Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Aug 4.
Published in final edited form as: Curr Opin Allergy Clin Immunol. 2007 Dec;7(6):481–494. doi: 10.1097/ACI.0b013e3282f1d690

Genetic Heterogeneity in Severe Congenital Neutropenia: How Many Aberrant Pathways Can Kill a Neutrophil?

Alejandro A Schäffer 1, Christoph Klein 2
PMCID: PMC2720578  NIHMSID: NIHMS41834  PMID: 17989524

Abstract

Purpose of review

Severe congenital neutropenia (SCN) is a primary immunodeficiency in which lack of neutrophils causes inadequate innate immune host response to bacterial infections. SCN occurs with sporadic, autosomal dominant (AD), autosomal recessive (AR), and X-linked recessive (XLR) inheritance, as well as in a variety of multi-system syndromes. A principal stimulus for this review is the identification of novel genetic defects and pathophysiological insights into the role of neutrophil apoptosis.

Recent findings

Identification of mutations in HAX1 in autosomal recessive SCN (Kostmann disease), large epidemiological study estimating the risk of progression from SCN to leukemia, better understanding of how heterozygous mutations in neutrophil elastase (ELA2) cause SCN, molecular characterization of a novel syndromic form of SCN called p14 deficiency, and new animal models for several syndromic forms of SCN.

Summary

We consider the numerous genes mutated in SCN, many attempts to make animal models of SCN, and results from both human and mouse studies investigating the molecular mechanisms of neutrophil apoptosis. Investigations of how SCN genes and apoptosis pathways are connected should lead to better understanding of the pathogenesis of neutropenia and apoptosis pathways relevant to many cell types.

Keywords: severe congenital neutropenia, neutrophils, genetics, apoptosis, primary immune deficiencies

Introduction

This review is about severe congenital neutropenia (SCN), a primary immunodeficiency in which lack of neutrophils causes susceptibility to infections. Neutrophils are leukocytes in the innate immune system [1] that help their host fight infections using secreted molecules toxic to microbes, proteases to cut microbial proteins, and phagocytosis to ingest and degrade dying microbes [2]. Neutrophils are classified along with basophils and eosinophils as “granulocytes” because they possess interior granules including “primary” or “azurophilic granules” that generally sequester, and when needed secrete microbicidal peptides and proteinases such as cathepsin G, neutrophil elastase, and proteinase 3 [2]. Neutrophil “secondary” or “specific” granules contain lactoferrin, transcobalamin II, lysozyme, lipocalin, and some membrane proteins [2]. Neutrophils have a high turnover, since they are produced at a rate exceeding 106 cells/min/kg [3] and have an estimated half-life of under 12 hours in healthy, uninfected individuals [4,5]. When the host is fighting an infection, neutrophils die even more quickly [1, 6]. Neutrophils die by apoptosis rather than necrosis, thus limiting tissue damage in the host [7]. In addition, a peculiar mechanism by which neutrophils launch “neutrophil extracellular traps” to kill bacteria and trigger their own suicide has recently been detected [8••].

Neutropenia definition

Neutropenia refers to the condition of having an inadequate number of neutrophils. Neutropenia is diagnosed by measuring the absolute neutrophil count (ANC) in peripheral blood and defined as an ANC below a commonly-used threshold such as 1500/µl; severe neutropenia is typically diagnosed when the ANC is below 500/µl, while some clinicians prefer the threshold 200/µl [9]. However, these thresholds were derived mostly by testing white individuals, and members of other ethnicities may have lower, yet protective ANC [9]. For the purpose of fighting infections what matters more are the microbicidal functions of neutrophils and the capability to rapidly ramp up the production of neutrophils in the bone marrow and their release into the bloodstream.

In adults, neutropenia is most commonly an acquired condition. Neutropenia is commonly seen in cancer patients being treated with chemotherapy that indiscriminately kills rapidly proliferating cells or in association with autoimmune disorders. In this review, we focus on severe congenital neutropenia (SCN), in which low ANC is observed from birth and it remains low, unless the patient is treated. The incidence of SCN has been estimated at 1/200,000 [3], but more epidemiological studies are needed.

Neutropenia treatment

SCN was almost always fatal in infancy until effective supportive treatment regimens became available. These include powerful antibiotics and recombinant human granulocyte colony stimulating factor (G-CSF) [10, 11]. G-CSF both stimulates the production of more neutrophils [4, 12] and delays their apoptosis [13,14]. The vast majority of SCN patients respond to G-CSF, with ANC increased toward the normal range and a concurrent reduction in infections [15, 16••]. Some patients also respond to a related hematopoietic growth factor, granulocyte-macrophage colony stimulating factor (GM-CSF) [e.g., 17], but some do not [18]. A recent in vitro study confirmed that G-CSF is more effective than GM-CSF at delaying neutrophil apoptosis and preserving the neutrophil chemotactic response to some cytokines; for example, G-CSF preserves the chemotactic response to interleukin 8, but GM-CSF does not [19•].

Although G-CSF keeps many SCN patients alive and functioning into adulthood [20], a recent study estimated that SCN patients die from sepsis at an elevated rate of 0.9% per year [16••]. Prolonged survival with SCN is associated with a substantially increased risk of acute myeloid leukemia (AML)[21]. The same study estimated the 10-year incidence of AML or related myelodysplastic syndrome at 21% [16••]. The occurrence of AML has been associated with somatic mutations in the gene CSF3R, which encodes the receptor for G-CSF [2224]. Mermel et al. [25•] suggested that these somatic mutations are leukemogenic because they eliminate signaling to Src family kinases Hck and Lyn, which are negative regulators of granulopoiesis. A recent study found CSF3R mutations in newly diagnosed SCN patients who had not yet started on G-CSF treatment [24]. While this suggests that G-CSF treatment does not initiate the leukemogenesis, Sloand et al. [26•] suggested that G-CSF helps the progression to leukemia by favoring the proliferation of cells with a monosomy of chromosome 7 over healthy cells disomic for that human chromosome.

Genetics and etiology of SCN

Neutropenia, as measured by low ANC, could be the consequence of: a) a defect in granulopoiesis b) a defect in moving the neutrophils from the bone marrow to the bloodstream or c) excessive apoptosis. Identifying genes mutated in the germline of SCN patients and studying the normal function of the encoded proteins is one strategy to recognize novel mechanisms of neutrophil homeostasis and/or function. Two stimuli for this review are the discoveries of mutations in the gene HAX1 in some cases of nonsyndromic autosomal recessive neutropenia [27••] and the molecular characterization of a new neutropenia syndrome caused by a homozygous mutation in the gene p14 [28••]. Because HAX1 protein was known to be a negative regulator of apoptosis [29], the first of these findings has brought increased attention to the possibility that excess apoptosis is a central mechanism in the etiology of SCN [30]. Genetic heterogeneity and the role of apoptosis in the etiology of SCN are themes of this review. From this perspective, central questions in the molecular biology of SCN include:

  1. Which genes mutated in SCN lead to excessive apoptosis of neutrophils?

  2. In which apoptosis pathways do the encoded proteins participate?

In the next section, we explain the genetic underpinnings of these two questions by reviewing the molecular etiologies of syndromic and nonsyndromic SCN. Grenda and Link [31•] give a lengthier review of this topic. In this article, we define syndromic SCN as those clinical phenotypes that include a non-immunological aspect, while nonsyndromic forms have the phenotype confined to the immune response. We only use the childhood phenotype (e.g., not including progression to leukemia) to make this distinction. “Recent advances” are formally defined as those not published in print before 1 January 2006.

Molecular Causes of Syndromic and Nonsyndromic Severe Congenital Neutropenia

Both syndromic and non-syndromic SCN can occur in autosomal dominant, autosomal recessive, X-linked recessive, or sporadic forms. Table 1 summarizes genes currently known to be mutated in monogenic syndromic and non-syndromic SCN. A substantial fraction of non-syndromic SCN cases have no gene mutation identified, but quantifying this has been confusing because of clinical and geographic biases in ascertainment.

Table 1.

Genes mutated in human syndromic and non-syndromic congenital neutropenias, not including some genes mutated in combined immunodeficiencies. The references given here are to papers that identified the initially discovered mutation(s). Other papers describing the clinical phenotypes and additional mutations are cited in the text. Inheritance: AD= autosomal dominant; AR = autosomal recessive; XLR= X-linked recessive; M = mitochondrial. Gene symbols are the current HUGO Gene Nomenclature Committee-approved symbols with the exception of p14 that has no approved symbols; therefore, gene symbols may differ from gene names used in the cited papers.

Disease Inheritance Mutated Gene Reference(s) comments
Severe Congenital Neutropenia (SCN) AD ELA2 [32] Mostly missense mutations
SCN AD GFII [33] Missense mutations in the zinc finger domain
SCN XLR WAS [34, 35•] Missense mutations that lead to constitutive activation
SCN AR HAX1 [27••]
Cyclic neutropenia AD ELA2 [36] Mutations mostly different from those causing SCN
Glycogen storage disease type Ib AR SLC37A4 [37]
Hermansky-Pudlak syndrome, type 2 AR AP3B1 [38] Other types of Hermansky-Pudlak syndrome do not include neutropenia
Griscelli syndrome, type 2 AR RAB27A [39] Other types of Griscelli syndrome do not include neutropenia
Chediak-Higashi syndrome AR LYST [4042]
p14 deficiency AR p14 [28••]
WHIM syndrome AD CXCR4 [43] Mutations truncate the cytoplasmic tail of the protein
Cohen syndrome AR VPS13B [44]
Shwachman-Diamond syndrome AR SBDS [45] Most patients have two compound heterozygous mutations
Barth syndrome XLR TAZ [46]
Cartilage hair hypoplasia AR RMRP [47] This is an RNA gene that does not code for a protein.
Pearson’s syndrome M Mitochondrial deletion [48] Mutations in mitochondrial DNA

A variety of monogenic, multi-system syndromes including neutropenia as a symptom have been reported. New, rare neutropenia syndromes continue to be discovered [28••, 49•]. We list many as “miscellaneous” because there are no known connections between the functions of the mutated genes. One exception to this confusing heterogeneity is a category of four syndromes (Hermansky-Pudlak syndrome type 2, Griscelli syndrome type 2, Chediak-Higashi syndrome, and the recently discovered p14 deficiency) that combine neutrophil dysfunction with hypopigmentation.

Autosomal dominant non-syndromic SCN

The most commonly mutated gene in non-syndromic SCN is ELA2, which codes for neutrophil elastase, a protease contained in the azurophilic granules [50•]. Heterozygous mutations in ELA2 were discovered first in a related disorder called cyclic neutropenia, in which the ANC cycles from low to normal levels over a period of approximately 21 days [36]. Cyclic neutropenia is inherited as an autosomal dominant trait, and most cases appear to be associated with heterozygous mutations of ELA2 [50•]. Subsequent to the discovery of ELA2 mutations in cyclic neutropenia, ELA2 mutations were also found in sporadic and familial cases of SCN [32]. To date, over 40 ELA2 mutations have been found, including some which are associated with both phenotypes [50•].

Before the discovery of ELA2 mutations, dominant inheritance of SCN had been occasionally documented [e.g., 51], but was controversial. Recently, a striking example of dominant inheritance was reported where an anonymous sperm donor fathered five neutropenic children all of whom inherited the S97L mutation [52•]. It remains mysterious how the sperm donor escaped clinical attention, but one possibility is that he is mosaic for the mutation.

ELA2 was a plausible candidate gene for neutropenia because it encodes a known neutrophil protein, but both the complete function of neutrophil elastase and the mechanism of dominance are not fully understood. Neutrophil elastase cleaves bacterial proteins as one infection fighting mechanism. Neutrophil elastase may also bind to and cleave host proteins. One candidate for this role is N2N, a member of the Notch family of proteins, which has been shown to be cleaved by ELA2 in vitro [53]. Other candidates are G-CSF and G-CSFR [54]. Li and Horwitz [55] showed that a variety of heterozygous mutations in ELA2 act in a truly dominant negative fashion, inhibiting the enzymatic activity of the protein encoded by the wild type allele. ELA2 with a disease-associated G185R mutation mis-localizes and accelerates apoptosis of differentiating early myeloid cells [56].

Two recent studies have added considerable insight to these earlier findings. Thusberg and Vihinen [57•] used 30 bioinformatics tools to predict, in silico, the effects of 32 missense mutations in ELA2. They found that mutations could: affect a conserved residue, change the electrostatic surface potential, affect critical contacts, cause the protein to become disordered, change the protein structure in other ways, but that there was no single cause that could explain all mutations. This finding is consistent with in vitro results of Köllner et al. [58•] showing that several different missense mutations could trigger the unfolded protein response causing the neutrophil with mutant ELA2 to die via apoptosis. We explore the apoptosis theme in a later section. The difficulty in understanding the mechanism of action of ELA2 mutations stems from the lack of an adequate animal model, as discussed in the next section.

Another rare cause of autosomal dominant SCN is heterozygous mutations in the zinc finger domain of the transcription factor GFI1 [33]. The original report showed that GFI1 protein binds to ELA2 DNA and speculated that repression of ELA2 is related to the neutropenia.

X-linked recessive SCN

X-linked recessive SCN was associated with an activating missense mutation in WASP (Wiskott-Aldrich syndrome protein), initially in a single pedigree [34]. Recently, two more male SCN patients with mutations in WASP were reported, confirming the initial finding [35•]. As its name indicates, WASP was originally found to be mutated in a different immunodeficiency, Wiskott-Aldrich syndrome, for which the known mutations generally eliminate or reduce the protein activity. The hallmark symptoms of Wiskott-Aldrich syndrome are excessive bleeding secondary to (micro-) thrombocytopenia, recurrent infections, and eczema. A milder phenotype is seen in a condition termed X-linked thrombocytopenia (reviewed in [59]).

Autosomal recessive SCN

In contrast to dominant SCN, recessive inheritance in SCN was established over 50 years ago by Kostmann [60] who published a large pedigree with many cases and proved the inheritance mode by statistical tests. However, no gene for autosomal recessive SCN had been found until recently. One discovery that stimulates this review is that homozygous nonsense mutations in the gene HAX1 explain some cases of autosomal recessive SCN [27••], including surviving descendants of the original SCN pedigree published by Kostmann. A substantial fraction of recessive non-syndromic SCN cases do not yet have an associated mutation [27••, unpublished observation].

The full name of HAX1 is “HS1 interacting protein 1” because HAX1 was first discovered via a yeast two-hybrid screen with HS1, which is a member of the Src family of kinases and plays roles in proliferation and apoptosis of lymphoid cells [61]. The ‘X’ comes from the “X-Gal colony filter assay”, used in the yeast two-hybrid experiment. HAX1 was shown to localize to the mitochondria, and this was a useful clue to make HAX1 a candidate gene for SCN. In SCN patients with two null mutations of HAX1, it is presumed that the HS1-HAX1 protein interaction cannot take place, but how this relates to the SCN phenotype is unclear. More pertinent to the SCN phenotype are the results showing that HAX1 is an anti-apoptotic protein that is cleaved by Omi/HTRA2 [29], as part of the mitochondrial apoptosis pathway, covered in a later section. This function of HAX1 suggested that patients with mutations should have a loss of mitochondrial membrane potential and consequent excessive neutrophil apoptosis, and this hypothesis was confirmed [27••].

Syndromes that combine SCN or neutrophil dysfunction with hypopigmentation

Some of the granules in neutrophils also belong to a more general class of lysosome-related organelles, sometime called “secretory lysosomes”. Other cell types with secretory lysosomes include: cytotoxic T lymphocytes, platelets, and melanocytes [62]. The secretory lysosomes serve to sequester, transport, and secrete substances such as pigment and microbicidal peptides that could be toxic to the cell if released at the wrong time and place. When secretory lysosome proteins are defective, the phenotypic consequence may be a multi-system disorder affecting different cell types. When the syndrome affects melanocytes, the phenotypic consequence is hypopigmentation and that symptom is more obvious to clinicians, but less dangerous to the patient than any associated leukocyte symptoms. All known secretory lysosome defect syndromes are purely recessive. Here we focus on four such syndromes that combine hypopigmentation and either SCN or another neutrophil dysfunction: Hermansky-Pudlak syndrome (HPS), type 2; Griscelli syndrome (GS), type 2; Chediak-Higashi syndrome (CHS); and the recently described p14 deficiency.

The original syndrome described by Hermansky and Pudlak [63] consists primarily of hypopigmentation and prolonged bleeding times due to defective platelet granules. There are 8 known human genes each of which is mutated in a different recessive form of HPS, and these forms are numbered HPS1-HPS8 according to the chronological order of gene discovery. Only HPS2, which is caused by a mutation in the gene AP3B1, includes a neutrophil dysfunction. The dysfunction was originally characterized as neutropenia [38], but recent work extends this phenotype. The AP3B1 protein is part of a protein transport complex called AP3. Neutrophil dysfunction has not been associated with mutations in other members of the complex. It is unknown what AP3 cargo is most relevant to neutrophil count or function, but Benson et al. [64] suggested that neutrophil elastase is an AP3 cargo in both dogs and humans. Support for this hypothesis comes from the observation that when G185R mutant ELA2 mis-localizes, the AP3 complex also mis-localizes [56]. Recent findings on HPS2 include: 1) extension of the phenotype to include dysfunction of other cell types such as NK cells, NKT cells and some antigen presenting cells [65,66]; 2) description of a patient whose phenotype also included hemophagocytic lymphohistiocytosis and whose genotype also included a heterozygous mutation in RAB27A, the gene mutated in GS2 [67]; and 3) positive resolution of a conjecture of Huizing et al. [68] that the affected members of a pedigree described by Kotzot et al. [69] have a form of HPS2 and not a different hypopigmentation-SCN syndrome [66].

GS2 is also characterized by albinism and (intermittent) neutropenia. Unlike HPS2, the platelet granules in GS2 patients look normal, but the platelet count may be reduced [70]. GS2 patients are susceptible to infections both because of the neutropenia and defective function of cytotoxic lymphocytes. The protein deficient in GS2, RAB27A is part of a three-protein complex including myosin VA and melanophilin. Deficiency of either of the other two proteins is also associated with hypopigmentation, but is not associated with any immune dysfunction. Neutrophils in GS2 patients appear to be defective in bacterial killing [71]. Recent progress includes the discovery that RAB27A plays a role in the release of myeloperoxidase (MPO) from azurophilic granules of neutrophils [72•], and this could explain the functional defect in GS2 neutrophils.

CHS is characterized by hypopigmentation, a bleeding disorder, immunodeficiency partly due to neutropenia and partly due to defective natural killer cells [31•, 7375]. The most severely affected CHS patients also have a progressive neurological disease [31•]. In CHS, neutrophils and other secretory granule cells have mis-shaped granules with inclusion bodies [31•] In neutrophils, these defective granules are now known to be the azurophilic granules, essential for bacterial killing [76]. As noted in Table 1, some CHS patients have double mutations in the gene LYST. Karim et al. [77] showed both that there is a genotype-phenotype correlation among those CHS patients with LYST mutations, and that some CHS patients do not have LYST mutations. No second CHS gene or genetic locus has been found.

Recently a new rare syndrome combining neutropenia and hypopigmentation was characterized in a single family. This syndrome is tentatively called “p14 deficiency” because it is caused by a homozygous point mutation in the 3’ untranslated region of the gene that encodes p14 [28••]. The patients do have a small amount of p14 and this likely explains why they can live, while p14-knockout mice are not viable [78•]. Other symptoms of p14 deficiency include short stature, hypogammaglobulinemia and reduced numbers of B cell subsets, and defective function of cytotoxic T cells, which contribute to a general immunodeficiency [28••]. p14 is an endosomal adaptor protein that acts as a scaffold when binding to MP1, enabling MP1 to participate in the ERK signaling cascade [79,80]. As for GS2, and CHS, neutrophils in p14-deficient patients are defective in bacterial killing [28••].

Miscellaneous syndromes that include neutropenia as a symptom

WHIM syndrome derives its name from the combination of symptoms: warts, hypogammaglobulinemia, infections and myelokathexis [81]. “Myelokathexis” means that neutrophils are not released properly from the bone marrow to the blood stream. Myelokathexis occurs also as a non-syndromic form of neutropenia [82], but no mutated genes have been found for non-syndromic myelokathexis. In the Introduction, we suggested that inadequate release of neutrophils to the blood stream might be a problem separate from granulopoiesis or apoptosis, but Aprikyan et al. [83] showed that myelokathexis is associated with excessive apoptosis and under-expression of the apoptosis inhibitor bcl-x. The hypogammaglobulinemia in WHIM syndrome is mild [81].Since neutrophils and immunoglobulins mainly protect against bacterial infections, it is surprising that infection with human papillomavirus (HPV) is common in WHIM syndrome, although immunity to other viral pathogens is normal [31•].

WHIM syndrome is unique among the syndromes we discuss in that inheritance of most cases is autosomal dominant [43, 81]. These cases are usually associated with a heterozygous activating mutation in the C-terminal section of the chemokine receptor gene CXCR4 [43]. No mutated gene has been found for sporadic or recessive cases of WHIM syndrome. The molecular pathogenesis appears to be due to damaged signaling between stromal-derived factor 1 (SDF-1) and CXCR4 [31•]. Three recent findings strengthen and clarify this hypothesis: 1) in an in vitro assay, addition of extra SDF-1 to WHIM patient cells eliminates excess apoptosis [84•]; 2) in a mouse model, G-CSF (known to be effective for WHIM syndrome patients) downregulates CXCR4 expression and this stimulates release of neutrophils into the bloodstream [85•]; 3) human CD34+ cells transduced with mutant CXCR4 showed increased engraftment into bone marrow associated with increased apoptosis upon transplantation into NOD/SCID mice [86•].

Cohen et al. [87] described an autosomal recessive syndrome including obesity, hypotonia, mental deficiency, craniofacial anomalies, large incisors, limb anomalies, and spinal anomalies. Norio et al. [88] observed that in a majority of Finnish Cohen syndrome patients the phenotype also included chorioretinal dystrophy and SCN. A clinical consensus to address the phenotypic heterogeneity and including neutropenia as a primary symptom can be found in [89]. Despite the phenotypic heterogeneity, Cohen syndrome is genetically homogeneous . all patients have a double mutation of VPS13B (a.k.a. COH1) [44]. The human protein VPS13B is homologous to a yeast protein VPS13, which has a function in protein sorting and intracellular trafficking. However, there is no detailed explanation of how VPS13B deficiency causes the symptoms of Cohen syndrome, including the neutropenia.

Shwachman-Diamond syndrome (SDS) is autosomal recessive and is characterized by pancreatic insufficiency, failure to gain weight, skeletal abnormalities, and bone marrow dysfunction [90, 91]. Most, but not all SDS patients have neutropenia [92]. Progression to leukemia is common [93]. The majority of SDS patients have two distinct mutations in the gene named SBDS [45,94]. A major recent advance was the characterization of the function of the orthologous yeast protein SDO1 (SBDS ortholog 1). SDO1 participates in a complex that facilitates release of Tif6 from pre-60S ribosomes in the process of their maturation [95••]. Understanding of the proteins in this complex led Menne et al. [95••] to propose that EFTUD1 (elongation factor Tu GTP binding domain containing 1), which is the human ortholog of yeast EFL1 (elongation factor like 1) may be the gene mutated in SDS patients who do not have SBDS mutations.

Glycogen storage disease (GSD) type I comprises at least two variants of an autosomal recessive syndrome that share the symptoms of glycogen accumulation in the liver leading to hypoglycemia, hepatomegaly, growth retardation, and other metabolic problems such as lactic aciduria. The disease is due to difficulty in converting glycogen to glucose. Type Ib was initially distinguished from type Ia by a metabolic test showing that patients with the much more common type Ia [96] have deficient activity of the enzyme glucose-6-phosphatase, while patients with type Ib have adequate glucose-6-phosphatase activity [97]. It was shown later that type Ib differs from type Ia in including neutropenia as a common symptom [98]. The gene mutated in GSD Ib is now called SLC37A4 and it encodes a protein that transports glucose 6-phosphate from the cytosol to the endoplasmic reticulum (glucose-6-phosphate-translocase) [31•, 37]. The cellular reason why neutropenia occurs in GSD Ib but not GSD Ia was elucidated via knockout mouse models, as explained in the next section. Why glucose and glycolysis may be more important to neutrophils than some other cell types is discussed in the section on apoptosis.

Barth syndrome is an X-linked recessive syndrome. Symptoms include: dilated cardiomyopathy, skeletal myopathy, carnitine deficiency, 3-methylglutaconic aciduria and neutropenia [99, 100]. Two decades after its initial characterization, survival for Barth syndrome patients had improved dramatically due to recognition and treatment of the cardiac defects and the neutropenia [101]. Barth syndrome patients were found to have a deficiency of cardiolipin [101], which is a protein uniquely synthesized in the inner membrane of mitochondria [102]. The mutated gene, now called TAZ, was identified by positional cloning [46]. Female carriers of TAZ mutations have skewed X inactivation selecting against the chromosome with the mutated gene, which explains why they have no reported phenotype [103].

Cartilage hair hypoplasia was originally characterized by short stature and unusually kinked hair [104]. Susceptibility to viral infections and severe chickenpox are mentioned in the original report, but immunodeficiency was not considered a defining clinical finding [104]. Later studies recognized neutropenia as a recurrent finding and it was estimated that ¼ of Finnish CHH patients have neutropenia [105]. Cartilage hair hypoplasia (CHH) like Cohen syndrome is a rare recessive syndrome seen most commonly in the Amish and Finnish populations [104,105]. Another feature in common with Cohen syndrome is that there is substantial phenotypic variability, but locus homogeneity. All CHH patients have a double mutation in the RNA coding gene RMRP, which is transcribed to be a subunit of the RNAse mitochondrial RNA processing complex [47]. The phenotypic variability may be due to allelic heterogeneity, since some patients have two “mild” mutations, some more severely affected patients have one “mild” mutation and one null mutation, and no CHH patients have two null mutations [31•]. The neutropenia in CHH appears to be due to a defect in granulopoiesis, not migration or apoptosis [31•].

Pearson’s syndrome combines the symptoms of anemia, neutropenia, thrombocytopenia, pancreatic insufficiency, and a vacuolization of erythroid and myeloid precursor cells in the bone marrow [106]. Pearson’s syndrome is unique among the diseases discussed here in that it is caused by deletions in the mitochondrial genome [48]. Just as for ELA2 mutations in SCN, these deletions are usually new, and could occur in mitosis leading to germline mosaicism [31•].

Attempts to Make Animal Models

Animal models of SCN could provide opportunities to test the effects of specific infections and to do invasive experiments at a molecular level. Since neutrophils are part of the innate immune system, one might hope that neutrophil development and function would be similar in mice and humans. Non-mammalian models could be relevant since neutrophils exist in bony fish; similar cells, called “heterophils” exist in birds and reptiles [107]. Moreover, cells with phagocytic capability of neutrophils exist in many invertebrates; of historical note, Metchnikoff won the 1908 Nobel Prize for his studies of phagocytosis in starfish. Recently, fish and fruitfly models of Barth syndrome have been developed, but neither study examined neutrophils or other phagocytic cells [108, 109]. Despite the deep evolutionary history of neutrophil-like cells, the development of animal models may be hampered by lineage-related specificity in the granule contents, perhaps due to co-evolution of vertebrate hosts and their common bacterial pathogens [1, 107]. Attempts to make animal models, especially mouse models of human monogenic SCN, have yielded mixed results.

Table 2 summarizes some models for the disorders discussed in the previous section, and a few other models such as Csf3−/− and Csf3r−/− mice that are neutropenic and are relevant to human SCN because Csf3 codes for mouse G-CSF and Csf3r codes for its receptor. The mouse models that best correspond to the human SCN phenotype are for some of the miscellaneous neutropenia syndromes such as: WHIM syndrome, Chediak-Higashi syndrome, and glycogen storage disease (GSD) Ib using respectively a xenotransplant model [86•], a natural mouse model [40], and engineered mice with a single gene knocked out unconditionally [119] or conditionally [120]. In the case of GSD Ib, the neutropenia documented in a recently developed mouse knockout of G6pc3 [116] helps explain why GSD Ib includes neutropenia as a symptom, but GSD Ia does not. GSD Ia is caused by mutations in G6PC, which encodes glucose-6 phosphatase α, while the GSDIb gene, SLC37A4 encodes the glucose-6 phosphatase transporter. These two proteins interact in glycogen to glucose conversion in the liver and kidney, and deficiency of either one causes an equivalent GSD I phenotype in those cells. In neutrophils, the glucose-6 phosphatase transporter is present, but G6PC is replaced by a paralogous protein G6PC3 [116]. Therefore, Cheung et al. tested and confirmed the hypothesis that in neutrophils, knocking out either Slc37a4 or G6pc3 causes an equivalent neutropenic phenotype [116,120].

Table 2.

Animal models used to study the pathogenesis of the diseases listed in Table 1. This Table includes both naturally occurring and engineered mutants. This Table includes animals with mutations in genes orthologous to those in Table 1 as well as some animals with mutations in genes relevant to neutrophil development and apoptosis.

Organism Gene mutation Reference Phenotypic comments
Mouse Csf3 null [110] Neutropenia, but ANC > 0
Mouse Csf3r null [111] Neutropenia, but ANC > 0; expedited apoptosis
Mouse Ela2 null [112] Mice have poor neutrophil function, but normal neutrophil counts
Mouse Ela2 heterozygous knockin of V72M [113] Neutrophil count and function appear normal
Mouse Gfi1 null [114, 115] Severe immunodeficiency, including neutropenia; heterozygote mice have no phenotype
Mouse G6pc3 null [116] Neutropenia
Mouse A1-a (homolog of human BCL2A1) null [117] Neutrophils have enhanced apoptosis
Mouse Mcl1 conditional null in macrophages and neutrophils [118•] Neutrophils have excess apoptosis, but macrophages do not.
Mouse Slc37a4 null [119] Hypoglycemia, hyperlipidemia, neutropenia (similar to human GSDIb)
Mouse Slc37a4 knockout+transplant to bone marrow [120] Defects in neutrophil number and function
Dog (collies) AP3B1 null [64] Gray coat color and cyclic neutropenia
Mouse Ap3b1 null [121] This mouse is called pearl due to the hypopigmentation similar to that in human HPS2, but there is no mention of neutropenia or any immunodeficiency
Mouse Rab27a null [122] The mouse is called ashen due to the hyopigmentation similar to that in human Griscelli syndromes, but there is no mention of neutropenia or any immunodeficiency
Mouse Lyst null? [40] Name is beige and it resembles human CHS
Mouse p14 null [78•] Embryonic lethal
Mouse Truncated Cxcr4 xenotransplantion of human cells into NOD/SCID mice [86•] Myelokathexis
Yeast SDO1/YLR022C (homolog of SBDS) [95••] Deficiency in pre-60S ribosome maturation
Zebrafish TAZ knockdown [109] Neutrophils not investigated
Drosophila TAZ ortholog null [110] Phagocytic cells not investigated

Another gene for which knockout mouse models were helpful to understand a human form of SCN is Gfi1. That Gfi1−/− mice are severely neutropenic [114,115], led Person et al. to sequence GFI1 in human patients and find heterozygous point mutations [33]. The phenotype of humans with heterozygous GFI1 mutations is much milder than the knockout mice, but heterozygous Gfi1+/− mice are not neutropenic [114]. To our knowledge, no knock-in models of the point mutations in GFI1 have been attempted.

Human SCN-associated mutations in GFI1, WASP, or ELA2 appear not to be functionally null, and this has been especially problematic in the case of ELA2, the most commonly mutated gene in human nonsyndromic SCN. Ela2 knockout mice have normal ANC, but poor neutrophil function [112]. The latter finding is consistent with a role for neutrophil elastase in bacterial killing [2, 50•]. The former finding is consistent with the fact that no humans have been reported with a phenotype SCN and a genotype including mutations in both alleles of ELA2. Grenda et al. [113] engineered a mouse with a heterozygous Ela2 mutation knocked in, but the mice do not have neutropenia. Thus, there is no animal model well-suited to studying heterozygous ELA2 mutations.

A similarly discouraging situation exists for the hypopigmentation+neutropenia syndromes Hermansky-Pudlak syndrome, type 2 and Griscelli syndrome, type 2, even though some affected humans appear to have double null mutations in the genes AP3B1 [68] and RAB27A [39]. Ashen mice, which have a natural homozygous null mutation in Rab27a, have a pigment defect, but no apparent immunodeficiency [122]. Pearl and mocha mice, natural mutants which have different components of the AP3 complex disrupted, also have a pigment defect, but no apparent immunodeficiency [e.g., 121]. AP3B1-deficient gray collies have a pigment phenotype and cyclic neutropenia, but not SCN [64].

Finally, we consider prospects for mouse models of HAX1 deficiency and p14 deficiency. The p14 situation appears challenging since the only known human mutation is in the 3’ UTR and affected individuals have low levels of p14 protein [28••]. A constitutive knockout of the orthologous mouse gene is not viable [78•]. Since some amount of p14 seems necessary for development of many tissues, a strategy such as reducing expression levels by siRNA may be more effective than modifying the p14 locus in the germline. For HAX1, all three known human mutations lead to premature stop codons in translation [27••], giving hope that a knockout of mouse Hax1 could be relevant. Indeed, a Hax1−/− mouse was developed independently of and in parallel with the HAX1 human gene hunt, but its phenotype does not recapitulate the phenotype of human SCN patients [J. Ihle, personal communication].

Apoptosis and Severe Congenital Neutropenia

The recent discovery of HAX1 mutations in SCN [27••] combined with the previous knowledge that HAX1 is an anti-apoptotic protein in the intrinsic/mitochondrial pathway of apoptosis [29] increases interest in the role of mitochondria in neutrophil apoptosis [123•]. The recent discovery of neutrophil extracellular traps [8••] emphasizes the need to study specific cell death pathways in neutrophils, which may not exist or work the same way in other cell types. There are two core molecular pathways for apoptosis. The “extrinsic” or “death receptor-dependent” pathway is initiated by signals outside the cell and a critical step is signaling via a transmembrane protein with a death receptor domain to caspase 8 inside the cell. The “intrinsic” or “mitochondrial” pathway is initiated by signals inside the cell and a critical step is release of cytochrome c from the mitochondria. The two pathways share the final step of signaling via caspase 3, but otherwise are mostly disjoint.

Neutrophils may be an attractive cell type in which to study apoptosis because the high turnover under normal physiological conditions means that there are lots of apoptosis events to observe and slight perturbations in apoptosis pathway signaling could lead to neutrophil phenotypes that would be hard to observe in longer-lived cells. Therefore, Table 3 summarizes some molecules whose apoptotic roles have been studied in neutrophils specifically. Many of these proteins, including HAX1, participate in the intrinsic/mitochondrial pathway.

Table 3.

Proteins related to apoptosis in neutrophils for which at least one neutrophil-specific study has been published. The pathway could be Intrinsic (a.k.a. mitochondrial), Extrinsic (a.k.a. death receptor), Both (meaning both Intrinsic or Extrinsic), or UPR (meaning unfolded protein response, whose relationship to the Intrinsic and Extrinsic pathways is uncertain). The human/mouse column records whether any of the studies cited in the references column were done on human cells or mouse cells. As suggested by the text associated with Table 2, inferences about pathways of human neutrophil apoptosis that rely on studies done exclusively on mouse cells should be made with extreme caution.

Protein Pro or Anti(-apoptotic) Pathway Reference(s) Mouse or Human
A1-a Anti Intrinsic ? [117] Mouse
Bax Pro (when translocated to the mitochondria) Intrinsic [124127] Both
Bcl-x Anti Intrinsic [83, 118•, 125] Both
Bcl-2 Anti Intrinsic [30, 118•] Both
Bid Pro Intrinsic [127, 128] Human
Bim Pro Intrinsic ? [129] Mouse
Calpain-1 Pro Intrinsic [126] Human
Caspase 3 Pro Both [e.g., 14, 116, 126] Both
CXCR4 Mutations enhance apoptotis ? [84•]; (see [86•] for a partly contradictory study on a transplant model) Human
ELA2 Mutations enhance apoptosis UPR [54, 58•, 130] Human
G-CSF Anti Intrinsic (only?) [14, 19•, 111, 127] Both
GM-CSF Anti ? [19•, 131] Both
G6PC3 Anti UPR? [116] Mouse
G6PT (encoded by SLC37A4) Anti Intrinsic (only?) [132] Human
HAX1 Anti Intrinsic [27••] Human
Mcl-1 Anti Intrinsic [118•, 128] Both
Omi/HTRA2 Pro, but see [133] for a more nuanced perspective Intrinsic [124] Human
SBDS Mutations may enhance apoptosis Extrinsic [134] contradicted by [135] Human
Smac/DIABLO Pro, but see [133] for a more nuanced perspective Intrinsic [124, 126] Human
TNF-α Pro Both [124, 136138] Both

The apparent importance of the mitochondrial pathway in neutrophils may be surprising because neutrophil mitochondria are few in number and hard to see [123•, 124]. In many cell types, mitochondria play at least three important roles: housing the mitochondrial genome, generating energy in the form of ATP, and controlling apoptosis via the intrinsic pathway. The observation of mitochondrial genome deletions in Pearson’s syndrome [48] suggests that the mitochondrial genome is essential to normal neutrophil homeostasis. Whether any proteins encoded by the mitochondrial genome are essential to control neutrophil apoptosis has not been studied, to our knowledge. Several studies suggested that ATP production in neutrophil mitochondria is unimportant because neutrophils get energy from glucose via glycolysis, which is necessary in the microbe-infested, hypoxic micro-environments where neutrophils do their killing [123•, 124, 139]. The reliance on glycolysis over mitochondrial respiration is seen also in eosinophils [140]. The importance of glucose to neutrophils can be understood via the phenotypes of human GSD Ib (Table 1) and mouse knockouts of Slc37a4 and G6pc3 (Table 2). Also in contrast to other cell types, neutrophil mitochondria may have neutrophil-specific roles in phagocytosis and chemotaxis [141].

Increased apoptosis in neutrophils, as for other cell types can be detected for example, by staining for annexin V binding to phosphatidylserine or by measuring mRNA or protein levels of caspase 3. However, these methods do not identify the pathway by which the apoptotic signal cascade reached caspase 3, and this explains some of the question marks in the Pathway column of Table 3. To distinguish mitochondrial pathway apoptosis one can stain with JC-1 and observe the loss of potential, denoted by Δψm, in the mitochondrial inner membranes [e.g., 27••, 141]. Increased mitochondrial apoptosis was seen in HAX1 deficiency [27••] and GSD Ib [132]. Increased apoptosis by other or unknown mechanisms has been seen in WHIM syndrome or models thereof [84•, 86•] and dominant or cyclic SCN associated with ELA2 mutations [58•, 130]. In contrast, TAZ mutations do not cause increased neutrophil apoptosis [142].

For most of the other genes listed in Table 1, apoptosis has not been studied in neutrophils from affected individuals. One intriguing gene in this category is GFI1. Person et al. [33] hypothesized that the transcription factor GFI1 is relevant to neutrophils via an interaction with the DNA for ELA2. However, GFI1 participates in the transcription of other genes, and Nakazawa et al. [143•] recently showed that the domain of GFI1 mutated in SCN plays a role in the repression of pro-apoptotic Bax, suggesting that GFI1 mutations cause SCN via increased apoptosis, unrelated to ELA2 levels.

GSD 1b is one of the more appealing SCN syndromes to study because of the good match between the phenotypes seen in the human disease and animal models (Table 2). As Kuijpers et al. [132] observed excess apoptosis in cells from human patients, Cheung et al. [116] observed excess apoptosis in neutrophils of mice deficient in G6pc3. However, they arrived at possibly divergent conclusions about the pathways that are used in the apoptosis. Kuijpers et al observed translocation of Bax to the mitochondria, which is considered one hallmark of mitochondrial pathway apoptosis. Cheung et al. used Annexin V staining and caspase 3 assays, which cannot determine the pathway, but they speculate that the apoptosis arises due to the lack of glucose, which may cause stress to the endoplasmic reticulum [116]. This possibility is interesting because stress in the endoplasmic reticulum is what causes the unfolded protein response (UPR). Increased UPR has been demonstrated in mutant ELA2 cells [58•]. The unfolded protein response is a danger signal, not an inherent apoptosis signal, which can arise due to a variety of problems in the cellular environment, including low glucose [144]. It has been suggested that the low glucose is problematic in any cell because it interferes with N-linked glycosylation [144]. In neutrophils, low glucose may be an even more severe problem because of the reliance on glycolysis for energy.

In summary, the molecular analysis of patients with SCN has shown that several genes may lead to increased apoptosis of neutrophils, yet the exact pathways of apoptosis remain elusive. Furthermore, we do not yet fully understand how these mutated genes connect separate pathways of cell death.

Discussion

SCN is an exception to two paradigms in molecular medicine:

  1. Identification of molecular causes of genetic diseases precedes the development of effective treatment, and detection of different mutated genes in different patients will lead to individualized treatments (sometimes called “pharmacogenomics”).

  2. When functions and genes are clearly conserved from fish or mice to humans, disrupting the orthologous genes in model organisms will lead to comparable phenotypes.

Table 1 lists 14 genes that can be mutated in a phenotype that includes neutropenia, yet there are few known connections between these genes or the proteins they encode. The genetic heterogeneity in SCN is so extreme that one affected descendant in the Kostmann family has an ELA2 mutation [145], while the rest have a shared homozygous HAX1 mutation [27••].

Identification of the genes mutated in human neutropenia could aid in the engineering and analysis of animal models to understand what the encoded proteins do normally, and what pathways are disrupted when the proteins are missing or mutated. For understanding some neutropenia genes and proteins, such as SBDS, studies in lower organisms such as yeast can be very useful [87••]. Since neutrophils are phagocytic cells and part of the more primitive innate immune system, one might hope for good correspondence between the phenotypes of mice and humans with orthologous mutations. Most encouraging are the excellent phenotypic correspondences between mice and humans with defects in glucose metabolism leading to either nonsyndromic SCN or glycogen storage type Ib. Discouraging is the discordance of phenotypes between mice and humans with heterozygous mutations of ELA2 or homozygous mutations of AP3B1.

The discrepancy between human and mouse models is also made more surprising by recent findings that increasingly implicate excessive apoptosis as a proximal cause of SCN. It remains to be explained why the apoptosis pathways affected in humans behave differently in mice. Another apoptosis-related neutrophil mystery is: Why does expedited apoptosis when neutrophils kill microbes lead to increased neutrophil production, but mutant-ELA2-associated apoptosis and HAX1-deficiency-associated apoptosis do not?

Possible explanations for these conundrums might be found by studying differentiation pathways in neutrophil development. For example, Skokowa et al. recently reported that the transcription factor LEF1 (lymphoid enhancer-binding factor 1) has low levels of expression in SCN and that restoration of expression levels of LEF1 leads to improved granulopoiesis in vitro [146••]. LEF1 controls expression of C/EBPα, an important transcription factor governing myelopoiesis. Moreover, LEF1 can be speculatively connected to the transcription of several genes mutated in SCN; for example, the promoter of ELA2 has a binding site for LEF1 [147]. If the targets of LEF1 protein or the timing of LEF1 expression differ between mice and humans this could help explain the phenotypic discrepancies.

Another possible partial explanation for these conundrums is that the ANC is determined by poorly understood signaling pathways more so than by apoptosis pathways. Mathematical modeling suggests that cyclic neutropenia is related to a defect in one or more feedback loops [148]. The facts that mutations in ELA2 can cause either SCN or cyclic neutropenia and AP3B1 deficiency causes cyclic neutropenia in dogs, suggest that feedback loops defective in cyclic neutropenia may be more severely damaged in and relevant to SCN. If G-CSF acts by amplifying a signaling pathway in which the mechanism of apoptosis is irrelevant, as modeled for example in [149], then this could explain why patients with so many different genetic causes of SCN benefit from G-CSF. Identifying the participants in such a signaling pathway could be aided by finding what genes are mutated in those SCN patients who do not respond to G-CSF. However, keeping those patients alive is a clinical challenge.

In sum, severe congenital neutropenia can be caused by defects in a variety of single genes and pathways. Surprisingly, many of these mutated genes, such as the recently identified HAX1 [27••] and p14 [28••], are expressed in a variety of cell types, not just neutrophils or just leukocytes or just immunity-related cells. The neutrophil specificity of the resulting phenotypes appears to be due to the high turnover and short lifespan of neutrophils, making them more sensitive to defective pathways of proliferation and especially apoptosis. Therefore, laboratory studies of naturally occurring human forms of SCN and engineered animal models are yielding new general molecular insights into birth and death of both neutrophils and other types of cells.

Acknowledgments

Our neutropenia research has been supported by a grant from the Deutsche Forschungsgemeinschaft (DFG-KliFo 110), José-Carreras-Leukemia-Foundation, Bundesministerium für Bildung und Forschung (Congenital Bone Marrow Failure Syndromes) and in part by the Intramural Research program of the NIH, NLM. This work was supported by a fellowship from the Yamagiwa Yoshida foundation. We thank Karl Welte for critical review of this paper. We thank Bodo Grimbacher for initiating and encouraging our collaboration.

References and recommended reading

Papers of particular interest, published after 31 December 2005, have been highlighted as:

• of special interest

•• of outstanding interest

  • 1.Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol. 2006 Jun;:173–182. doi: 10.1038/nri1785. [DOI] [PubMed] [Google Scholar]
  • 2.Segal AW. How neutrophils kill microbes. Ann Rev Immunol. 2005;23:197–223. doi: 10.1146/annurev.immunol.23.021704.115653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Skokowa J, Welte K. LEF-1 is a decisive transcription factor in neutrophil granulopoiesis. Ann NY Acad Sci. 2007;1106:143–151. doi: 10.1196/annals.1392.012. [DOI] [PubMed] [Google Scholar]
  • 4.Lord BI, Bronchud MH, Owens S, Chang J, Howell A, Souza L, Dexter TM. The kinetics of human granulopoiesis following treatment with granulocyte colony-stimulating factor in vivo. Proc Natl Acad Sci USA. 1989;86:9499–9503. doi: 10.1073/pnas.86.23.9499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Mackey MC, Aprikyan AAG, Dale DC. The rate of apoptosis in post mitotic and neutrophil precursors of normal and neutropenic humans. Cell Prolif. 2003;36:27–34. doi: 10.1046/j.1365-2184.2003.00251.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kobayashi SD, Braughton KR, Whitney AR, Voyich JM, Schwan TG, Musser JM, DeLeo FR. Bacterial pathogens modulate an apoptosis differentiation program in human neutrophils. Proc Natl Acad Sci USA. 2003;100:10948–10953. doi: 10.1073/pnas.1833375100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Watson RW, Redmond HP, Wang JH, Condron C, Bouchier-Hayes D. Neutrophils undergo apoptosis following ingestion of Escherichia coli. J Immunol. 1996;156:3986–3992. [PubMed] [Google Scholar]
  • 8.Fuchs TA, Abed U, Goosmann C, Hurwitz R, Schulze I, Wahn V, Weinrauch Y, Brinkmann V, Zychlinsky Novel cell death program leads to neutrophil extracellular traps. J Cell Biol. 2007;176:231–241. doi: 10.1083/jcb.200606027. [Neutrophils construct “neutrophil extracellular traps” “NETs” as yet another mechanism to kill microbes. NETs are released as the cell membrane breaks, which has the additional virtue that the neutrophil does not stay around to create inflammation. Neutrophils in patients with chronic granulomatous disease, an immunodeficiency in which neutrophils are deficient in function but not in number (ANC), cannot make NETs or die by this pathway.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Haddy TB, Rana SR, Castro O. Benign ethnic neutropenia: what is a normal absolute neutrophil count? J Lab Clin Med. 1999;133:15–22. doi: 10.1053/lc.1999.v133.a94931. [DOI] [PubMed] [Google Scholar]
  • 10.Souza LM, Boone TC, Gabrilove J, Lai PH, Zsebo KM, Murdock DC, Chazin VR, Bruszewski J, Lu H, Chen KK, Barendt J, Platzer E, Moore MAS, Mertelsmann R, Welte K. Recombinant human granulocyte colony stimulating factor: effects on normal and leukemic myeloid cells. Science. 1986;232:61–65. doi: 10.1126/science.2420009. [DOI] [PubMed] [Google Scholar]
  • 11.Bonilla MA, Gillio AP, Ruggiero M, Kernan NA, Brochstein JA, Abboud M, Fumagalli L, Vincent M, Gabrilove JL, Welte K, Souza LM, O’Reilly RJ. Effects of recombinant human granulocyte colony-stimulating factor on neutropenia in patients with congenital agranulocytosis. New Engl J Med. 1989;320:1574–1580. doi: 10.1056/NEJM198906153202402. [DOI] [PubMed] [Google Scholar]
  • 12.Anderlini P, Przepiorka D, Champlin R, Körbling M. Biologic and clinical effects of granulocyte colony stimulating factor in normal individuals. Blood. 1996;88:2819–2825. [PubMed] [Google Scholar]
  • 13.Rex JH, Bhalla SC, Cohen DM, Hester JP, Vartivarian SE, Anaissie EJ. Protection of human polymorphonuclear leukocyte function from the deleterious effects of isolation, irradiation, and storage by interferon-γ and granulocyte-colonystimulating factor. Transfusion. 1995;35:605–611. doi: 10.1046/j.1537-2995.1995.35795357886.x. [DOI] [PubMed] [Google Scholar]
  • 14.Maianksi NA, Mul FPJ, van Buul JD, Roos D, Kuijpers TW. Granulocyte colonystimulating factor inhibits the mitochondria-dependent activation of caspase-3 in neutrophils. Blood. 2002;99:672–679. doi: 10.1182/blood.v99.2.672. [DOI] [PubMed] [Google Scholar]
  • 15.Dale DC, Bonilla MA, Davis MW, Nakanishi AM, Hammond WP, Kurtzberg J, Wang W, Jakubowski A, Winton E, Lalezari P, Robinson W, Glaspy JA, Emerson S, Gabrilove J, Vincent M, Boxer LA. A randomized controlled phase III trial of recombinant human granulocyte colony-stimulating factor (filgrastim) for treatment of severe chronic neutropenia. Blood. 1993;81:2496–2502. [PMC free article] [PubMed] [Google Scholar]
  • 16.Rosenberg PS, Alter BP, Bolyard AA, Bonilla MA, Boxer LA, Cham B, Fier C, Freedman M, Kannourakis G, Kinsey S, Schwinzer B, Zeidler C, Welte K, Dale DC. for the Severe Chronic Neutropenia International Registry: The incidence of leukemia and mortality from sepsis in patients with severe congenital neutropenia receiving longterm G-CSF therapy. Blood. 2006;107:4628–4635. doi: 10.1182/blood-2005-11-4370. [After the introduction of granulocyte-colony stimulating factor therapy, it is estimated that the ten-year incidence for sepsis decreased to 8%, but the ten-year incidence of myelodysplastic syndrome or acute myeloid leukemia (MDS/AML) increased to 21%. Those patients receiving higher doses of G-CSF have a higher incidence of MDS/AML.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hurst D, Kilpatrick L, Becker J, Lipani J, Kleman K, Perrine S, Douglas SD. Recombinant human GM-CSF treatment of neutropenia in glycogen storage disease-1b. Am J Pediatr Hematol Oncol. 1993;15:71–76. doi: 10.1097/00043426-199302000-00008. [DOI] [PubMed] [Google Scholar]
  • 18.Welte K, Zeidler C, Reiter A, Müller W, Odenwald E, Souza L, Riehm H. Differential effects of granulocyte-macrophage colony-stimulating factor and granulocyte colony-stimulating factor in children with severe congenital neutropenia. Blood. 1990;75:1056–1063. [PubMed] [Google Scholar]
  • 19.Wolach B, van der Laan LJW, Maianski NA, Tool ATJ, van Bruggen R, Roos D, Kuijpers TW. Growth factors G-CSF and GM-CSF differentially preserve chemotaxis of neutrophils aging in vitro. Exp Hematol. 2007;35:541–550. doi: 10.1016/j.exphem.2006.12.008. [G-CSF is more effective than GM-CSF at preserving chemotaxis and delaying apoptosis in cultured neutrophils.] [DOI] [PubMed] [Google Scholar]
  • 20.Welte K, Zeidler C, Dale DC. Severe congential neutropenia. Semin Hematol. 2006;43:189–195. doi: 10.1053/j.seminhematol.2006.04.004. [DOI] [PubMed] [Google Scholar]
  • 21.Gilman PA, Jackson DP, Guild HG. Congenital agranulocytosis: prolonged survival and terminal acute leukemia. Blood. 1970;36:576–585. [PubMed] [Google Scholar]
  • 22.Dong F, Hoefsloot LH, Schelen AM, Broeders LCAM, Meijer Y, Veerman AJP, Touw IP, Löwenberg B. Identification of a nonsense mutation in the granulocytecolony-stimulating factor receptor in severe congenital neutropenia. Proc Natl Acad Sci USA. 1994;91:4480–4484. doi: 10.1073/pnas.91.10.4480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Dong F, Byrnes RK, Tidow N, Welte K, Löwenberg B, Touw IP. Mutations in the gene for the granulocyte colony-stimulating-factor receptor in patients with acute myeloid leukemia preceded by severe congenital neutropenia. N Engl J Med. 1995;333:487–493. doi: 10.1056/NEJM199508243330804. [DOI] [PubMed] [Google Scholar]
  • 24.Germeshausen M, Ballmaier M, Welte K. Incidence of CSF3R mutations in severe congenital neutropenia and relevance for leukemogenesis: results of a long-term survey. Blood. 2007;109:93–99. doi: 10.1182/blood-2006-02-004275. [DOI] [PubMed] [Google Scholar]
  • 25.Mermel CH, McLemore ML, Liu F, Pereira S, Woloszynek J, Lowell CA, Link DC. Src family kinases are important negative regulators of G-CSF-dependent granulopoiesis. Blood. 2006;108:2562–2568. doi: 10.1182/blood-2006-05-024307. [Src family kinases are Hck and Lyn provide negative regulation at different stages of granulopoiesis. The authors propose that CSF3R mutations may be leukemogenic because signaling between CSF3R and the kinases is lost.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sloand EM, Yong ASM, Ramkissoon S, Solomou E, Bruno TC, Kim S, Fuhrer M, Kajigaya S, Barrett AJ, Young NS. Granulocyte colony-stimulating factor preferentially stimulates proliferation of monosomy 7 cells bearing the isoform IV receptor. Proc Natl Acad Sci USA. 2006;103:14483–1488. doi: 10.1073/pnas.0605245103. [In vitro, G-CSF promotes the proliferation of cells with monosomy for human chromosome 7 over cells with the normal two copies. This could be a factor in leukemogenesis in SCN patients treated with G-CSF. It is proposed that transcription factor CDP/cut, whose official name is CUTL1, is the gene on chromosome 7 whose low dosage is problematic.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Klein C, Grudzien M, Appaswamy G, Germeshausen M, Sandrock I, Schäffer AA, Rathinam C, Boztug K, Zeidler C, Schwinzer B, Rezaei N, Bohn G, Melin M, Carlsson G, Fadeel B, Dahl N, Palmblad J, Henter J-I, Grimbacher B, Welte K. Deficiency of HAX1 causes autosomal recessive severe congenital neutropenia (Kostmann disease) Nat Genet. 2007;39:86–92. doi: 10.1038/ng1940. [Some cases of autosomal recessive SCN are caused by homozygous mutations in HAX1, which encodes a protein that inhibits apoptosis. This includes three surviving affected descendants of the original SCN family published by Kostmann [60]. HAX1 stabilizes the inner mitochondrial membrane potential, yet the exact mechanism by which neutrophil apoptosis is prevented remains to be determined.] [DOI] [PubMed] [Google Scholar]
  • 28.Bohn G, Allroth A, Brandes G, Thiel J, Glocker E, Schäffer AA, Rathinam C, Taub N, Teis D, Zeidler C, Dewey RA, Geffers R, Buer J, Huber LA, Welte K, Grimbacher B, Klein C. A novel human primary immunodeficiency syndrome caused by deficiency of the endosomal adaptor protein p14. Nat Med. 2007;13:38–45. doi: 10.1038/nm1528. [Four siblings affected with a syndrome including neutropenia, low memory B cell counts, short stature, hypopigmentation, and coarse facial features have a homozygous missense mutation in the 3’ untranslated region of the gene encoding p14, and endosomal adaptor protein.] [DOI] [PubMed] [Google Scholar]
  • 29.Cilenti L, Soundarapandian MM, Kyriazis GA, Stratico V, Singh S, Gupta S, Bonventre JV, Alnemri ES, Zervos AS. Regulation of HAX-1 anti-apoptotic protein by Omi/HtrA2 protease during cell death. J Biol Chem. 2004;279:50295–50301. doi: 10.1074/jbc.M406006200. [DOI] [PubMed] [Google Scholar]
  • 30.Carlsson G, Aprikyan AAG, Tehranchi R, Dale DC, Porwit A, Hellstrom-Lindberg E, Palmblad J, Henter J-I, Fadeel B. Kostmann syndrome: severe congenital neutropenia associated with defective expression of Bcl-2, constitutive mitochondrial release of cytochrome c, and excessive apoptosis of myeloid progenitor cells. Blood. 2004;103:3355–3361. doi: 10.1182/blood-2003-04-1011. [DOI] [PubMed] [Google Scholar]
  • 31.Grenda DS, Link DC. Mechanisms of disordered granulopoiesis in congenital neutropenia. Curr Top Dev Biol. 2006;74:133–176. doi: 10.1016/S0070-2153(06)74005-4. [This review is well-suited for readers who want more information on any of the neutropenia syndromes that we covered superficially.] [DOI] [PubMed] [Google Scholar]
  • 32.Dale DC, Person RE, Bolyard AA, Aprikyan AG, Bos C, Bonilla MA, Boxer LA, Kannourakis G, Zeidler C, Welte K, Benson KF, Horwitz M. Mutations in the gene encoding neutrophil elastase in congenital and cyclic neutropenia. Blood. 2000;96:2317–2322. [PubMed] [Google Scholar]
  • 33.Person RE, Li F-Q, Duan Z, Benson KF, Wechsler J, Papadaki HA, Eliopoulos G, Kaufman C, Bertolone SJ, Nakamoto B, Papayannopoulou T, Grimes HL, Horwitz M. Mutations in proto-oncogene GFI1 cause human neutropenia and target ELA2. Nat Genet. 2003;34:308–312. doi: 10.1038/ng1170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Devriendt K, Kim AS, Mathijs G, Frints SGM, Schwartz M, Van den Oord JJ, Verhoef GEG, Boogaerts MA, Fryns J-P, You D, Michael K, Rosen MK, Vandenberghe P. Constitutively activating mutation in WASP causes X-linked severe congenital neutropenia. Nat Genet. 2001;27:313–317. doi: 10.1038/85886. [DOI] [PubMed] [Google Scholar]
  • 35.Ancliff PJ, Blundell MP, Cory GO, Calle Y, Worth A, Kempski H, Burns S, Jones GE, Sinclair J, Kinnon C, Hann IM, Gale RE, Linch DC, Thrasher AJ. Two novel activating mutations in the Wiskott-Aldrich syndrome protein result in congenital neutropenia. Blood. 2006;108:2182–2189. doi: 10.1182/blood-2006-01-010249. [Two male patients with SCN have activating mutations in WAS on the X chromosome. This confirms and extends the results of [34] showing that the original mutation and family are not unique.] [DOI] [PubMed] [Google Scholar]
  • 36.Horwitz M, Benson KF, Person RE, Aprikyan AG, Dale DC. Mutations in ELA2, encoding neutrophil elastase, define a 21-day biological clock in cyclic haematopoiesis. Nat Genet. 1999;23:433–436. doi: 10.1038/70544. [DOI] [PubMed] [Google Scholar]
  • 37.Gerin I, Veiga-da-Cunha M, Achouri Y, Collet JF, Van Schaftingen E. Sequence of a putative glucose 6-phosphate translocase, mutated in glycogen storage disease type Ib. FEBS Lett. 1997;419:235–238. doi: 10.1016/s0014-5793(97)01463-4. [DOI] [PubMed] [Google Scholar]
  • 38.Dell'Angelica EC, Shotelersuk V, Aguilar RC, Gahl WA, Bonifacino JS. Altered trafficking of lysosomal proteins in Hermansky-Pudlak syndrome due to mutations in the β3A subunit of the AP-3 adaptor. Mol Cell. 1999;3:11–21. doi: 10.1016/s1097-2765(00)80170-7. [DOI] [PubMed] [Google Scholar]
  • 39.Ménasché G, Pastural E, Feldmann J, Certain S, Ersoy F, Dupuis S, Wulffraat N, Bianchi D, Fischer A, Le Deist F, de Saint Basile G. Mutations in RAB27A cause Griscelli syndrome associated with haemophagocytic syndrome. Nat Genet. 2000;25:173–176. doi: 10.1038/76024. [DOI] [PubMed] [Google Scholar]
  • 40.Nagle DL, Karim MA, Woolf EA, Holmgren L, Bork P, Misumi DJ, McGrail SH, Dussault BJ, Jr, Perou CM, Boissy RE, Duyk GM, Spritz RA, Moore KJ. Identification and mutation analysis of the complete gene for Chediak–Higashi syndrome. Nat Genet. 1996;14:307–311. doi: 10.1038/ng1196-307. [DOI] [PubMed] [Google Scholar]
  • 41.Barbosa MDFS, Nguyen QA, Tchernev 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;382:262–265. doi: 10.1038/382262a0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Perou CM, Moore KJ, Nagle DL, Misumi DJ, Woolf EA, McGrail SH, Lisa HolmgrenL, Brody TH, Dussault BJ, Jr, Monroe CA, Duyk GM, Pryor RJ, Li L, Justice MJ, Jerry Kaplan J. Identification of the murine beige gene by YAC complementation and positional cloning. Nat Genet. 1996;13:303–308. doi: 10.1038/ng0796-303. [DOI] [PubMed] [Google Scholar]
  • 43.Hernandez PA, Gorlin RJ, Lukens JN, Shoichiro Taniuchi S, Bohinjec J, Francois F, Klotman ME, Diaz GA. Mutations in the chemokine receptor gene CXCR4 are associated with WHIM syndrome, a combined immunodeficiency disease. Nat Genet. 2003;34:70–74. doi: 10.1038/ng1149. [DOI] [PubMed] [Google Scholar]
  • 44.Kolehmainen J, Black GCM, Saarinen A, Chandler K, Clayton-Smith J, Träskelin AL, Perveen R, Kivitie-Kallio S, Norio R, Warburg M, Fryns J-P, de la Chapelle A, Lehesjoki A-E. Cohen syndrome is caused by mutations in a novel gene, COH1, encoding a transmembrane protein with a presumed role in vesicle-mediated sorting and intracellular protein transport. Am J Hum Genet. 2003;72:1359–1369. doi: 10.1086/375454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Boocock GRB, Morrison JA, Popovic M, Richards N, Ellis L, Durie PR, Rommens JM. Mutations in SBDS are associated with Shwachman–Diamond syndrome. Nat Genet. 2003;33:97–101. doi: 10.1038/ng1062. [DOI] [PubMed] [Google Scholar]
  • 46.Bione S, D’Adamo P, Maestrini E, Gedeon AK, Bolhuis PA, Toniolo D. A novel Xlinked gene, G4.5. is responsible for Barth syndrome. Nat Genet. 1996;12:385–389. doi: 10.1038/ng0496-385. [DOI] [PubMed] [Google Scholar]
  • 47.Ridanpää M, van Eenennaam H, Pelin K, Chadwick R, Johnson C, Yuan B, van Venrooij W, Pruijn G, Salmela R, Rockas S, Mäkitie O, Kaitila I, de la Chapelle A. Mutations in the RNA component of RNase MRP cause a pleiotropic human disease, cartilage-hair hypoplasia. Cell. 2001;104:195–203. doi: 10.1016/s0092-8674(01)00205-7. [DOI] [PubMed] [Google Scholar]
  • 48.Rotig A, Colonna M, Bonnefont JP, Blanche S, Fischer A, Saudubray JM, Munnich A. Mitochondrial DNA deletion in Pearson's marrow/pancreas syndrome. Lancet I. 1989;(issue 8643):902–903. doi: 10.1016/s0140-6736(89)92897-3. [DOI] [PubMed] [Google Scholar]
  • 49.Hochberg JC, Miron PM, Hay BN, Woda BA, Wang SA, Richert-Przygonska M, Aprikyan AAG, Newburger PE. Pediatr Blood Cancer. Mosaic tetraploidy and transient GFI1 mutation in a patient with severe chronic neutropenia. in press. [ This paper describes a father-son pair with mild neutropenia and mosaic tetraploidy of some myeloid cells. Sequencing of genomic DNA from the son showed a reproducible GFI1 mutation in one sample, but not in other samples. The authors speculate that both the tetraploidy and the neutropenia are due to some defect in mitosis.] [DOI] [PubMed] [Google Scholar]
  • 50.Horwitz MS, Duan Z, Korkmaz B, Lee H-H, Mealiffe ME, Salipante SJ. Neutrophil elastase in cyclic and severe congenital neutropenia. Blood. 2007;109:1817–1824. doi: 10.1182/blood-2006-08-019166. [This review proposes a central role for NE (encoded by ELA2) in the etiology of SCN. It discusses genotype/phenotype correlations distinguishing between mutations associated with SCN that could lead to general mis-trafficking anywhere in the cell and apoptosis, while cyclic neutropenia mutations might lead to excess granular accumulation. Failure of mutant NE to cleave some target protein could lead to disruption of a feedback loop and cyclic neutropenia. One possible target is N2N [53].] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Briars GL, Parry HF, Ansari BM. Dominantly inherited severe congenital neutropenia. J Infect. 1996;33:123–126. doi: 10.1016/s0163-4453(96)93081-9. [DOI] [PubMed] [Google Scholar]
  • 52.Boxer LA, Stein S, Buckley D, Bolyard AA, Dale DC. Strong evidence for autosomal dominant inheritance of severe congenital neutropenia associated with ELA2 mutations. J Pediatr. 2006;148:633–636. doi: 10.1016/j.jpeds.2005.12.029. [Five different children with four different mothers are all affected with SCN and are heterozygous for the same S97L mutation in the gene ELA2. Subsequent investigation showed that the father contributed sperm to a sperm bank, which was used to impregnate all the mothers. This is interpreted as strong evidence for autosomal dominant transmission since the mothers have different genetic material. However, it is unclear why the father was not diagnosed with SCN; perhaps, he is a germline mosaic.] [DOI] [PubMed] [Google Scholar]
  • 53.Duan Z, Li F-Q, Wechsler J, Meade-White K, Williams K, Benson KF, Horwitz M. A novel notch protein, N2N, targeted by neutrophil elastase and implicated in hereditary neutropenia. Mol Cell Biol. 2004;24:58–70. doi: 10.1128/MCB.24.1.58-70.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hunter MG, Druhan LJ, Pam R, Massullo PR, Avalos BR. Proteolytic cleavage of granulocyte colony-stimulating factor and its receptor by neutrophil elastase induces growth inhibition and decreased cell surface expression of the granulocyte colony-stimulating factor receptor. Am J Path. 2003;74:149–155. doi: 10.1002/ajh.10434. [DOI] [PubMed] [Google Scholar]
  • 55.Li F-Q, Horwitz M. Characterization of mutant neutrophil elastase in severe congenital neutropenia. J Biol Chem. 2001;276:14230–14241. doi: 10.1074/jbc.M010279200. [DOI] [PubMed] [Google Scholar]
  • 56.Massullo P, Druhan LJ, Bunnell BA, Hunter MG, Robinson JM, Marsh CB, Avalos BR. Aberrant subcellular targeting of the G185R neutrophil elastase mutant associated with severe congenital neutropenia induces premature apoptosis of differentiating promyelocytes. Blood. 2005;105:3397–3404. doi: 10.1182/blood-2004-07-2618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Thusberg J, Vihinen M. Bioinformatic analysis of protein structure-function relationships: case study of leukocyte elastase (ELA2) missense mutations. Hum Mutat. 2006;27:1230–1243. doi: 10.1002/humu.20407. [This paper uses over 30 computational tools to assess the effects of 32 missense mutations in the ELA2 protein. The authors predict which mutations affect a conserved residue, change the electrostatic surface potential, affect critical contacts, cause the protein to become disordered, change the protein structure in other ways, etc.] [DOI] [PubMed] [Google Scholar]
  • 58.Köllner I, Sodeik B, Schreek S, Heyn H, Neuhoff N, Germeshausen M, Zeidler C, Kruger M, Schlegelberger B, Welte K, Beger C. Mutations in neutrophil elastase causing congenital neutropenia lead to cytoplasmic protein accumulation and induction of the unfolded protein response. Blood. 2006;108:493–500. doi: 10.1182/blood-2005-11-4689. [This paper addresses the question of how diverse heterozygous mutations in ELA2 cause neutropenia. The mutations all cause the ELA2 protein encoded by the mutant allele to be aberrant in some structural sense (see [57•]) and accumulate in the cytoplasm where it does not belong. This accumulation of aberrant ELA2 triggers apoptosis via the unfolded protein response, regardless of the fact that the other allele encodes wild type ELA2.] [DOI] [PubMed] [Google Scholar]
  • 59.Notarangelo LD, Notarangelo LD, Ochs HD. WASP and the phenotypic range associated with deficiency. Curr Opin Allergy Clin Immunol. 2005;5:485–490. doi: 10.1097/01.all.0000191243.25757.ce. [DOI] [PubMed] [Google Scholar]
  • 60.Kostmann R. Infantile genetic agranulocytosis (Agranulocystosis infantilis hereditaria): a new recessive lethal disease in man. Acta Paediatr. 1956;45 (Suppl.):1–78. [PubMed] [Google Scholar]
  • 61.Suzuki Y, Demoliere C, Kitamura D, Takeshita H, Deuschle U, Watanabe T. HAX-1, a novel intracellular protein, localized on mitochondria, directly associates with HS1, a substrate of Src family tyrosine kinases. J Immunol. 1997;158:2736–2744. [PubMed] [Google Scholar]
  • 62.Stinchcombe J, Bossi G, Griffiths GM. Linking albinism and immunity: the secrets of secretory lysosomes. Science. 2004;305:55–59. doi: 10.1126/science.1095291. [DOI] [PubMed] [Google Scholar]
  • 63.Hermansky F, Pudlak P. Albinism associated with hemorrhagic diathesis and unusual pigmented reticular cells in the bone marrow: report of two cases with histochemical studies. Blood. 1959;14:162–169. [PubMed] [Google Scholar]
  • 64.Benson KF, Li F-Q, Person RE, Albani D, Duan Z, Wechsler J, Meade-White K, Williams K, Acland GM, Niemeyer G, Lothrop CD, Horwitz M. Mutations associated with neutropenia in dogs and humans disrupt intracellular transport of neutrophil elastase. Nat Genet. 2003;35:90–96. doi: 10.1038/ng1224. [DOI] [PubMed] [Google Scholar]
  • 65.Fontana S, Parolini S, Vermi W, Booth S, Gallo F, Donini M, Benassi M, Gentili F, Ferrari D, Notarangelo LD, Cavadini P, Marcenaro E, Dusi S, Cassatella M, Facchetti F, Griffiths GM, Moretta A, Notarangelo LD, Badolato R. Innate immunity defects in Hermansky-Pudlak type 2 syndrome. Blood. 2006;107:4857–4864. doi: 10.1182/blood-2005-11-4398. [DOI] [PubMed] [Google Scholar]
  • 66.Jung J, Bohn G, Allroth A, Boztug K, Brandes G, Sandrock I, Schäffer AA, Rathinam C, Köllner I, Beger C, Schilke R, Welte K, Grimbacher B, Klein C. Identification of a homozygous deletion in the AP3B1 gene causing Hermansky-Pudlak syndrome, type 2. Blood. 2006;108:362–369. doi: 10.1182/blood-2005-11-4377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Enders A, Zieger B, Schwarz K, Yoshimi A, Speckmann C, Knoepfle E-M, Kontny U, Muller C, Nurden A, Rohr J, Henschen M, Pannicke U, Niemeyer C, Nurden P, Ehl S. Lethal hemophagocytic lymphohistiocytosis in Hermansky-Pudlak syndrome type II. Blood. 2006;108:81–87. doi: 10.1182/blood-2005-11-4413. [DOI] [PubMed] [Google Scholar]
  • 68.Huizing M, Scher CD, Strovel E, Fitzpatrick DL, Hartnell LM, Anikster Y, Gahl WA. Nonsense mutations in ADTB3A cause complete deficiency of the β3A subunit of adaptor complex-3 and severe Hermansky-Pudlak syndrome type 2. Pediatr Res. 2002;51:150–158. doi: 10.1203/00006450-200202000-00006. [DOI] [PubMed] [Google Scholar]
  • 69.Kotzot D, Richter K, Gierth-Fiebig K. Oculocutaneous albinism, immunodeficiency, hematological disorders, and minor anomalies: a new autosomal recessive syndrome? Am J Med Genet. 1994;50:224–227. doi: 10.1002/ajmg.1320500303. [DOI] [PubMed] [Google Scholar]
  • 70.Griscelli C, Durandy A, Guy-Grand D, Daguillard F, Herzog C, Prunieras M. A syndrome associating partial albinism and immunodeficiency. Am J Med. 1978;65:691–702. doi: 10.1016/0002-9343(78)90858-6. [DOI] [PubMed] [Google Scholar]
  • 71.Klein C, Phillipe N, Le Deist F, Fraitag S, Prost C, Durandy A, Fischer A, Griscelli C. Partial albinism with immunodeficiency (Griscelli syndrome) J Pediatr. 1994;125:886–895. doi: 10.1016/s0022-3476(05)82003-7. [DOI] [PubMed] [Google Scholar]
  • 72.Munafó DB, Johnson JL, Ellis BA, Rutschmann S, Beutler B, Catz SD. Rab27a is a key component of the secretory machinery of azurophilic granules in granulocytes. Biochem J. 2007;402:229–239. doi: 10.1042/BJ20060950. [RAB27A in conjunction with SYTL1 (previously called JFC1/Slp1) (synaptotagmin-like protein 1) plays a key role in the release of myeloperoxidase (MPO) from azurophilic granules of neutrophils. This explains why neutrophils from patients with Griscelli syndrome type 2, caused by mutations in RAB27A, have defective bacterial killing. It does not directly explain the neutropenia, however.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Beguez-Cesar AB. Neutropenia cronica maligna familiar con granulaciones atipicas de los leucocitos. Bol Soc Cubana Pediat. 1943;15:900–922. [Google Scholar]
  • 74.Chediak M. Nouvelle anomalie leucocytaire de caractere constitutionnel et familial. Rev Hemat. 1952;7:362–367. [PubMed] [Google Scholar]
  • 75.Higashi O. Congenital gigantism of peroxidase granules: the first case ever reported of qualitative abnormity of peroxidase. Tohoku J Exp Med. 1954;59:315–332. doi: 10.1620/tjem.59.315. [DOI] [PubMed] [Google Scholar]
  • 76.Kjeldsen L, Calafat J, Borregaard N. Giant granules of neutrophils in Chediak-Higashi syndrome are derived from azurophil granules but not from specific and gelatinase granules. J Leukoc Biol. 1998;64:72–77. doi: 10.1002/jlb.64.1.72. [DOI] [PubMed] [Google Scholar]
  • 77.Karim MA, Suzuki K, Fukai K, Oh J, Nagle DL, Moore KJ, Barbosa E, Falik-Borenstein T, Filipovich A, Ishida Y, Kivrikko S, Klein C, Kreuz F, Levin A, Miyajima H, Regueiro J, Russo C, Uyama E, Outi Vierimaa O, Spritz RA. Apparent genotypephenotype correlation in childhood, adolescent, and adult Chediak-Higashi syndrome. Am J Med Genet. 2002;108:16–22. [PubMed] [Google Scholar]
  • 78.Teis D, Taub N, Kurzbauer R, Hilber D, de Araujo ME, Erlacher M, Offterdinger M, Villunger A, Geley S, Bohn G, Klein C, Hess MW, Huber LA. p14–MP1-MEK1 signaling regulates endosomal traffic and cellular proliferation during tissue homeostasis. J Cell Biol. 2006;175:861–868. doi: 10.1083/jcb.200607025. [A homozygous knockout of p14 conditional to the epidermis is embryonic lethal in mice. The p14 protein forms a scaffold along with MEK1 (mitogen-activated protein kinase and ERK kinase 1) and MP1 (MEK1 partner 1) that aid trafficking other proteins to late endosomes and in ERK (extracellular signal-regulated kinase) signaling that is essential for cellular proliferation. Since Bohn et al. [28••] found p14 deficiency in one set of siblings with an albinism+neutropenia syndrome, defects in p14 or related proteins may be suspected in other patients with clinically overlapping syndromes. Assays developed in this paper may aid in determining whether late endosomal trafficking is defective in patients with albinism+neutropenia of unknown molecular etiology before sequencing of specific genes is undertaken.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wunderlich W, Fialka I, Teis D, Alpi A, Pfeifer A, Parton RG, Lottspeich F, Huber LA. A novel 14-kilodalton protein interacts with the mitogen-activated protein kinase scaffold MP1 on a late endosomal/lysosomal compartment. J Cell Biol. 2001;152:765–776. doi: 10.1083/jcb.152.4.765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Teis D, Wunderlich W, Huber LA. Localization of the MP1-MAPK scaffold complex to endosomes is mediated by p14 and required for signal transduction. Dev Cell. 2002;3:803–814. doi: 10.1016/s1534-5807(02)00364-7. [DOI] [PubMed] [Google Scholar]
  • 81.Gorlin RJ, Gelb B, Diaz GA, Lofsness KG, Pittelkow MR, Fenyk JR., Jr WHIM syndrome, an autosomal dominant disorder: Clinical, hematological, and molecular studies. Am J Med Genet. 2000;91:368–376. [PubMed] [Google Scholar]
  • 82.Wetzler M, Talpaz M, Kellagher MJ, Gutterman JU, Kurzrock R. Myelokathexis: normalization of neutrophil counts and morphology by GM-CSF. JAMA. 1992;267:2179–2180. [PubMed] [Google Scholar]
  • 83.Aprikyan AAG, Liles WC, Park JR, Jonas M, Chi EY, Dale DC. Myelokathexis, a congenital disorder of severe neutropenia characterized by accelerated apoptosis and defective expression of bcl-x in neutrophil precursors. Blood. 2000;95:320–327. [PubMed] [Google Scholar]
  • 84.Sanmun D, Garwicz D, Smith CIE, Palmblad J, Fadeel B. Stromal-derived factor-1 abolishes constitutive apoptosis of WHIM syndrome neutrophils harbouring a truncating CXCR4 mutation. Br J Haematol. 2006;134:640–644. doi: 10.1111/j.1365-2141.2006.06240.x. [Stromal-derived factor-1 (SDF1) eliminates the excessive apoptosis in cells from a WHIM patient, but not in cells from SCN patients, indicating heterogeneous use of apoptosis pathways. SDF-1 acts through the MEK1 and PI3K pathways, while G-CSF inhibits apoptosis via the PI3K and JNK pathways.] [DOI] [PubMed] [Google Scholar]
  • 85.Kim HK, De La Luz Sierra M, Williams CK, Gulino AV, Tosato G. G-CSF downregulation of CXCR4 expression identified as a mechanism for mobilization of myeloid cells. Blood. 2006a;108:812–820. doi: 10.1182/blood-2005-10-4162. [G-CSF reduces the expression of CXCR4. Combined with the fact that activating mutations of CXCR4 cause WHIM syndrome, this new finding helps explain why G-CSF is effective in treating the myelokathexis in WHIM syndrome.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kawai T, Choi U, Cardwell L, DeRavin SS, Naumann N, Whiting-Theobald NL, Linton GF, Moon J, Philip M, Murphy PM, Malech HL. WHIM syndrome myelokathexis reproduced in the NOD/SCID mouse xenotransplant model engrafted with healthy human stem cells transduced with C-terminus–truncated CXCR4. Blood. 2007;109:78–84. doi: 10.1182/blood-2006-05-025296. [In a mouse xenotransplant model, cells with mutant CXCR4 have increased apoptosis, but this effect was not seen in culture. This seems to be contradictory to [84•].] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Cohen MM, Jr, Hall BD, Smith DW, Graham CB, Lampert KJ. A new syndrome with hypotonia, obesity, mental deficiency, and facial, oral, ocular and limb anomalies. J Pediatr. 1973;83:280–284. doi: 10.1016/s0022-3476(73)80493-7. [DOI] [PubMed] [Google Scholar]
  • 88.Norio R, Raitta C, Lindahl E. Further delineation of the Cohen syndrome; report on chorioretinal dystrophy, leukopenia and consanguinity. Clin Genet. 1984;25:1–14. doi: 10.1111/j.1399-0004.1984.tb00456.x. [DOI] [PubMed] [Google Scholar]
  • 89.Chandler KE, Kidd A, Al-Gazali L, Kolehmainen J, Lehesjoki A-E, Black GCM, Clayton-Smith J. Diagnostic criteria, clinical characteristics and natural history of Cohen syndrome. J Med Genet. 2003;40:233–241. doi: 10.1136/jmg.40.4.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Bodian M, Sheldon W, Lightwood R. Congenital hypoplasia of the exocrine pancreas. Acta Paediatr. 1964;53:282–293. doi: 10.1111/j.1651-2227.1964.tb07237.x. [DOI] [PubMed] [Google Scholar]
  • 91.Shwachman H, Diamond LK, Oski FA, Khaw K-T. The syndrome of pancreatic insufficiency and bone marrow dysfunction. J Pediatr. 1964;65:645–663. doi: 10.1016/s0022-3476(64)80150-5. [DOI] [PubMed] [Google Scholar]
  • 92.Ginzberg H, Shin J, Ellis L, Morrison J, Ip W, Dror Y, Freedman M, Heitlinger LA, Belt MA, Corey M, Rommens JM, Durie PR. Shwachman syndrome: phenotypic manifestations of sibling sets and isolated cases in a large patient cohort are similar. J Pediatr. 1999;135:81–88. doi: 10.1016/s0022-3476(99)70332-x. [DOI] [PubMed] [Google Scholar]
  • 93.Smith OP, Hann IM, Chessels JM, Reeves BR, Milla P. Haematological abnormalities in Shwachman-Diamond syndrome. Br J Haematol. 1996;94:279–284. doi: 10.1046/j.1365-2141.1996.d01-1788.x. [DOI] [PubMed] [Google Scholar]
  • 94.Woloszynek JR, Rothbaum RJ, Rawls AS, Minx PJ, Wilson RK, Mason PJ, Bessler M, Link DC. Mutations of the SBDS gene are present in most patients with Shwachman-Diamond syndrome. Blood. 2004;104:3588–3590. doi: 10.1182/blood-2004-04-1516. [DOI] [PubMed] [Google Scholar]
  • 95.Menne TF, Goyenchea B, Sánchex-Puig N, Wong CC, Tonkin LM, Ancliff PJ, Brost RL, Costanzo M, Boone C, Warren AJ. The Schwachman-Bodian-Siamond syndrome proteins mediates translational activation of ribosomes in yeast. Nat Genet. 2007;39:486–495. doi: 10.1038/ng1994. [Yeast has an ortholog of SBDS called SDO1 (SBDS ortholog) or YLR022C. SDO1 participates in a complex that facilitates release of Tif6 from pre-60S ribosomes in the process of their maturation. Another protein in the complex is EFL1 (elongation factor like 1). Therefore, the human ortholog of EFL1 becomes a candidate to be mutated in the Shwachman-Diamond syndrome patients that do not have SBDS mutations.] [DOI] [PubMed] [Google Scholar]
  • 96.Visser G, Rake J-P, Fernandes J, Labrune P, Leonard JV, Moses S, Ulrich K, Smit GPA. Neutropenia, neutrophil dysfunction, and inflammatory bowel disease in glycogen storage disease type Ib: results of the European Study on Glycogen Storage Disease type I. J Pediatr. 2000;137:187–191. doi: 10.1067/mpd.2000.105232. [DOI] [PubMed] [Google Scholar]
  • 97.Senior B, Loridan L. Studies of liver glycogenoses, with particular reference to the metabolism of intravenously administered glycerol. New Eng J Med. 1968;279:958–965. doi: 10.1056/NEJM196810312791802. [DOI] [PubMed] [Google Scholar]
  • 98.Beaudet AL, Anderson DC, Michels VV, Arion WJ, Lange AJ. Neutropenia and impaired neutrophil migration in type IB glycogen storage disease. J Pediatr. 1980;97:906–910. doi: 10.1016/s0022-3476(80)80418-5. [DOI] [PubMed] [Google Scholar]
  • 99.Barth PG, Scholte HR, Berden JA, Van der Klei-Van Moorsel JM, Luyt-Houwen IEM, Van't Veer-Korthof ET, Van der Harten JJ, Sobotka-Plojhar MA. An X-linked mitochondrial disease affecting cardiac muscle, skeletal muscle and neutrophil leucocytes. J Neurol Sci. 1983;62:327–355. doi: 10.1016/0022-510x(83)90209-5. [DOI] [PubMed] [Google Scholar]
  • 100.Kelley RI, Cheatham JP, Clark BJ, Nigro MA, Powell BR, Sherwood GW, Sladky JT, Swisher WP. X-linked dilated cardiomyopathy with neutropenia, growth retardation, and 3-methylglutaconic aciduria. J. Pediatr. 1991;119:738–747. doi: 10.1016/s0022-3476(05)80289-6. [DOI] [PubMed] [Google Scholar]
  • 101.Barth PG, Valianpour F, Bowen VM, Lam J, Duran M, Vaz FM, Wanders RJA. X-linked cardioskeletal myopathy and neutropenia. Am J Med Genet. 2004;126A:349–354. doi: 10.1002/ajmg.a.20660. [DOI] [PubMed] [Google Scholar]
  • 102.McMillin JB, Dowhan W. Cardiolipin and apoptosis. Biochim Biophys Acta. 2002;1585:97–107. doi: 10.1016/s1388-1981(02)00329-3. [DOI] [PubMed] [Google Scholar]
  • 103.Orstavik KH, Orstavik RE, Naumova AK, D'Adamo P, Gedeon A, Bolhuis PA, Barth PG, Toniolo D. X chromosome inactivation in carriers of Barth syndrome. Am J Hum Genet. 1998;63:1457–1463. doi: 10.1086/302095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.McKusick VA, Eldridge R, Hostetler JA, Egeland JA, Ruangwit U. Dwarfism in the Amish. II. cartilage-hair hypoplasia. Bull Johns Hopkins Hosp. 1965;116:285–326. [PubMed] [Google Scholar]
  • 105.Mäkitie O, Kaitila I. Cartilage-hair hypoplasia – clinical manifestations in 108 patients. Eur J Pediatr. 1993;152:211–217. doi: 10.1007/BF01956147. [DOI] [PubMed] [Google Scholar]
  • 106.Pearson HA, Lobel JS, Kocoshis SA, Naiman JL, Windmiller J, Lammi AT, Hoffman R, Marsh JC. A new syndrome of refractory sideroblastic anemia with vacuolization of marrow precursors and exocrine pancreatic dysfunction. J Pediatr. 1979;95:976–984. doi: 10.1016/s0022-3476(79)80286-3. [DOI] [PubMed] [Google Scholar]
  • 107.Styrt B. Species variation in neutrophil biochemistry and function. J Leuk Biol. 1989;46:63–74. doi: 10.1002/jlb.46.1.63. [DOI] [PubMed] [Google Scholar]
  • 108.Khuchua Z, Ye Z, Batts L, Strauss AW. A zebrafish mdel of human Barth syndrome reveals the essential role of tafazzin in cardiac development and function. Circulation Res. 2006;99:201–208. doi: 10.1161/01.RES.0000233378.95325.ce. [DOI] [PubMed] [Google Scholar]
  • 109.Xu Y, Condell M, Plesken H, Edelman-Novemsky I, Ma J, Ren M, Schlame M. A Drosophila model of Barth syndrome. Proc Natl Acad Sci USA. 2006;103:11584–11588. doi: 10.1073/pnas.0603242103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Lieschke GJ, Grail D, Hodgson G, Metcalf D, Stanley E, Cheers C, Fowler KJ, Basu S, Zhan YF, Dunn AR. Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization. Blood. 1994;84:1737–1746. [PubMed] [Google Scholar]
  • 111.Liu F, Wu HY, Wesselschmidt R, Tad Kornaga T, Link DC. Impaired production and increased apoptosis of neutrophils in granulocyte colony-stimulating factor receptor–deficient mice. Immunity. 1995;5:491–501. doi: 10.1016/s1074-7613(00)80504-x. [DOI] [PubMed] [Google Scholar]
  • 112.Belaaouaj A, McCarthy R, Baumann M, Gao Z, Ley TJ, Abraham SN, Shapiro SD. Mice lacking neutrophil elastase reveal impaired host defense against gram negative bacterial sepsis. Nat Med. 1998;4:615–618. doi: 10.1038/nm0598-615. [DOI] [PubMed] [Google Scholar]
  • 113.Grenda DS, Johnson SE, Mayer JR, McLemore ML, Benson KF, Horwitz M, Link DC. Mice expressing a neutrophil elastase mutation derived from patients with severe congenital neutropenia have normal granulopoiesis. Blood. 2002;100:3221–3228. doi: 10.1182/blood-2002-05-1372. [DOI] [PubMed] [Google Scholar]
  • 114.Karsunky H, Zen H, Schmidt T, Zevnik B, Kluge R, Schmid KW, Dührsen U, Möröy T. Inflammatory reactions and severe neutropenia in mice lacking the transcriptional repressor Gfi1. Nat Genet. 2002;30:295–300. doi: 10.1038/ng831. [DOI] [PubMed] [Google Scholar]
  • 115.Hock H, Hamblen MJ, Rooke HM, Traver D, Bronson RT, Cameron S, Orkin SH. Intrinsic requirement for zinc finger transcription factor Gfi-1 in neutrophil differentiation. Immunity. 2003;18:109–120. doi: 10.1016/s1074-7613(02)00501-0. [DOI] [PubMed] [Google Scholar]
  • 116.Cheung YY, Kim SY, Yiu WH, Pan C-J, Jun H-S, Ruef RA, Lee EJ, Heiner Westphal H, Brian C, Mansfield BC, Chou JY. Impaired neutrophil activity and increased susceptibility to bacterial infection in mice lacking glucose-6-phosphatase–β. J Clin Invest. 2007;117:784–793. doi: 10.1172/JCI30443. [Mice with G6PC3, which encodes glucose-6-phosphatase–β, knocked out develop neutropenia. In neutrophils, glucose-6-phosphatase–β interacts with the glucose-6-phosphate transporter (G6PT), which translocates glucose-6-phosphate (G6P) from the cytoplasm into the lumen of the ER. G6PT is the gene mutated in glycogen storage type 1b, and this finding helps explain why GSD1b patients have neutropenia, while GSD1a patients do not.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Hamasaki A, Sendo F, Nakayama K, Ishida N, Negishi I, Nakayama K, Hatakeyama S. Accelerated neutrophil apoptosis in mice lacking A1-a, a subtype of the bcl-2-related A1 gene. J Exp Med. 1998;188:1985–1992. doi: 10.1084/jem.188.11.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Dzhagalov I, St. John A, He Y-W. The antiapoptotic protein Mcl-1 is essential for the survival of neutrophils but not macrophages. Blood. 2007;109:1620–1626. doi: 10.1182/blood-2006-03-013771. [Mice with a knockout of Mcl1 conditional to neutrophils and macrophages have excess apoptosis in neutrophils, but not macrophages.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Chen L-Y, Shieh J-J, Baochuan Lin B, Pan C-J, Gao J-L, Murphy PM, Roe TF, Moses S, Ward JM, Lee EJ, Westphal H, Mansfield BC, Chou JY. Impaired glucose homeostasis, neutrophil trafficking and function in mice lacking the glucose-6-phosphate transporter. Hum Mol Genet. 2003;12:2547–2558. doi: 10.1093/hmg/ddg263. [DOI] [PubMed] [Google Scholar]
  • 120.Kim SY, Nguyen AD, Gao J-L, Murphy PM, Brian C, Mansfield BC, Chou JY. Bone marrow-derived cells require a functional glucose 6-phosphate transporter for normal myeloid functions. J Biol Chem. 2006b;39:28794–28801. doi: 10.1074/jbc.M604964200. [DOI] [PubMed] [Google Scholar]
  • 121.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 β3A subunit gene (Ap3b1) of the AP-3 adaptor complex is altered in the mouse hypopigmentation mutant pearl, a model for Hermansky-Pudlak syndrome and night blindness. Hum Molec Genet. 1999;8:323–330. doi: 10.1093/hmg/8.2.323. [DOI] [PubMed] [Google Scholar]
  • 122.Wilson SM, Yip R, Swing DA, O'Sullivan TN, Zhang Y, Novak EK, Swank RT, Russell LB, Copeland NG, Jenkins NA. A mutation in Rab27a causes the vesicle transport defects observed in ashen mice. Proc Natl Acad Sci U S A. 2000;97:7933–7938. doi: 10.1073/pnas.140212797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.van Raam BJ, Verhoeven AJ, Kuijpers TW. Mitochondria in neutrophil apoptosis. Int J Hematol. 2006;84:199–204. doi: 10.1532/IJH97.06131. [This is a review article focused on the role of mitochondria in cell death. Some key points in the review are as follows. Neutrophils are short-lived cells with a very high turnover, so committed neutrophil death is inevitable. Neutrophils can rely on glycolysis alone as a source of ATP, which allows them to function in hypoxic environments. There are both caspase-dependent and caspase-independent pathways of cell death in neutrophils, but both involve mitochondrial signaling. Bcl-2 family members play essential pro- and anti-apoptotic roles in neutrophil lifespan. Which pathway(s) is/are used to accelerate apoptosis after phagocytosis of bacteria is unknown. While the hypoxic state promotes apoptosis in many cell types, it seems to delay apoptosis in neutrophils.] [DOI] [PubMed] [Google Scholar]
  • 124.Maianski NA, Geissler J, Srinivasula SM, Alnemri ES, Roos D, Kuijpers TW. Functional characterization of mitochondria in neutrophils: a role restricted to apoptosis. Cell Death Differ. 2004a;11:143–153. doi: 10.1038/sj.cdd.4401320. [DOI] [PubMed] [Google Scholar]
  • 125.Weinmann P, Scharffetter-Kochanek K, Forlow SB, Peters T, Walzog B. A role for apoptosis in the control of neutrophil homeostasis in the circulation: insights from CD18-deficient mice. Blood. 2003;101:739–746. doi: 10.1182/blood-2002-01-0239. [DOI] [PubMed] [Google Scholar]
  • 126.Altznauer F, Conus S, Cavalli A, Folkers G, Simon H-U. Calpain-1 regulates Bax and subsequent Smac-dependent caspase-3 activation in neutrophil apoptosis. J Biol Chem. 2004;279:5947–5957. doi: 10.1074/jbc.M308576200. [DOI] [PubMed] [Google Scholar]
  • 127.Maianski NA, Roos D, Kuijpers TW. Bid truncation, Bid/Bax targeting to the mitochondria, and caspase activation associated with neutrophil apoptosis are inhibited by granulocyte colony-stimulating factor. J Immunol. 2004b;172:7031–7042. doi: 10.4049/jimmunol.172.11.7024. [DOI] [PubMed] [Google Scholar]
  • 128.Blomgran R, Zheng L, Stendahl O. Cathepsin-cleaved Bid promotes apoptosis in human neutrophils via oxidative stress-induced lysosomal membrane permeabilization. J Leukoc Biol. 2007;81:1213–1223. doi: 10.1189/jlb.0506359. [When a neutrophil is attached to a bacterium, the cascade of apoptosis has reactiveoxygen-species dependent lysosomal membrane permeabilization as an early event. This is followed by cleavage of Bid by cathepsins, then mitochondrial relase of cytochrome c, and then triggering caspases (presumably caspase 9). In contrast, non-phagosomal generation of reactive oxygen species does not lead to apoptosis.] [DOI] [PubMed] [Google Scholar]
  • 129.Bouillet P, Metcalf D, Huang DCS, Tarlinton DM, Kay TWH, Köntgen F, Adams JM, Andreas Strasser A. Proapoptotic Bcl-2 relative Bim required for certain apoptotic responses, leukocyte homeostasis, and to preclude autoimmunity. Science. 1999;286:1735–1738. doi: 10.1126/science.286.5445.1735. [DOI] [PubMed] [Google Scholar]
  • 130.Aprikyan AAG, Liles WC, Rodger E, Jonas M, Chi EY, Dale DC. Impaired survival of bone marrow hematopoietic progenitor cells in cyclic neutropenia. Blood. 2001;97:147–153. doi: 10.1182/blood.v97.1.147. [DOI] [PubMed] [Google Scholar]
  • 131.Yasui K, Sekiguchi Y, Ichikawa M, Nagumo H, Yamazaki T, Komiyama A, Suzuki H. Granulocyte colony macrophage stimulating factor delays neutrophil apoptosis and primes its function through Ia-type phosphoinositide 3-kinase. J Leukocyte Biol. 2002;72:1020–1026. [PubMed] [Google Scholar]
  • 132.Kuijpers TW, Maianski NA, Tool ATJ, G. Smit GPA, Rake JP, Roos D, Visser G. Apoptotic neutrophils in the circulation of patients with glycogen storage disease type 1b (GSD1b) Blood. 2003;101:5021–5024. doi: 10.1182/blood-2002-10-3128. [DOI] [PubMed] [Google Scholar]
  • 133.Ekert PG, Vaux Dl. The mitochondrial death squad: hardened killers or innocent bystanders? Curr Opin Cell Biol. 2005;17:626–630. doi: 10.1016/j.ceb.2005.09.001. [DOI] [PubMed] [Google Scholar]
  • 134.Dror Y, Freedman MH. Shwachman-Diamond syndrome marrow cells show abnormally increased apoptosis mediated through the Fas pathway. Blood. 2001;97:3011–3016. doi: 10.1182/blood.v97.10.3011. [DOI] [PubMed] [Google Scholar]
  • 135.Kuijpers TW, Alders M, Tool ATJ, Mellink C, Roos D, Hennekam RCM. Hematologic abnormalities in Shwachman Diamond syndrome: lack of genotypephenotype relationship. Blood. 2005;106:356–361. doi: 10.1182/blood-2004-11-4371. [DOI] [PubMed] [Google Scholar]
  • 136.Liu C-Y, Takemasa A, Liles WC, Goodman RB, Jonas M, Rosen H, Chi E, Winn RK, Harlan JM, Chuang PL. Broad-spectrum caspase inhibition paradoxically augments cell death in TNF-α-stimulated neutrophils. Blood. 2003;101:295–304. doi: 10.1182/blood-2001-12-0266. [DOI] [PubMed] [Google Scholar]
  • 137.Maianski NA, Roos D, Kuijpers TW. Tumor necrosis factor α induces a caspaseindependent death pathway in human neutrophils. Blood. 2003;101:1987–1995. doi: 10.1182/blood-2002-02-0522. [DOI] [PubMed] [Google Scholar]
  • 138.Allenbach C, Zufferey C, Perez C, Launois P, Mueller C, Tacchini-Cottier Macrophages induce neutrophil apoptosis through membrane TNF, a process amplified by Leishmania major. J Immunol. 2006;176:6656–6664. doi: 10.4049/jimmunol.176.11.6656. [DOI] [PubMed] [Google Scholar]
  • 139.Borregaard N, Herlin T. Energy metabolism of human neutrophils during phagocytosis. J Clin Invest. 1982;70:550–557. doi: 10.1172/JCI110647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Peachman KK, Lyles DS, Bass DA. Mitochondria in eosinophils: functional role in apoptosis but not respiration. Proc Natl Acad Sci USA. 2001;98:1717–1722. doi: 10.1073/pnas.98.4.1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Fossati G, Moulding DA, Spiller DG, Moots RJ, White MRH, Edwards SW. The mitochondrial network of human neutrophils: role in chemotaxis, phagocytosis, respiratory burst activation, and commitment to apoptosis. J Immunol. 2003;170:1964–1972. doi: 10.4049/jimmunol.170.4.1964. [DOI] [PubMed] [Google Scholar]
  • 142.Kuijpers TW, Maianski NA, Tool ATJ, Becker K, Plecko B, Valianpour F, Wanders RJA, Pereira R, Van Hove J, Verhoeven AJ, Roos D, Baas F, Barth PG. Neutrophils in Barth syndrome (BTHS) avidly bind annexin-V in the absence of apoptosis. Blood. 2004;103:3915–3923. doi: 10.1182/blood-2003-11-3940. [DOI] [PubMed] [Google Scholar]
  • 143.Nakazawa Y, Suzuki M, Manabe N, Yamada T, Kihara-Negishi F, Sakurai T, Tenen DG, Iwama A, Mochizuki M, Oikawa T. Cooperative interaction between ETS1 and GFI1 transcription factors in the repression of Bax gene expression. Oncogene. 2007;26:3541–3550. doi: 10.1038/sj.onc.1210140. [The zinc-finger domains of GFI1 are essential for it to bind with ETS1, and the GFI1-ETS1 complex regulates transcription of Bax. This opens the possibility that GFI1 dominant negative mutations lead to neutropenia via overexpression of Bax leading to increased apoptosis.] [DOI] [PubMed] [Google Scholar]
  • 144.Xu C, Bailly-Maitre B, Reed JC. Endoplasmic reticulum stress: cell life and cell decisions. J Clin Invest. 2005;115:2656–2664. doi: 10.1172/JCI26373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Carlsson G, Aprikyan AAG, Ericson KG, Stein S, Makaryan V, Dale DC, Nordenskjöld M, Fadeel B, Palmblad J, Henter JI. Neutrophil elastase and granulocyte colony-stimulating factor receptor mutation analyses and leukemia evolution in severe congenital neutropenia patients belonging to the original Kostmann family in northern Sweden. Haematologica. 2006;91:589–595. [PubMed] [Google Scholar]
  • 146.Skokowa J, Cario G, Uenalan M, Schambach A, Germeshausen M, Battmer K, Zeidler C, Lehmann U, Eder M, Baum C, Grosschedl R, Stanulla M, Scherr M, Welte K. LEF-1 is crucial for neutrophil granulocytopoiesis and its expression is severely reduced in congenital neutropenia. Nat Med. 2006;12:1191–1197. doi: 10.1038/nm1474. [The transcription factor LEF1 is deficient in cells of congenital neutropenia patients, but not cyclic neutropenia patients. LEF1 targets CCND1 (encodes cyclin D1), MYC (encodes c-Myc), BIRC5 (encodes survivin), and the transcription factor CEBPA (encodes CEBP/α) which plays a central role in granulopoiesis. Enforced expression of LEF1 upon retroviral gene transfer led to increase proliferation and differentiation in SCN patient cells. No mutations in LEF1 were found in patient cells, so it is unclear by what mechanism LEF1 expression is reduced. The binding site for LEF1 on the promoter of CEBPA was confirmed.] [DOI] [PubMed] [Google Scholar]
  • 147.Li F-Q, Person RE, Takemaru K-I, Williams K, Meade-White K, Ozsahin AH, Güngör T, Moon RT, Horwitz M. Lymphoid enhancer factor-1 links two hereditary leukemia syndromes through core-binding factor a regulation of ELA2. J Biol Chem. 2004;279:2873–2884. doi: 10.1074/jbc.M310759200. [DOI] [PubMed] [Google Scholar]
  • 148.Haurie C, Dale DC, Mackey MC. Cyclical neutropenia and other periodic hematological disorders: A review of mechanisms and mathematical models. Blood. 1998;92:2629–2640. [PubMed] [Google Scholar]
  • 149.Vainstein V, Ginosar Y, Shoham M, Ranmar DO, Ianovski A, Agur Z. The complex effect of granulocyte colonoy-stimulating factor on human granulopoiesis analyzed by a new physiologically-based mathematical model. J Theor Biol. 2005;234:311–327. doi: 10.1016/j.jtbi.2004.11.026. [DOI] [PubMed] [Google Scholar]

RESOURCES