and theirFYVE domain-containing effectors as regulators of vacuolar/lysosomalmembrane trafficking pathways. J Biol Chem 1999; 274: 9129-32.

135. Brown WJ, DeWald DB, Emr SD, Plutner H, Balch WE. Role for thephophatidylinositol 3-kinase in the sorting and transport of newly synthe-sized lysosomal enzymes in mammalian cells. J Cell Biol 1995; 130:781-96.

136. Davidson HW. Wortmannin causes mistargeting of procathepsin D.Evidence for the involvement of a phosphatidylinositol 3-kinase in vesicu-lar transport to lysosomes.J Cell Biol 1995; 130: 797-805.

137. Klionsky DJ, Emr SD. A new class of lysosomal/vacuolar protein sortingsignals. J Biol Chem 1990; 265: 5349-52.

138. Stack JH, Herman PK, Schu PV, Emr SD. A membrane-associated com-plex containing the VPS15 protein kinase and the VPS34 PI 3-kinase isessential for protein sorting to the yeast lysosome-like vacuole. EMBO J1993; 12: 2195-204.

139. De Camilli P, Emr SD, McPherson PS, Novick P. Phospho-inositides asregulators in membrane traffic. Science 1996; 271: 1533-8.

140. Panaretou C, Domin J, Cockroft S, Waterfield MD. Characterization ofp150, an adaptor protein for the human phosphatidylinositol(PtdIns)3-kinase. J Biochem Chem 1997; 272: 2477-85.

141. Davies JP, Cotter PD, Ioannou YA. Cloning and mapping of human Rab7and Rab9 cDNA sequences and identification of a Rab9 pseudogene.Genomics 1997; 41: 131-4.

142. Bucci C, Thomsen P, Nicoziani P, McCarthy J, van Deurs B. Rab7: A keyto lysosome biogenesis. Mol Biol Cell 2000; 11: 467-80.

143. Stein MP, Feng Y, Wandinger-Ness A. Binding of Rab7 to human Vps34,a cytosolic phosphatidyl inositol-3-kinase is nucleotide dependent. MolBiol Cell 2000; 11: Suppl 505a (Abstr).

144. Pastural E, Barrat FJ, Dufourcq-Lagelouse R, Certain S, Sanal O, JabadoN, Seger R, Griscelli C,Fischer A, de Saint Basile G. Griscelli diseasemaps to chromosome 15q21 and is associated with mutations in themyosin-Va gene. Nat Genet 1997; 16: 289-92.

145. Mercer JA, Seperack PK, Strobel MC, Copeland NG, Jenkins NA. Novelmyosin heavy chain encoded by murine dilute coat colour locus. Nature1991; 349: 709-13.

For personal or educational use only. No other uses without permission. All rights reserved.Downloaded from www.thrombosis-online.com on 2018-05-18 | ID: 1001066444 | IP: 54.70.40.11