Summary
Since their description by Metchnikoff in 1905, phagocytes have been increasingly recognized to be the entities that traffic to sites of infection and inflammation, engulf and kill infecting organisms, and clear out apoptotic debris all while making antigens available and accessible to the lymphoid organs for future use. Therefore, phagocytes provide the gateway and the first check on host protection and immune response. Disorders in killing and chemotaxis lead not only to infection susceptibility but also to autoimmunity. We aim to describe chronic granulomatous disease and the leukocyte adhesion deficiencies as well as myeloperoxidase deficiency and G6PD deficiency as paradigms of critical pathways.
Keywords: Neutrophil, Phagocytes, Inflammation, Colitis, Autoimmunity
Chronic Granulomatous Disease
Since it was first reported in 1954 in children with hypergammaglobulinemia and infections, Chronic Granulomatous Disease (CGD), has been characterized by both infection susceptibility and dysregulation of inflammation1–3 Genetically linked to 6 genotypes with similar a phenotype, the defect is in phagocyte production of reactive oxygen species (ROS) or superoxide, which is associated with ineffective microbial killing. This defect in microbial killing leads to specific bacterial and fungal infection susceptibilities. In the US, CGD occurs approximately in 1 in 200,000 live births4, but the numbers are likely higher. The inheritance can be either X-linked or autosomal recessive (AR) with increased rates of AR inheritance in areas of the world in which there are higher rates of consanguinity. In the US, the X-linked form accounts for approximately 65% of all cases.
ROS and CGD
CGD is due to mutations in any of the proteins that make up the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex itself or lead to its assembly. Impairment of the NADPH complex leads to an inability to transfer an electron from NADPH to molecular oxygen to form superoxide. This results in impaired killing in neutrophils, macrophages and monocytes. The NADPH complex per se is made up of six proteins. gp91phox and p22phox are lodged in the membrane of the secondary granules and together form the heterodimer cytochrome b558; the cytoplasmic tail of gp91phox (encoded by CYBB) binds flavin adenine dinucleotide (FAD), NADPH (on the C-terminus end of gp91phox) and heme (on the N-terminus) permitting the transfer of an electron to molecular oxygen; p22phox (encoded by CYBA) is bound to gp91phox and is required for the expression of the heterodimer. Upon phagocyte activation, three cytosolic proteins p47phox (encoded by NCF1), p67phox (encoded by NCF2), and p40phox (encoded by NCF4) are phosphorylated, aggregate and translocate to cytochrome b558 along with RAC1/RAC2 at the membrane surface. The critical interaction is the binding of the p47phox/p67phox complex to p22phox5. RAC1/RAC2 are GTPases which are bound to the complex, with RAC2 being the predominant phagocyte form. Electron transfer from NADPH to molecular oxygen results in a superoxide anion (O2−), which is converted to hydrogen peroxide by superoxide dismutase (SOD). Hydrogen peroxide is then converted to HOCl (hypohalous acid; bleach in neutrophils because the halide is chlorine) by myeloperoxidase (MPO). The intravacuolar generation of superoxide changes the intravacuolar electrical charge, eliciting a K+ flux that enhances intracellular killing through proteases which had been delivered by the primary granule6 (Figure 1.). Another genetic cause of CGD is due to a deficiency of the chaperone protein required for the transport of the gp91phox/p22phox complex from the endoplasmic reticulum into a plasma membrane, named essential for reactive oxidant signaling (EROS, encoded by CYBC1)7. Neutrophil extracellular traps (NETs) appear to be dependent on ROS, therefore are also defective in CGD, which may also affect microbial killing8,9. NETs are released decondensed nuclear chromatin coated with histones and proteases that require MPO for protease activation after nuclear elastases promote decondensation of chromatin. Defects of microbial killing via NETosis in CGD cells have been reversed with the addition of tamoxifen, which induced NET formation in a ROS independent manner and neutrophils treated with tamoxifen showed significant improvement in microbial killing9. NET formation was restored in a patient who underwent gene therapy for CGD10.
Figure 1.

Assembly of the NADPH complex leads to both the production of HOCl and the influx of potassium as a result of the net negative intraphagosomic charge resulting in activation of proteases. This leads to killing in intracellular organisms to which the patients with CGD are susceptible.
Genetics of CGD
The most common affected gene causing CGD in the United States is CYBB, located at Xp21.1, encoding gp91phox. There are 681 mutations identified in CYBB so far11. Overall, mortality is inversely linked to ROS production12, with survival increasing in patients with mutations that allow more residual ROS production. Early missense mutations before amino acid 310 (except for His 222) tend to result in an intact protein with some residual ROS production and higher rates of overall survival, while those mutations after amino acid 310, which are in the NADPH, FAD, and heme binding domain, tend to be unable to support ROS production. In contrast, nonsense mutations, deletions and some splice mutations result in no protein production and have the lowest residual ROS production and lower overall survival12. Additionally, liver abscess and liver disease as evidenced by platelet decline of >9000/mm3/y and elevation in alkaline phosphatase by >0.25U/L/y have been independently linked to mortality13. Given its location on Xp21, large deletions involving CYBB may also encompass other genes: telomeric deletions involving KX lead to McLeod syndrome (Kell antigen deletion), as CYBB and KX are < 50 kbp apart. In this deletion, initial red blood cell transfusion can result in formation of anti-Kx and anti-Km antibodies making future transfusions extremely dangerous14. Duchenne muscular dystrophy (DMD) and X-linked retinitis pigmentosa (RPGR) can also be impacted by large deletions in Xp21.1 as part of a contiguous deletion. Centromeric deletions can cause ornithine transcarbamylase deficiency, which has also been described in X-linked carriers15–17. This is why chromosomal microarrays to detect the boundaries of large deletions are important in X-linked CGD.
The second most common CGD mutation in the US is in NCF1, the recessive gene encoding p47phox and accounting for ~25% of cases. The vast majority of mutations are homozygous for a GT deletion at the start of exon 218. Of ~900 mutations in NCF1 worldwide, about 80% were homozygous for the GT deletion. This common deletion occurs because most individuals carry two pseudogenes, each of which carries the deletion and flanks the functional gene. Recombination events in which a GT-deleted pseudogene replaces the functional NCF1 result in early truncation and no protein production18,19. Despite this lack of protein, p47phox deficient CGD typically has residual ROS, albeit not normal amounts, and is a milder infection susceptibility phenotype.
The second subunit of the cytochrome b558 is autosomal recessive p22phox (encoded by CYBA at 16q24) accounts for 7% of the mutations leading to US CGD18. Because it is required for gp91phox expression and stability, mutations in CYBA are clinically almost indistinguishable from mutations in CYBB, except that CYBA is autosomal recessive, leading to females with the X-linked phenotype. p67phox is another autosomal recessive cytosolic component (encoded by NCF2, at 1q25.3), and the critical cytosolic factor for NADPH activation. P47phox is the partner protein crucial for getting p67phox into the NADPH complex, partly explaining why p67phox deficiency is usually more severe than p47phox deficiency. p40phox (encoded by NCF4 at 22q12.3) is another autosomal recessive member of the NADPH oxidase that serves a more regulatory role. The standard functional test, Dihydrorhodamine- 1,2,3 (DHR) oxidation, may be normal to PMA and ionomycin but abnormal to testing with fMLF or LPS. This form of CGD should be considered in patients with significant inflammatory bowel disease, even without infections20,21. This diagnosis should be considered in patients severe inflammatory bowel disease, and in particular with very early onset inflammatory bowel disease (VEO-IBD). Essential for reactive oxidant signaling (EROS) is encoded by CYBC1 at 17q25.3. EROS is the critical chaperone for the gp91phox/p22phox complex to get out of the ER and into the secondary granule membrane. It is autosomal recessive and tends to have high levels of inflammatory bowel disease7,22,23.
Clinical Manifestations
Infection
CGD infection susceptibility has been well described in numerous reviews24–27. Infections typically occur in infancy or early childhood. However, later diagnoses occur in patients with significant residual superoxide production, which can be missed when the phenotype is less severe. Characteristic sites of infection include lungs, liver, skin and bone24,25. The most common pathogens in the US are Staphylococcus aureus, Burkholderia cepacia complex, Serratia marcescens, Nocardia spp., and Aspergillus spp. In other parts of the world Salmonella spp, Mycobacterium tuberculosis and Bacille Calmette-Guerin (BCG) are also important. There are a few bacteria that are almost pathognomonic for CGD and should prompt diagnostic testing: Chromobacterium violaceum, Francisella philomiragia and Granulibacter bethesdensis28–32.Both Chromobacterium and Francisella are gram negative organisms that are found in warm brackish water and can cause sepsis in CGD. Chromobacterium and Francisella are both well treated with carbapenems. Granulibacter, a fastidious gram-negative rod, can present as chronic or recurrent necrotizing lymphadenitis and can be particularly difficult to eradicate, and often responds to ceftriaxone30,31. In the US there has been one case of fatal sepsis secondary to G. bethesdensis33. European strains of Granulibacter have been more severe in presentation34. Granulibacter requires special culture media, such as Charcoal yeast extract (CYE) and can be identified by 16S sequencing30,35. In some instances, after parenteral therapy with ceftriaxone, longer term cefdinir has been helpful.
Staphylococcus aureus is a gram-positive bacterium that tends to cause deep seated infections in CGD including lymph nodes, skin, liver and bone. Liver abscess was previously treated with surgery and antibiotics. This approach was complicated by high rates of relapse and recurrence. The liver abscesses of CGD are not very amenable to percutaneous drainage as the abscess is often fibrocaseous and septated36,37. The seemingly counterintuitive approach of simultaneous therapy with steroids and antibiotics for staphylococcal liver abscess has resulted in decreased need for surgery lower morbidity and mortality36. However, diagnosis of the microbiological cause of liver abscess still requires definitive determination, such as percutaneous aspiration36.
Infections caused by Burkholderia spp often cause pneumonia, but bacteremia can result in both sepsis and hemophagocytic lymphohistiocytosis (HLH)38. B. gladioli and B. pseudomallei have also been reported in CGD39,40
Aspergillus and other mold infections can be fatal in CGD. With the advent of highly effective antifungal prophylaxis and newer azoles, A. fumigatus is largely curable. However, the non-fumigatus Aspergillus species can be intrinsically resistant to amphotericin and many azole antifungal therapies and can be invasive, eroding through tissue planes41. In a review of non-Aspergillus infections in CGD the most frequent offenders were Rhizopus spp. and Trichosporon spp. and overall mortality was 26%42. Other molds that have been described to cause invasive and often fatal disease in CGD include A. viridinutans, A. nidulans, A. tanneri, Paecilomyces variotii, P. lilacinus, Neosartorya udagawae, and Phellinus spp.41,43–46. In these cases, if there is isolated disease then surgical resection may be advisable. Because of the stark therapeutic divide, it is critical to definitively diagnose invasive fungal disease to the species level as medical therapies are not always sufficient and adjunctive treatments take time to arrange (hematopoietic stem cell transplant, allogeneic granulocyte infusions).
A variation on fungal pneumonia is “mulch pneumonitis” described in CGD presenting acutely, with fever and dyspnea, typically within 24 hours of exposure and with potentially minimal changes on conventional X-ray47–51. The history often includes exposure to decaying matter such as mulching, landscaping, demolition, lawn debris or animal feed. Patients may present for dyspnea with what is called community acquired pneumonia. Left untreated, patients invariably return in respiratory distress with abnormal X-ray and widespread diffuse nodules on CT. This condition is life threatening. Treatment typically requires both antifungal (mold-active azole) and antibacterial (carbapenem) and corticosteroids50.
Inflammatory complications
Gastrointestinal complications occur in about 50% of patients with X-linked CGD, appearing usually by 7 years, whereas in p47phox CGD the rate is about 40% but the age of onset is 22 years52. Signs and symptoms can range from failure to thrive, irritability and diarrhea in the very young to urgency, frequency, tenesmus, iron deficiency anemia, diarrhea, and hematochezia in adults. Functional gastric, esophageal, and duodenal obstruction have also been described53,54. Common radiographic findings can include peri-colonic fat stranding, mesenteric adenopathy and bowel wall thickening55. CGD-related IBD does not affect overall mortality, but it does cause significant morbidity12,56. IBD may be the initial presentation of any of the genotypes of CGD. In particular NCF4 deficiency often presents as IBD only, without infections18,20,21. Typically, the colon is most affected, with the perirectal region most involved. IBD can progress from distal to proximal affecting the entirety of the colon53,57. Disease is morphologically similar to Crohn disease (CD) with ulcerations, friability and erythema of the mucosa with acute colitis, and crypt abscesses58,59. There is a high rate of perianal disease, with fistula formation and fissures59. In addition to the colonic disease, video capsule endoscopy (VCE) in CGD found that 85% had some sort of small bowel involvement, not predicted by radiographic appearance60. Therefore, comprehensive staging of CGD patients with IBD is indicated.
CGD IBD does not correlate with residual superoxide production12,53. In addition, within families who all share the same mutation rates of IBD are discordant. A comparison of patients with CGD IBD against those with IBD without CGD found that patients with CGD IBD had many of the same genetic risks as in the general IBD population but their burden was lower overall, consistent with the idea that ROS deficiency is per se an important contributor to the development of CGD IBD61. Inflammasome activation and defective autophagy in the setting of elevated IL-1beta has also been implicated in a mouse model of CGD as a cause of CGD IBD62. Treatment of CGD IBD with IL-1 blockade has shown some small improvements in disease, but has not been sustained in small reports62,63. The intestinal microbiome regulates immunity and inflammasome activation and may be important in the development of CGD IBD64. The microbiota of CGD patients with and without IBD show different levels of alpha diversity in CGD patients compared with healthy controls64,65. Moreover, the beta diversity of the fecal microbiome can distinguish CGD patients from healthy controls65. The microbiome in symptomatic CGD IBD is distinct from that in CGD without IBD and can shift from one to the other upon institution of an elemental diet66,67. There are ongoing trials targeting the microbiome in CGD, both through the use of fecal transplant and also using elemental diets.
Medical therapy for CGD IBD is directed at the dysregulated inflammation, ideally without increasing the risk of infection. Traditionally, the mainstay of therapy has been corticosteroids and lumenal non-steroidal gastrointestinal medications such as mesalamine, or sulfasalazine; for more refractory cases azathioprine is usually added53,68. Unfortunately, long term steroids can lead to aseptic necrosis and osteopenia53. Therapies aimed at Crohn IBD have been utilized with varying degrees of success. Despite improvement in fistulizing disease with TNF-α blockade in CGD IBD, fatal infections and increased infection susceptibility have been associated with their use69,70. The humanized monoclonal antibody that targets the integrin α4β7, vedolizumab, has shown modest improvement in CGD IBD. Although it did not show significant sustained improvement it may have a role as a bridge to more definitive therapy71,72. Ustekinumab, a monoclonal antibody targeting IL-12/IL-23 has been reported to improve CGD IBD but has been associated with some increase in infection susceptibility. However, it can take several months to see results and disease may recur if therapy is stopped and73,74. We use a long steroid taper until the effects of ustekinumab can be established. There is still not an immunomodulatory therapy that is both safe and effective for severe CGD IBD. The only definitive therapy for CGD IBD is hematopoietic stem cell transplantation (HSCT)75,76. Encouragingly, CGD IBD largely resolves with transplant and its presence does not impact transplant outcomes. Therefore, HSCT is an attractive option even for patients with inflammatory bowel disease as their sole CGD morbidity75–78.
The liver is another part of the GI tract that can be affected by dysregulated inflammation, with complications including non-cirrhotic portal hypertension, nodular regenerative hyperplasia, and hepato-splenomegaly resulting in sequestration and thrombocytopenia13,79.
Approximately one quarter of patients with CGD exhibit signs of pulmonary inflammatory complications80,81. These can be severe enough to result in hypoxemia and pulmonary hypertension. In one European cohort, upwards of 77% of their patients had pulmonary abnormalities on computed tomography scanning, including 43% with bronchiectasis or fibrosis82. Unpublished data suggest that some of this can resolve with transplantation.
Urological manifestations of CGD can include eosinophilic cystitis as well as obstructive uropathy. Both are effectively treated with corticosteroids; however, the episodes can recur and may require low dose steroids over a prolonged period54,83.
Chorioretinitis, uveitis, and ocular granulomata have also been described in CGD, most of which do not progress. However, there are reports of loss of vision sometimes resulting in enucleation. The etiology of this complication is poorly understood and no clear effective treatments have been described80,84,85. Patients with CGD should be screened and followed routinely for this inflammatory complication.
Curative therapy
Much of the therapy for CGD is aimed at prevention and treatment of both infections and inflammatory complications. Currently the only available cure for CGD is HSCT, with overall survival >90% for children transplanted < 14 years of age76,77,86–88. Resolution of inflammatory bowel disease and full engraftment with both matched related and unrelated donors have been reported, making transplant a viable option even in the setting of ongoing infection or inflammatory complications. Transplant has also been reported in highly Lyonized carriers, either in the setting of incurable infection or for the same inflammatory complications of the GI tract as in X-linked boys. Gene therapy for X-linked CGD has been performed with success, but has been complicated in some cases by loss of gene expression and in rare cases by development of myelodysplasia or leukemia, as has been the case with some other early efforts at gene therapy89. However, some of the newer vectors and approaches are promising. Whether gene therapy can fully control the inflammatory complications of CGD remains to be demonstrated. Newer ex-vivo using technologies like CRISPR/Cas9 have shown promise and are on the cusp of trials in humans90,91.
Myeloperoxidase deficiency
After the superoxide free radical is converted by superoxide dismutase to hydrogen peroxide (H2O2), MPO is the heme-containing enzyme necessary for its conversion to hypochlorous acid (HOCl). MPO is expressed early in myeloid differentiation and resides in the azurophilic (primary) granules of neutrophils and the lysosomes of monocytes. Located at 17q23, this is the most common inherited neutrophil disorder, occurring in approximately 1/2000–4000. It is rarely associated with infection despite its demonstrated role in oxidation of phagocytosed organisms92. There are case reports of invasive Candida infections and paracoccidioidomycosis infection, but these infections are typically associated with other co-morbidities such as diabetes mellitus93–95. It is important to keep in mind that MPO deficiency is another of the causes of an abnormal DHR assay, which can on occasion be quite misleading. More commonly, MPO has been associated with inflammatory disorders given its role as a mediator of inflammation96. Recent work in mouse models suggests that MPO is involved in mediating apoptosis and that in its absence inflammation is enhanced, offering a possible overlap between MPO deficiency, in which HOCl is not generated because of an enzyme defect and CGD, in which HOCl is not generated because of a substrate defect97
Glucose-6-Phosphate Dehydrogenase Deficiency (G6PD)
Glucose-5-phosphate dehydrogenase deficiency (G6PD) belongs in the category of neutrophil disorders, as it is one of the first two reactions of the hexose monophosphate shunt pathway that generates NADPH. G6PD deficiency affects about 400 million persons worldwide with an increased prevalence in Africa, Asia and the Middle East, making it the most common X-linked disorder98. The amount of NADPH available for use in a number of reactions, including for the NADPH oxidase, is dependent on G6PD, and therefore less or less effective enzyme results in insufficient NADPH and reviewed recently11,99. G6PD deficiency is most commonly associated with hemolysis of red blood cells. Located at Xq23, G6PD deficiency can range from mild to severe, with 5 different classes of variants. Infections occur in the most severe form of G6PD deficiency and generally phenocopy CGD susceptibility100. G6PD deficiency should be considered in a patient with severe hemolysis and CGD-associated infections.
Leukocyte Adhesion Deficiency
Defects in migration resulting in infection susceptibility and inflammatory complications can be due to one of the three types of Leukocyte Adhesion Deficiency (LAD). These defects can result in variable phenotypes as a result of impaired adhesion to the endothelium or transendothelial migration out of the bloodstream101. Neutrophil and monocyte adhesion to the endothelium and to other cells and to some pathogens is mediated in part by molecules collectively known as integrins [CD11a/CD18 (LFA-1), CD18/CD11b (CR3 or Mac-1), CD18/CD11c (CR4), CD18/CD11d] which are stored in intracellular vesicles and translocate to the leukocyte cell surface where they can interact with intercellular adhesion molecules (ICAMs) on the endothelial surface. Lymphocyte function-associated antigen (LFA-1) is a heterodimeric integrin present on the surface of all leukocytes that is essential for neutrophil recruitment to sites of tissue inflammation, but also for processes such as homotypic adhesion leading to T cell activation102. Mac-1 or Complement receptor (CR) 3 is essential in responding to chemotactic factors, such as the complement fragment C5a, IL-8, leukotriene B4 (LTB4), or the bacterial product formyl-methionyl-leucyl-phenylalanine (fMLF) and leukocyte adhesion to the endothelium101,102. CR4 (also known as p150,95) mediates binding to lipopolysaccharide (LPS) directly as well as fibrinogen. It is also highly expressed on monocyte-derived dendritic cells103. CD18/CD11d is an integrin whose expression is more complexly regulated and whose role in human health and disease is less well understood103. There is a bidirectional interplay between the neutrophil (integrin expression) and the endothelium (ICAM expression) required for appropriate adhesion and extravasation. Neutrophil integrin expression is entirely dependent on the common subunit of all 4 integrins, CD18 encoded by ITGB2 at 21q22104. Therefore, defects in CD18 expression or function lead to defects in any or all of the CD11 family molecules and therefore defects in adhesion, trafficking and killing. Disease severity in LAD-1 is determined by the amount of CD18 expression and categorized as severe (<2%), moderate (2%−30%), or mild (>30%). Inherited in a autosomal recessive manner, patients with LFA-1 severe mutations frequently present with delayed umbilical cord separation, poor wound healing, neutrophilia, oral ulcers, and aggressive gingivitis with accelerated apical bone loss105,106. The severe periodontitis typically results in loss of all adult teeth by early adulthood. The mechanism of the inflammation associated with LAD1 is somewhat counterintuitive, as the inability of neutrophils to exit the vascular space leads to tissue neutropenia, which is associated with hypoinflammatory lesions, such as in neutropenia. However, tissue resident macrophages are constantly monitoring for adequate neutrophil levels in the tissue, and when they decline macrophages produce IL-23 which eventually leads to IL-17 and G-CSF production. Therefore, it is the local production of high levels of IL-17 as a result of macrophage IL-23 that leads to the severe inflammatory complications of LAD1, including severe gingivitis and inflammatory skin lesions107. This latter observation led to the use of ustekinumab in the treatment of gingivitis and inflammation in LAD1108. HSCT is curative for LAD1 but results are complicated by high rates of graft failure and graft versus host disease109. In view of the persisting difficulties of HSCT there has been significant effort for gene therapy for LAD1, which has so far been successful with survival of all recipients past one year110.
LAD-2 is due to defect in the fucosylation of the protein CD15s (sialyl LewisX, SLeX) on neutrophils, thus impairing the early low-avidity rolling step of neutrophil adhesion which is mediated by selectins on the endothelium. The molecular defect is in the guanine diphosphate (GDP)–fucose transporter-1 (FUCT1 or SLC35C1). This defect in glycosylation places LAD2 within the congenital disorders of glycosylation (CDG), making this CDGIIc. Inherited in an autosomal recessive manner, patients with LAD2 have the Bombay blood group phenotype, which is due to the absence of the H antigen, which is also dependent on fucosylation105. There are many fucoslylated proteins, all of which are affected, so patients often present with short stature and cognitive deficits. The infection phenotype in LAD2 appears to improve with age and is usually less prominent in adulthood, but the number of cases reports is small. There may be a subset of cases who respond to fucose supplementation, as well111. Because the integrin, phagocyte killing and T-cell receptor are still functional in this LAD2, the infectious complications tend to be less severe106.
Initially thought to be a variant of LAD-1, LAD-III was first described in a Turkish patient with mutations in FERMT3, which encodes KINDLIN3, an adaptor protein expressed in hematopoietic cells that regulates integrin activation through binding to the short intracellular tails of β2 integrins112. The process of KINDLIN3 binding to the intracellular portion of the integrins is called inside-out signaling, since the KINDLIN3 interaction strengthens integrin binding. Described in only a handful of patients and families, LAD III is autosomal recessive and presents with infections and a Glanzmann thrombasthenia phenotype bleeding tendency113–115. Patients typically have a purpuric (“mulberry”) rash at birth and may have early fatal infections. LAD-III platelets have decreased binding to soluble fibrinogen, and do not respond properly to thrombin via thrombin receptors, resulting in improper platelet granule secretion via integrin activation and hence the purpuric rash. HSCT is curative for LAD3 and should be considered early in the course109,116–118.
Table 1.
Genetic testing for clinicians who suspect a neutrophil disorder and the genes affected.
| Disease and Protein | Gene | Diagnostic Test | Chromosome | Inheritance | Sequencing |
|---|---|---|---|---|---|
| CGD | |||||
| - gp91 | CYBB | DHR | Xp21.1 | X-linked | Yes |
| - p22phox | CYBA | DHR | 16q24.2 | AR | Yes |
| - p47phox | NCF1 | DHR | 7q11.23 | AR | Possible1 |
| - p67phox | NCF2 | DHR | 1q25 | AR | Yes |
| - p40phox | NCF4 | May look normal2 | 22q12.3 | AR | Yes |
| - EROS | CYBC1 | DHR may be low | 17q25.3 | AR | Yes |
| - RAC2 | Rac2 | 22q13 | AD | Yes | |
| G6PD | G6PD | Enzymatic activity | Xq28 | X-linked | Yes |
| Myeloperoxidase deficiency | 3DHR may appear abnormal | 17q23 | AR | Yes | |
| LAD | |||||
| - LAD1 | ITGB2 | Flow Cytometry4 | 21q22.3 | AR | Yes |
| - LAD2 | SLC35C1 | Flow Cytometry4 | 11p11.2 | AR | Yes |
| - LAD3 | FERMT3 | Flow Cytometry4 | 11q13.1 | AR | Yes |
May be difficult given that there are two pseudo genes which may miss the typical GT deletion
If clinical suspicion is high – request DHR with serum-opsonized zymosan or E.coli rather than PMA. However direct sequencing is indicated for high clinical suspicion.
Patient may erroneously be diagnosed with CGD, however direct visualization of neutrophils when stained for peroxidase is more specific.
Abnormal flow cytometry for CD11a, CD11b, CD11c and CD18 in combination with clinical suspicion fitting the phenotype
Acknowledgement:
Funding for this review was provided in part by the Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does the mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government.
Contributor Information
Christa S. Zerbe, Laboratory of Clinical Immunology, National Institutes of Allergy and Infectious Disease, The National Institutes of Health, 10 Center Drive, MSC 1899, Rm 12C103, Bethesda, MD 20817.
Steven M. Holland, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bldg. 10/11N248 MSC 1960, Bethesda, MD 20892-1960.
References
- 1.Bridges RA, Berendes H, Good RA. A fatal granulomatous disease of childhood; the clinical, pathological, and laboratory features of a new syndrome. AMA J Dis Child. Apr 1959;97(4):387–408. [PubMed] [Google Scholar]
- 2.Janeway CA, Craig J, Davidson M, Downey W, Gitlin D. Hypergammaglobulinemia Associated with Severe Recurrent and Chronic Nonspecific Infection. Ama Am J Dis Child. 1954;88(3):388–392. [Google Scholar]
- 3.Berendes H, Bridges RA, Good RA. A fatal granulomatosus of childhood: the clinical study of a new syndrome. Minn Med. May 1957;40(5):309–12. [PubMed] [Google Scholar]
- 4.Winkelstein JA, Marino MC, Johnston RB Jr., et al. Chronic granulomatous disease. Report on a national registry of 368 patients. Medicine (Baltimore). May 2000;79(3):155–69. doi: 10.1097/00005792-200005000-00003 [DOI] [PubMed] [Google Scholar]
- 5.Segal BH, Leto TL, Gallin JI, Malech HL, Holland SM. Genetic, biochemical, and clinical features of chronic granulomatous disease. Medicine (Baltimore). May 2000;79(3):170–200. doi: 10.1097/00005792-200005000-00004 [DOI] [PubMed] [Google Scholar]
- 6.Segal AW. How neutrophils kill microbes. Annu Rev Immunol. 2005;23:197–223. doi: 10.1146/annurev.immunol.23.021704.115653 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Thomas DC, Charbonnier LM, Schejtman A, et al. EROS/CYBC1 mutations: Decreased NADPH oxidase function and chronic granulomatous disease. J Allergy Clin Immunol. Feb 2019;143(2):782–785 e1. doi: 10.1016/j.jaci.2018.09.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Papayannopoulos V Neutrophil extracellular traps in immunity and disease. Nat Rev Immunol. Feb 2018;18(2):134–147. doi: 10.1038/nri.2017.105 [DOI] [PubMed] [Google Scholar]
- 9.Sharma A, McCann K, Tripathi JK, et al. Tamoxifen restores extracellular trap formation in neutrophils from patients with chronic granulomatous disease in a reactive oxygen species-independent manner. J Allergy Clin Immunol. Aug 2019;144(2):597–600 e3. doi: 10.1016/j.jaci.2019.04.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bianchi M, Hakkim A, Brinkmann V, et al. Restoration of NET formation by gene therapy in CGD controls aspergillosis. Blood. Sep 24 2009;114(13):2619–22. doi: 10.1182/blood-2009-05-221606 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Roos D, van Leeuwen K, Hsu AP, et al. Hematologically important mutations: X-linked chronic granulomatous disease (fourth update). Blood Cells Mol Dis. Sep 2021;90:102587. doi: 10.1016/j.bcmd.2021.102587 [DOI] [PubMed] [Google Scholar]
- 12.Kuhns DB, Alvord WG, Heller T, et al. Residual NADPH oxidase and survival in chronic granulomatous disease. N Engl J Med. Dec 30 2010;363(27):2600–10. doi: 10.1056/NEJMoa1007097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Feld JJ, Hussain N, Wright EC, et al. Hepatic involvement and portal hypertension predict mortality in chronic granulomatous disease. Gastroenterology. Jun 2008;134(7):1917–26. doi: 10.1053/j.gastro.2008.02.081 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Giblett ER, Klebanoff SJ, Pincus SH. Kell phenotypes in chronic granulomatous disease: a potential trnsfusion hazard. Lancet. Jun 12 1971;1(7711):1235–6. doi: 10.1016/s0140-6736(71)91738-7 [DOI] [PubMed] [Google Scholar]
- 15.Deardorff MA, Gaddipati H, Kaplan P, et al. Complex management of a patient with a contiguous Xp11.4 gene deletion involving ornithine transcarbamylase: a role for detailed molecular analysis in complex presentations of classical diseases. Mol Genet Metab. Aug 2008;94(4):498–502. doi: 10.1016/j.ymgme.2008.04.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Coman D, Yaplito-Lee J, La P, et al. Three Mendelian disorders (chronic granulomatous disease, retinitis pigmentosa, ornithine transcarbamylase deficiency) in a young woman with an X chromosome deletion, del(X)(p11.4p21.1). Mol Genet Metab. Mar 2010;99(3):329. doi: 10.1016/j.ymgme.2009.11.006 [DOI] [PubMed] [Google Scholar]
- 17.Balasubramaniam S, Rudduck C, Bennetts B, Peters G, Wilcken B, Ellaway C. Contiguous gene deletion syndrome in a female with ornithine transcarbamylase deficiency. Mol Genet Metab. Jan 2010;99(1):34–41. doi: 10.1016/j.ymgme.2009.08.007 [DOI] [PubMed] [Google Scholar]
- 18.Roos D, van Leeuwen K, Hsu AP, et al. Hematologically important mutations: The autosomal forms of chronic granulomatous disease (third update). Blood Cells Mol Dis. Dec 2021;92:102596. doi: 10.1016/j.bcmd.2021.102596 [DOI] [PubMed] [Google Scholar]
- 19.Hayrapetyan A, Dencher PC, van Leeuwen K, de Boer M, Roos D. Different unequal cross-over events between NCF1 and its pseudogenes in autosomal p47(phox)-deficient chronic granulomatous disease. Biochim Biophys Acta. Oct 2013;1832(10):1662–72. doi: 10.1016/j.bbadis.2013.05.001 [DOI] [PubMed] [Google Scholar]
- 20.Matute JD, Arias AA, Wright NA, et al. A new genetic subgroup of chronic granulomatous disease with autosomal recessive mutations in p40 phox and selective defects in neutrophil NADPH oxidase activity. Blood. Oct 8 2009;114(15):3309–15. doi: 10.1182/blood-2009-07-231498 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.van de Geer A, Nieto-Patlan A, Kuhns DB, et al. Inherited p40phox deficiency differs from classic chronic granulomatous disease. J Clin Invest. Aug 31 2018;128(9):3957–3975. doi: 10.1172/JCI97116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Mortimer PM, Nichols E, Thomas J, et al. A novel mutation in EROS (CYBC1) causes chronic granulomatous disease. Clin Immunol. Oct 2023;255:109761. doi: 10.1016/j.clim.2023.109761 [DOI] [PubMed] [Google Scholar]
- 23.Chiriaco M, De Matteis A, Cifaldi C, et al. Characterization of AR-CGD female patient with a novel homozygous deletion in CYBC1 gene presenting with unusual clinical phenotype. Clin Immunol. Jun 2023;251:109316. doi: 10.1016/j.clim.2023.109316 [DOI] [PubMed] [Google Scholar]
- 24.Marciano BE, Spalding C, Fitzgerald A, et al. Common severe infections in chronic granulomatous disease. Clin Infect Dis. Apr 15 2015;60(8):1176–83. doi: 10.1093/cid/ciu1154 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.van den Berg JM, van Koppen E, Ahlin A, et al. Chronic granulomatous disease: the European experience. PLoS One. 2009;4(4):e5234. doi: 10.1371/journal.pone.0005234 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Leiding JW, Holland SM. Chronic Granulomatous Disease. In: Adam MP, Mirzaa GM, Pagon RA, et al. , eds. GeneReviews((R)). 1993. [PubMed] [Google Scholar]
- 27.Arnold DE, Heimall JR. A Review of Chronic Granulomatous Disease. Adv Ther. Dec 2017;34(12):2543–2557. doi: 10.1007/s12325-017-0636-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Meher-Homji Z, Mangalore RP, P DRJ, K YLC. Chromobacterium violaceum infection in chronic granulomatous disease: a case report and review of the literature. JMM Case Rep. Jan 2017;4(1):e005084. doi: 10.1099/jmmcr.0.005084 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mailman TL, Schmidt MH. Francisella philomiragia adenitis and pulmonary nodules in a child with chronic granulomatous disease. Can J Infect Dis Med Microbiol. Jul 2005;16(4):245–8. doi: 10.1155/2005/486417 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Greenberg DE, Shoffner AR, Zelazny AM, et al. Recurrent Granulibacter bethesdensis infections and chronic granulomatous disease. Emerg Infect Dis. Sep 2010;16(9):1341–8. doi: 10.3201/eid1609.091800 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Greenberg DE, Porcella SF, Stock F, et al. Granulibacter bethesdensis gen. nov., sp. nov., a distinctive pathogenic acetic acid bacterium in the family Acetobacteraceae. Int J Syst Evol Microbiol. Nov 2006;56(Pt 11):2609–2616. doi: 10.1099/ijs.0.64412-0 [DOI] [PubMed] [Google Scholar]
- 32.Sirinavin S, Techasaensiri C, Benjaponpitak S, Pornkul R, Vorachit M. Invasive Chromobacterium violaceum infection in children: case report and review. Pediatr Infect Dis J. Jun 2005;24(6):559–61. doi: 10.1097/01.inf.0000164761.81491.3f [DOI] [PubMed] [Google Scholar]
- 33.Mayer EF, Gialanella P, Munjal I, Cunningham-Rundles C, Dara J. Fulminant Sepsis Due to Granulibacter bethesdensis in a 4-Year-Old Boy With X-Linked Chronic Granulomatous Disease. Pediatr Infect Dis J. Dec 2017;36(12):1165–1166. doi: 10.1097/INF.0000000000001659 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Rebelo M, Ding L, Cordeiro AI, et al. Fatal Meningitis in Patient with X-Linked Chronic Granulomatous Disease Caused by Virulent Granulibacter bethesdensis. Emerg Infect Dis. May 2019;25(5):976–979. doi: 10.3201/eid2505.181505 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Muszynski A, Zarember KA, Heiss C, et al. Granulibacter bethesdensis, a Pathogen from Patients with Chronic Granulomatous Disease, Produces a Penta-Acylated Hypostimulatory Glycero-D-talo-oct-2-ulosonic Acid-Lipid A Glycolipid (Ko-Lipid A). Int J Mol Sci. Mar 24 2021;22(7)doi: 10.3390/ijms22073303 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Straughan DM, McLoughlin KC, Mullinax JE, et al. The Changing Paradigm of Management of Liver Abscesses in Chronic Granulomatous Disease. Clin Infect Dis. Apr 17 2018;66(9):1427–1434. doi: 10.1093/cid/cix1012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lublin M, Bartlett DL, Danforth DN, et al. Hepatic abscess in patients with chronic granulomatous disease. Ann Surg. Mar 2002;235(3):383–91. doi: 10.1097/00000658-200203000-00010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Marzollo A, Conti F, Rossini L, et al. Neonatal Manifestations of Chronic Granulomatous Disease: MAS/HLH and Necrotizing Pneumonia as Unusual Phenotypes and Review of the Literature. J Clin Immunol. Feb 2022;42(2):299–311. doi: 10.1007/s10875-021-01159-4 [DOI] [PubMed] [Google Scholar]
- 39.Dorman SE, Gill VJ, Gallin JI, Holland SM. Burkholderia pseudomallei infection in a Puerto Rican patient with chronic granulomatous disease: case report and review of occurrences in the Americas. Clin Infect Dis. Apr 1998;26(4):889–94. doi: 10.1086/513928 [DOI] [PubMed] [Google Scholar]
- 40.Ross JP, Holland SM, Gill VJ, DeCarlo ES, Gallin JI. Severe Burkholderia (Pseudomonas) gladioli infection in chronic granulomatous disease: report of two successfully treated cases. Clin Infect Dis. Nov 1995;21(5):1291–3. doi: 10.1093/clinids/21.5.1291 [DOI] [PubMed] [Google Scholar]
- 41.Falcone EL, Holland SM. Invasive fungal infection in chronic granulomatous disease: insights into pathogenesis and management. Curr Opin Infect Dis. Dec 2012;25(6):658–69. doi: 10.1097/QCO.0b013e328358b0a4 [DOI] [PubMed] [Google Scholar]
- 42.Dotis J, Pana ZD, Roilides E. Non-Aspergillus fungal infections in chronic granulomatous disease. Mycoses. Jul 2013;56(4):449–62. doi: 10.1111/myc.12049 [DOI] [PubMed] [Google Scholar]
- 43.Vinh DC, Shea YR, Sugui JA, et al. Invasive aspergillosis due to Neosartorya udagawae. Clin Infect Dis. Jul 1 2009;49(1):102–11. doi: 10.1086/599345 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Vinh DC, Shea YR, Jones PA, Freeman AF, Zelazny A, Holland SM. Chronic invasive aspergillosis caused by Aspergillus viridinutans. Emerg Infect Dis. Aug 2009;15(8):1292–4. doi: 10.3201/eid1508.090251 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Sugui JA, Peterson SW, Clark LP, et al. Aspergillus tanneri sp. nov., a new pathogen that causes invasive disease refractory to antifungal therapy. J Clin Microbiol. Oct 2012;50(10):3309–17. doi: 10.1128/JCM.01509-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Haidar G, Zerbe CS, Cheng M, Zelazny AM, Holland SM, Sheridan KR. Phellinus species: An emerging cause of refractory fungal infections in patients with X-linked chronic granulomatous disease. Mycoses. Mar 2017;60(3):155–160. doi: 10.1111/myc.12573 [DOI] [PubMed] [Google Scholar]
- 47.Ameratunga R, Woon ST, Vyas J, Roberts S. Fulminant mulch pneumonitis in undiagnosed chronic granulomatous disease: a medical emergency. Clin Pediatr (Phila). Dec 2010;49(12):1143–6. doi: 10.1177/0009922810370057 [DOI] [PubMed] [Google Scholar]
- 48.Cowan J, Pakhale S, Angel JB. Mulch pneumonitis in a patient with chronic granulomatous disease. CMAJ. Nov 15 2022;194(44):E1510. doi: 10.1503/cmaj.220713 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Maaloul I, Ameur SB, Chabchoub I, et al. Fulminant mulch pneumonitis in a previously healthy child. Arch Pediatr. Nov 2018;25(8):495–496. doi: 10.1016/j.arcped.2018.09.003 [DOI] [PubMed] [Google Scholar]
- 50.Siddiqui S, Anderson VL, Hilligoss DM, et al. Fulminant mulch pneumonitis: an emergency presentation of chronic granulomatous disease. Clin Infect Dis. Sep 15 2007;45(6):673–81. doi: 10.1086/520985 [DOI] [PubMed] [Google Scholar]
- 51.Marois L, Drouin D, Leduc C, et al. Chronic granulomatous disease presenting at age 52 with fulminant mulch pneumonitis. J Allergy Clin Immunol Glob. Nov 2022;1(4):322–324. doi: 10.1016/j.jacig.2022.06.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Falcone EL, Holland SM. Gastrointestinal Complications in Chronic Granulomatous Disease. Methods Mol Biol. 2019;1982:573–586. doi: 10.1007/978-1-4939-9424-3_34 [DOI] [PubMed] [Google Scholar]
- 53.Marciano BE, Rosenzweig SD, Kleiner DE, et al. Gastrointestinal involvement in chronic granulomatous disease. Pediatrics. Aug 2004;114(2):462–8. doi: 10.1542/peds.114.2.462 [DOI] [PubMed] [Google Scholar]
- 54.Chin TW, Stiehm ER, Falloon J, Gallin JI. Corticosteroids in treatment of obstructive lesions of chronic granulomatous disease. J Pediatr. Sep 1987;111(3):349–52. doi: 10.1016/s0022-3476(87)80452-3 [DOI] [PubMed] [Google Scholar]
- 55.Bhattacharya S, Koethe Y, Ling A, et al. Gastrointestinal Computed Tomography Findings in Chronic Granulomatous Disease with Subgroup Clinicopathologic Analysis. Dig Dis Sci. May 2022;67(5):1831–1842. doi: 10.1007/s10620-021-06978-4 [DOI] [PubMed] [Google Scholar]
- 56.LaBere B, Gutierrez MJ, Wright H, et al. Chronic Granulomatous Disease With Inflammatory Bowel Disease: Clinical Presentation, Treatment, and Outcomes From the USIDNET Registry. J Allergy Clin Immunol Pract. May 2022;10(5):1325–1333 e5. doi: 10.1016/j.jaip.2021.12.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Khangura SK, Kamal N, Ho N, et al. Gastrointestinal Features of Chronic Granulomatous Disease Found During Endoscopy. Clin Gastroenterol Hepatol. Mar 2016;14(3):395–402 e5. doi: 10.1016/j.cgh.2015.10.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Alimchandani M, Lai JP, Aung PP, et al. Gastrointestinal histopathology in chronic granulomatous disease: a study of 87 patients. Am J Surg Pathol. Sep 2013;37(9):1365–72. doi: 10.1097/PAS.0b013e318297427d [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Marks DJ, Miyagi K, Rahman FZ, Novelli M, Bloom SL, Segal AW. Inflammatory bowel disease in CGD reproduces the clinicopathological features of Crohn’s disease. Am J Gastroenterol. Jan 2009;104(1):117–24. doi: 10.1038/ajg.2008.72 [DOI] [PubMed] [Google Scholar]
- 60.Bhattacharya S, Wong U, Khalid M, et al. Small bowel disease prevalence on video capsule endoscopy in chronic granulomatous disease-associated inflammatory bowel disease. J Gastroenterol Hepatol. Sep 24 2023;doi: 10.1111/jgh.16342 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Huang C, De Ravin SS, Paul AR, et al. Genetic Risk for Inflammatory Bowel Disease Is a Determinant of Crohn’s Disease Development in Chronic Granulomatous Disease. Inflamm Bowel Dis. Dec 2016;22(12):2794–2801. doi: 10.1097/MIB.0000000000000966 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.de Luca A, Smeekens SP, Casagrande A, et al. IL-1 receptor blockade restores autophagy and reduces inflammation in chronic granulomatous disease in mice and in humans. Proc Natl Acad Sci U S A. Mar 4 2014;111(9):3526–31. doi: 10.1073/pnas.1322831111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hahn KJ, Ho N, Yockey L, et al. Treatment With Anakinra, a Recombinant IL-1 Receptor Antagonist, Unlikely to Induce Lasting Remission in Patients With CGD Colitis. Am J Gastroenterol. Jun 2015;110(6):938–9. doi: 10.1038/ajg.2015.135 [DOI] [PubMed] [Google Scholar]
- 64.Chandrasekaran P, Han Y, Zerbe CS, et al. Intestinal microbiome and metabolome signatures in patients with chronic granulomatous disease. J Allergy Clin Immunol. Sep 1 2023;doi: 10.1016/j.jaci.2023.07.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Davrandi M, Harris S, Smith PJ, Murray CD, Lowe DM. The Relationship Between Mucosal Microbiota, Colitis, and Systemic Inflammation in Chronic Granulomatous Disorder. J Clin Immunol. Feb 2022;42(2):312–324. doi: 10.1007/s10875-021-01165-6 [DOI] [PubMed] [Google Scholar]
- 66.Falcone EL, Han Y, Kreuzburg S, et al. Exclusive enteral nutrition induced sustained changes in the microbiota and improved inflammatory bowel disease in a pediatric patient with chronic granulomatous disease. J Allergy Clin Immunol Pract. Feb 2021;9(2):1011–1014 e2. doi: 10.1016/j.jaip.2020.12.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Chandrasekaran P, Han Y, Zerbe CS, et al. Intestinal microbiome and metabolome signatures in patients with chronic granulomatous disease. J Allergy Clin Immunol. Aug 31 2023;doi: 10.1016/j.jaci.2023.07.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Angelino G, De Angelis P, Faraci S, et al. Inflammatory bowel disease in chronic granulomatous disease: An emerging problem over a twenty years’ experience. Pediatr Allergy Immunol. Dec 2017;28(8):801–809. doi: 10.1111/pai.12814 [DOI] [PubMed] [Google Scholar]
- 69.Uzel G, Orange JS, Poliak N, Marciano BE, Heller T, Holland SM. Complications of tumor necrosis factor-alpha blockade in chronic granulomatous disease-related colitis. Clin Infect Dis. Dec 15 2010;51(12):1429–34. doi: 10.1086/657308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Conrad A, Neven B, Mahlaoui N, et al. Infections in Patients with Chronic Granulomatous Disease Treated with Tumor Necrosis Factor Alpha Blockers for Inflammatory Complications. J Clin Immunol. Jan 2021;41(1):185–193. doi: 10.1007/s10875-020-00901-8 [DOI] [PubMed] [Google Scholar]
- 71.Campbell N, Chapdelaine H. Treatment of chronic granulomatous disease-associated fistulising colitis with vedolizumab. J Allergy Clin Immunol Pract. Nov-Dec 2017;5(6):1748–1749. doi: 10.1016/j.jaip.2017.07.035 [DOI] [PubMed] [Google Scholar]
- 72.Kamal N, Marciano B, Curtin B, et al. The response to vedolizumab in chronic granulomatous disease-related inflammatory bowel disease. Gastroenterol Rep (Oxf). Oct 2020;8(5):404–406. doi: 10.1093/gastro/goaa005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Bhattacharya S, Marciano BE, Malech HL, et al. Safety and Efficacy of Ustekinumab in the Inflammatory Bowel Disease of Chronic Granulomatous Disease. Clin Gastroenterol Hepatol. Feb 2022;20(2):461–464 e2. doi: 10.1016/j.cgh.2021.03.039 [DOI] [PubMed] [Google Scholar]
- 74.Butte MJ, Park KT, Lewis DB. Treatment of CGD-associated Colitis with the IL-23 Blocker Ustekinumab. J Clin Immunol. Oct 2016;36(7):619–20. doi: 10.1007/s10875-016-0318-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Marsh RA, Leiding JW, Logan BR, et al. Chronic Granulomatous Disease-Associated IBD Resolves and Does Not Adversely Impact Survival Following Allogeneic HCT. J Clin Immunol. Oct 2019;39(7):653–667. doi: 10.1007/s10875-019-00659-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Gungor T, Teira P, Slatter M, et al. Reduced-intensity conditioning and HLA-matched haemopoietic stem-cell transplantation in patients with chronic granulomatous disease: a prospective multicentre study. Lancet. Feb 1 2014;383(9915):436–48. doi: 10.1016/S0140-6736(13)62069-3 [DOI] [PubMed] [Google Scholar]
- 77.Slatter MA, Gennery AR. Haematopoietic Stem Cell Transplantation for Chronic Granulomatous Disease. J Clin Med. Sep 20 2023;12(18)doi: 10.3390/jcm12186083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Leiding JW, Arnold DE, Parikh SH, et al. Genotype, Oxidase Status, and Preceding Infection or Autoinflammation Do Not Affect Allogeneic HCT Outcomes for CGD. Blood. Aug 10 2023;doi: 10.1182/blood.2022019586 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Hussain N, Feld JJ, Kleiner DE, et al. Hepatic abnormalities in patients with chronic granulomatous disease. Hepatology. Mar 2007;45(3):675–83. doi: 10.1002/hep.21524 [DOI] [PubMed] [Google Scholar]
- 80.Magnani A, Brosselin P, Beaute J, et al. Inflammatory manifestations in a single-center cohort of patients with chronic granulomatous disease. J Allergy Clin Immunol. Sep 2014;134(3):655–662 e8. doi: 10.1016/j.jaci.2014.04.014 [DOI] [PubMed] [Google Scholar]
- 81.Salvator H, Mahlaoui N, Catherinot E, et al. Pulmonary manifestations in adult patients with chronic granulomatous disease. Eur Respir J. Jun 2015;45(6):1613–23. doi: 10.1183/09031936.00118414 [DOI] [PubMed] [Google Scholar]
- 82.Jones LB, McGrogan P, Flood TJ, et al. Special article: chronic granulomatous disease in the United Kingdom and Ireland: a comprehensive national patient-based registry. Clin Exp Immunol. May 2008;152(2):211–8. doi: 10.1111/j.1365-2249.2008.03644.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Claps A, Della Corte M, Gerocarni Nappo S, Francalanci P, Palma P, Finocchi A. How should eosinophilic cystitis be treated in patients with chronic granulomatous disease? Pediatr Nephrol. Nov 2014;29(11):2229–33. doi: 10.1007/s00467-014-2883-7 [DOI] [PubMed] [Google Scholar]
- 84.Chen TA, Rayess N, Afshar AR, Moshfeghi DM, Duncan JL, de Alba Campomanes AG. Chorioretinal Findings as the Initial Presentation of Chronic Granulomatous Disease. Ophthalmic Surg Lasers Imaging Retina. Apr 2022;53(4):234–238. doi: 10.3928/23258160-20220315-01 [DOI] [PubMed] [Google Scholar]
- 85.Kim SJ, Kim JG, Yu YS. Chorioretinal lesions in patients with chronic granulomatous disease. Retina. Jun 2003;23(3):360–5. doi: 10.1097/00006982-200306000-00012 [DOI] [PubMed] [Google Scholar]
- 86.Chiesa R, Wang J, Blok HJ, et al. Hematopoietic cell transplantation in chronic granulomatous disease: a study of 712 children and adults. Blood. Sep 3 2020;136(10):1201–1211. doi: 10.1182/blood.2020005590 [DOI] [PubMed] [Google Scholar]
- 87.Soncini E, Slatter MA, Jones LB, et al. Unrelated donor and HLA-identical sibling haematopoietic stem cell transplantation cure chronic granulomatous disease with good long-term outcome and growth. Br J Haematol. Apr 2009;145(1):73–83. doi: 10.1111/j.1365-2141.2009.07614.x [DOI] [PubMed] [Google Scholar]
- 88.Schuetz C, Hoenig M, Gatz S, et al. Hematopoietic stem cell transplantation from matched unrelated donors in chronic granulomatous disease. Immunol Res. 2009;44(1–3):35–41. doi: 10.1007/s12026-008-8068-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Kohn DB, Booth C, Kang EM, et al. Lentiviral gene therapy for X-linked chronic granulomatous disease. Nat Med. Feb 2020;26(2):200–206. doi: 10.1038/s41591-019-0735-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.De Ravin SS, Li L, Wu X, et al. CRISPR-Cas9 gene repair of hematopoietic stem cells from patients with X-linked chronic granulomatous disease. Sci Transl Med. Jan 11 2017;9(372)doi: 10.1126/scitranslmed.aah3480 [DOI] [PubMed] [Google Scholar]
- 91.Klatt D, Cheng E, Philipp F, et al. Targeted Repair of p47-CGD in iPSCs by CRISPR/Cas9: Functional Correction without Cleavage in the Highly Homologous Pseudogenes. Stem Cell Reports. Oct 8 2019;13(4):590–598. doi: 10.1016/j.stemcr.2019.08.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Klebanoff SJ, Kettle AJ, Rosen H, Winterbourn CC, Nauseef WM. Myeloperoxidase: a front-line defender against phagocytosed microorganisms. J Leukoc Biol. Feb 2013;93(2):185–98. doi: 10.1189/jlb.0712349 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hoyos S, Agudelo CC, Lozano DM, Buitrago LE. Systemic paracoccidioidomycosis in a patient with enzymatic myeloperoxidase deficiency and acute lymphoblastic leukemia: case report. Bull Natl Res Cent. 2022;46(1):286. doi: 10.1186/s42269-022-00973-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Milligan KL, Mann D, Rump A, et al. Complete Myeloperoxidase Deficiency: Beware the “False-Positive” Dihydrorhodamine Oxidation. J Pediatr. Sep 2016;176:204–6. doi: 10.1016/j.jpeds.2016.05.047 [DOI] [PubMed] [Google Scholar]
- 95.Chiang AK, Chan GC, Ma SK, Ng YK, Ha SY, Lau YL. Disseminated fungal infection associated with myeloperoxidase deficiency in a premature neonate. Pediatr Infect Dis J. Oct 2000;19(10):1027–9. doi: 10.1097/00006454-200010000-00024 [DOI] [PubMed] [Google Scholar]
- 96.Kutter D, Devaquet P, Vanderstocken G, Paulus JM, Marchal V, Gothot A. Consequences of total and subtotal myeloperoxidase deficiency: risk or benefit ? Acta Haematol. 2000;104(1):10–5. doi: 10.1159/000041062 [DOI] [PubMed] [Google Scholar]
- 97.Kremserova S, Kocurkova A, Chorvatova M, Klinke A, Kubala L. Myeloperoxidase Deficiency Alters the Process of the Regulated Cell Death of Polymorphonuclear Neutrophils. Front Immunol. 2022;13:707085. doi: 10.3389/fimmu.2022.707085 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Cappellini MD, Fiorelli G. Glucose-6-phosphate dehydrogenase deficiency. Lancet. Jan 5 2008;371(9606):64–74. doi: 10.1016/S0140-6736(08)60073-2 [DOI] [PubMed] [Google Scholar]
- 99.TeSlaa T, Ralser M, Fan J, Rabinowitz JD. The pentose phosphate pathway in health and disease. Nat Metab. Aug 2023;5(8):1275–1289. doi: 10.1038/s42255-023-00863-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.van Bruggen R, Bautista JM, Petropoulou T, et al. Deletion of leucine 61 in glucose-6-phosphate dehydrogenase leads to chronic nonspherocytic anemia, granulocyte dysfunction, and increased susceptibility to infections. Blood. Aug 1 2002;100(3):1026–30. doi: 10.1182/blood.v100.3.1026 [DOI] [PubMed] [Google Scholar]
- 101.Ley K, Hoffman HM, Kubes P, et al. Neutrophils: New insights and open questions. Sci Immunol. Dec 7 2018;3(30)doi: 10.1126/sciimmunol.aat4579 [DOI] [PubMed] [Google Scholar]
- 102.Sadik CD, Kim ND, Luster AD. Neutrophils cascading their way to inflammation. Trends Immunol. Oct 2011;32(10):452–60. doi: 10.1016/j.it.2011.06.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Sandor N, Lukacsi S, Ungai-Salanki R, et al. CD11c/CD18 Dominates Adhesion of Human Monocytes, Macrophages and Dendritic Cells over CD11b/CD18. PLoS One. 2016;11(9):e0163120. doi: 10.1371/journal.pone.0163120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Fekadu J, Modlich U, Bader P, Bakhtiar S. Understanding the Role of LFA-1 in Leukocyte Adhesion Deficiency Type I (LAD I): Moving towards Inflammation? Int J Mol Sci. Mar 25 2022;23(7)doi: 10.3390/ijms23073578 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Hanna S, Etzioni A. Leukocyte adhesion deficiencies. Ann N Y Acad Sci. Feb 2012;1250:50–5. doi: 10.1111/j.1749-6632.2011.06389.x [DOI] [PubMed] [Google Scholar]
- 106.Roos D, van Leeuwen K, Madkaikar M, et al. Hematologically important mutations: Leukocyte adhesion deficiency (second update). Blood Cells Mol Dis. Mar 2023;99:102726. doi: 10.1016/j.bcmd.2023.102726 [DOI] [PubMed] [Google Scholar]
- 107.Stark MA, Huo Y, Burcin TL, Morris MA, Olson TS, Ley K. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity. Mar 2005;22(3):285–94. doi: 10.1016/j.immuni.2005.01.011 [DOI] [PubMed] [Google Scholar]
- 108.Moutsopoulos NM, Zerbe CS, Wild T, et al. Interleukin-12 and Interleukin-23 Blockade in Leukocyte Adhesion Deficiency Type 1. N Engl J Med. Mar 23 2017;376(12):1141–1146. doi: 10.1056/NEJMoa1612197 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Bakhtiar S, Salzmann-Manrique E, Blok HJ, et al. Allogeneic hematopoietic stem cell transplantation in leukocyte adhesion deficiency type I and III. Blood Adv. Jan 12 2021;5(1):262–273. doi: 10.1182/bloodadvances.2020002185 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kohn LA, Kohn DB. Gene Therapies for Primary Immune Deficiencies. Front Immunol. 2021;12:648951. doi: 10.3389/fimmu.2021.648951 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Tahata S, Raymond K, Quade M, et al. Defining the mild variant of leukocyte adhesion deficiency type II (SLC35C1-congenital disorder of glycosylation) and response to l-fucose therapy: Insights from two new families and review of the literature. Am J Med Genet A. Jul 2022;188(7):2005–2018. doi: 10.1002/ajmg.a.62737 [DOI] [PubMed] [Google Scholar]
- 112.Alon R, Etzioni A. LAD-III, a novel group of leukocyte integrin activation deficiencies. Trends Immunol. Oct 2003;24(10):561–6. doi: 10.1016/j.it.2003.08.001 [DOI] [PubMed] [Google Scholar]
- 113.Etzioni A, Alon R. Leukocyte adhesion deficiency III: a group of integrin activation defects in hematopoietic lineage cells. Curr Opin Allergy Clin Immunol. Dec 2004;4(6):485–90. doi: 10.1097/00130832-200412000-00003 [DOI] [PubMed] [Google Scholar]
- 114.Xu Z, Jobe SM, Ma YQ, Shavit JA. A novel LAD-III mutation manifests functional importance of the compact FERM domain in Kindlin-3. J Thromb Haemost. Oct 20 2023;doi: 10.1016/j.jtha.2023.10.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Koker N, Deveci I, van Leeuwen K, et al. A Novel Deletion in FERMT3 Causes LAD-III in a Turkish Family. J Clin Immunol. May 2023;43(4):741–746. doi: 10.1007/s10875-022-01420-4 [DOI] [PubMed] [Google Scholar]
- 116.Qasim W, Cavazzana-Calvo M, Davies EG, et al. Allogeneic hematopoietic stem-cell transplantation for leukocyte adhesion deficiency. Pediatrics. Mar 2009;123(3):836–40. doi: 10.1542/peds.2008-1191 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Al-Dhekri H, Al-Mousa H, Ayas M, et al. Allogeneic hematopoietic stem cell transplantation in leukocyte adhesion deficiency type 1: a single center experience. Biol Blood Marrow Transplant. Aug 2011;17(8):1245–9. doi: 10.1016/j.bbmt.2010.12.714 [DOI] [PubMed] [Google Scholar]
- 118.Horikoshi Y, Umeda K, Imai K, et al. Allogeneic Hematopoietic Stem Cell Transplantation for Leukocyte Adhesion Deficiency. J Pediatr Hematol Oncol. Mar 2018;40(2):137–140. doi: 10.1097/MPH.0000000000001028 [DOI] [PubMed] [Google Scholar]
