Minireviews Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Methodol. Dec 20, 2024; 14(4): 96380
Published online Dec 20, 2024. doi: 10.5662/wjm.v14.i4.96380
Relation between dysbiosis and inborn errors of immunity
Öner Özdemir, Department of Pediatric Allergy and Immunology, Sakarya University, Medical Faculty, Adapazarı 54100, Sakarya, Türkiye
ORCID number: Öner Özdemir (0000-0002-5338-9561).
Author contributions: Özdemir Ö has done everything.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Öner Özdemir, MD, Full Professor, Department of Pediatric Allergy and Immunology, Sakarya University Research and Training Hospital, Adnan Menderes Cad, Adapazarı 54100, Sakarya, Türkiye. ozdemir_oner@hotmail.com
Received: May 5, 2024
Revised: May 27, 2024
Accepted: June 14, 2024
Published online: December 20, 2024
Processing time: 81 Days and 19.9 Hours

Abstract

Inborn errors of immunity (IEI) disorders, formerly primary immune deficiency diseases, are a heterogeneous group of disorders with variable hereditary transitions, clinical manifestations, complications and varying disease severity. Many of the clinical symptoms, signs and complications in IEI patients can be attributed to inflammatory and immune dysregulatory processes due to loss of microbial diversity (dysbiosis). For example, in common variable immunodeficiency patients, the diversity of bacteria, but not fungi, in the gut microbiota has been found to be reduced and significantly altered. Again, this was associated with a more severe disease phenotype. Compromise of the STAT3/Th17 pathway in hyper-IgE syndrome may lead to dysbiosis of the oral microbiota in these patients, causing Candida albicans to switch from commensal to pathogenic. Modification of the microbiota can be used as a therapeutic approach in patients with IEI. Prebiotics, probiotics, postbiotics and fecal microbiota transplantation can be used to restore the balance of the gut microbiota and reduce pathogenicity in IEI patients. Clinical trials are currently underway to understand the impact of this dysbiosis on the phenotype of IEI diseases and its role in their treatment.

Key Words: Immunodeficiency; Microbiome; Microbiota; Fecal microbiota transplantation; Immunoglobulin

Core Tip: Inborn errors of immunity (IEI) disease is associated with microbial dysbiosis and systemic inflammation, especially in the presence of immune dysregulation. Fecal microbiota transplantation in particular, besides not being used as a stand-alone treatment strategy, could be a potential therapeutic tool for dysbiosis in IEI, considering the complications and the complexity of IEI.



INTRODUCTION

Inborn errors of immunity (IEI, formerly primary immunodeficiency) diseases are a heterogeneous group of disorders with variable genetic transmissions, clinical manifestations, complications, and varying disease severity[1]. Many of the clinical symptoms, signs, and complications in IEI patients can be ascribed to inflammatory and immune dysregulation processes due to loss of microbial diversity (dysbiosis) and imbalances in the diet-microbiome-host-immune system axis[2]. The microbiome in IEI patients is reported to have a key role in serving the host immune system preserve homeostasis in the gut. In particular, the intestinal microbiome is a favorable area of study and interest in these aspects in IEI patients. Although the association between the microbiome and humoral immunodeficiency is better known, the relationship between combined immunodeficiency and the microbiome and its possible therapeutic benefits are still under investigation[2,3]. Additional studies investigating the efficacy of microbiota-based approaches and treatments in IEI-related dysbiosis in these patients are needed[2-4].

The contemporary understanding of the microbiome concerning the clinical symptoms and complications found in IEI patients and its potential as a therapeutic approach in IEI forms are the main basis of the aim of this minireview article.

Evaluation tools of microbiome/microbiota in studies

When carrying out microbiome studies, it is important to assess alpha diversity in the sample (single ecosystem) to be studied and beta diversity among individuals. It indicates the quantity and their prevalence of diverse taxa detected in each sample. Beta diversity is an indicator of the variety of the microbial population among different subjects or samples. It is also a marker for the number of different taxa and their prevalence in an environment. In brief, the average species variety of a place at the local measure is called alpha diversity[2,4]. Beta diversity reflects the diversity ratio between regional and local species[2,4].

Microbiome in most common IEI diseases

Here, microbiome and microbiota studies in more common and better-known IEI diseases will be presented in the light of current literature data (Table 1).

Table 1 Microbial dysbiosis on the clinical implications in various inborn errors of immunity.
Inborn errors of immunity/diseases
Microbiota
Clinical findings
Ref.
Hyper-IgE syndrome Predominance of Candida albicans, decreased abundance of C.
parapsilosis, Boletus, and Penicillium
STAT3/Th17 axis play an
important role in maintaining C. albicans as a commensal organism
[28]
Wiskott–Aldrich syndromeIncreased abundance of potentially, pathogenic Proteobacteria and
Roteobacteria. Decreased levels of
protective commensals, e.g., Faecalibacterium prausnitzii, Bacteroidetes and Verrucomicrobia
It may lead to periodontal lesions[18]
Severe combined immunodeficiencyIncreased abundances of Escherichia, Staphylococcus, Enterococcus, Veillonella, Enterobacteriaceae, Adenovirus, and BocavirusIncrease in disease severity[16,18]
Selective IgA deficiencyHigher abundance of Firmicutes, Bacteroidetes, Gammaproteobacteria and PrevotellaIncrease in systemic inflammation[1,6,24]
Common variable immunodeficiency Decreased abundance of beneficial bacteria, e.g., Bifidobacterium and Lactobacillus, Bacteroides and Firmicutes. Increased abundance
of Clostridia, Bacilli, Prevotella, and Gammaproteobacteria
Increase in systemic inflammation[10,14]
Common variable immunodeficiency

The foremost gastrointestinal tract symptom of common variable immunodeficiency (CVID), which is one of the most common immunodeficiencies, is temporary or persistent diarrhea seen in 21%-57% of cases[5]. Detectable pathogens include Cytomegalovirus, Salmonella species, Clostridium difficile, Giardia lamblia, Cryptosporidium parvum, and Campylobacter jejuni[6]. In some studies, Hungatella hathewayi from normal gut microbiota has been related to CVID, like some chronic inflammatory diseases (such as cystic fibrosis and IgA nephropathy)[7]. CVID patients with chronic diarrhea were found to have lower alpha diversity compared to healthy housemates or CVID patients without diarrhea[8]. New research indicates that the immune dysregulation yielding difficulties in CVID patients might be the result of a changed microbiome arrangement and augmented microbial translocation[2].

Decreased bacterial alpha diversity was also found in the more severe CVID phenotype with undetectable low serum IgA levels[2]. IgA production against commensal microbes occurs in Peyer’s patches (PP) where live bacteria are presented by dendritic cell (DC) to B and T cells. Furthermore, secretory IgA selectively adheres to M cells in the gut PP and contributes to exciting the acceptance of IgA-bound pathogen antigens for distribution to DCs, ensuring a positive feedback loop. IgA thus has an essential role in controlling the pro-inflammatory reaction to the bacteria it covers, in establishing and maintaining the integrity of the gut mucosal barrier, and in determining the content of the gut microbiota. IgA-deprived mice were found to have meaningfully lesser gut microbial diversity than wild-type offspring[9].

IgA deficiency detected in CVID patients is highly associated with augmented morbidity from inflammation. Intestinal epithelial damage occurring in CVID cases may be caused by IgA deficiency leading to mucus invasion and epithelial infection. The alpha diversity index of cases with severe IgA deficiency was meaningfully decreased compared with CVID patients with low/normal IgA levels. The 3 bacterial taxa that were more copious in CVID patients than in healthy donors were Clostridia (genera Lachnospiraceae Roseburia and Lachnospiraceae Dorea), Bacilli and Gammaproteobacteria. In contrast, Firmicutes (genus Blautia of Lachnospiraceae and family Christensenellaceae), Actinobacteria (e.g., family Bifidobacteriaceae), and Deltaproteobacteria (genus Desulfovibrionales) were detected to be greatly reduced in CVID[10].

One of the groundbreaking types of research on the microbiome in CVID cases showed low levels of alpha diversity and Bifidobacterium species. However, increased Bacilli, Clostridia, and Gammaproteobacteria species were detected in these cases[10,11]. Among the 3 diverse bacterial taxa, Geobacillus, Acinetobacter baumannii and the otu 15570 bacterium are among those that may contribute to CVID enteropathy[12].

The presence of dysbiosis and reduced alpha diversity is well known, especially in diseases such as CVID. There is no significant difference in beta diversity between adults and children, but alpha diversity is lesser in children than in adults. When compared to adults, the microbiota configuration in children shows a greater diversity of Ruminococcaceae, Bacilli, Actinobacteria, and Bacteroidetes phyla and a lesser diversity of Methanobacteriales phyla[13].

CVID patients have shown a less diverse and significantly altered bacterial, but not fungal, gut microbiota. Again, this has been associated with a more severe disease phenotype[14].

Selective IgA deficiency

Using a metagenomic approach, the researchers compared the intestinal microbial groups in the feces of 21 IgA-deficient cases with 34 age- and sex-matched healthy controls. Seventeen microbial species in the phyla Firmicutes, Bacteroidetes, and Proteobacteria (Gammaproteobacteria only, comprising E. coli) were greater in IgA-deficient patients. Moreover, 3 oral commensal bacteria (Haemophilus parainfluenza, Streptococcus sanguinis and Veillonella parvula) and two Prevotella species were also found at high rates in the selective IgA deficiency cohort. Most of the bacteria lost in IgA-deficient patients (13 of 14) belonged to the phylum Firmicutes (genus Faecalibacterium and family Lachnospiraceae), while one belonged to the phylum Bacteroides. In short, IgA deficiency caused a loss of some typical favorable symbionts and an increase in pathobionts[15].

Faecalibacterium, a genus with anti-inflammatory effects for the intestinal mucosa, was markedly reduced in patients with inflammatory bowel disease (IBD). However, the pro-inflammatory species, Gammaproteobacteria and Prevotella, are increased in patients with selective IgA deficiency. Analysis of the gut microbiota in these cases shows that the oral microbiota is ectopically localized in the lower digestive tract[6].

Severe combined immunodeficiency

A study showed that the bacterial taxonomy of the intestinal microbiota in severe combined immunodeficiency patients changes in long periods of time, resulting in altered microbiome spectrum before and after hematopoietic stem-cell transplantation (HSCT)[6]. Graft-vs-host disease, which can also be seen after HSCT, is affected by different factors, plus the intestinal microbiota. Decreased microbial miscellany after HSCT causes the predominance of Staphylococcus, Escherichia, and Enterococcus species[16,17].

Wiskott–Aldrich syndrome

Bacteroidetes and Verrucomicrobia were less abundant in patients with Wiskott–Aldrich syndrome (WAS), whereas Proteobacteria were significantly more abundant compared to healthy controls[18,19]. Roughly 10% of WAS patients live with dysbiosis causing severe, sporadic, and recurrent gastrointestinal inflammation. Therefore, WAS cases are inclined to develop an acute and early IBD that phenotypically resembles polygenetic IBD[4].

Hyper-IgE syndrome

Candida albicans are more common in autosomal dominant (AD)- Hyper-IgE syndrome (HIES) patients than others (e.g., C. parapsilosis, Boletus, and Penicillium) compared to healthy patients. AD-HIES patients have severe dysbiosis with Candida albicans predominating during active fungal infection. In uninfected patients, the genus Malassezia was predominant. A modification of the skin microbiota towards gram-negative colonization (especially Acinetobacter spp.) is linked with a weak in vitro immune reaction to C. albicans and S. aureus[18].

Immunodeficient cases with defects in the Th17/IL-17 pathway are more inclined to propagate oral fungal infections. Patients with HIES due to STAT3 deficiency have been reported to have low salivary antifungal activity as a result of the reduction of specific antimicrobial effectors containing human β-defensin 2 and multiple histatins. In these patients, the STAT3-Th17 axis is important for C. albicans commensalism and the formation of oral bacterial groups. In these cases, C. albicans forms a symbiotic life with orally present Streptococcus mutans and Streptococcus oralis[18]. A compromised STAT3/Th17 pathway may lead to dysbiosis of the oral microbiota in these patients, causing C. albicans to shift from commensal to pathogenic[4,20]. Selected oral streptococci can increase the virulence of C. albicans by invading oral tissue and causing mucosal lesions[6]. Another study reported a decrease in abundant gram-negative bacteria (such as Fusobacteria and Prevotella) in oral swabs of HIES patients.

In a study, the oral microbiome of a large cohort of 36 patients with AD-HIES (STAT3 deficiency) was analyzed. Oral bacterial groups were also found to be dysbiotic in AD-HIES, especially when active Candida infection was present. Many prevalent oral commensal bacteria, comprising Porphyromonas, Neisseria, and Haemophilus, had lesser comparative copiousness in autosomal dominat-HIES patients, even though the genus Capnocytophaga was unequally represented[19,20].

Management

Modification of the microbiota can be used as a therapeutic approach in IEI patients[4]. Prebiotics, probiotics, postbiotics, and fecal microbiota transplantation (FMT) have been established to renovate the equilibrium of intestinal microbiota and reduce pathogenicity in IEI patients[4,21,22].

Dysbiosis can be treated by oral intake of live, natural probiotic bacteria. Metabolites from prebiotic fibers serve as a material for probiotic commensal bacteria. The ability of prebiotics to modify immunity has been utilized to produce complementary immunomodulatory therapies for a range of IEIs. Probiotics increase the manufacture of bioactive peptides by enabling the interaction of gut epithelial and mucosal immune cells and intestinal microbiota and help protect intestinal epithelial barriers[4].

Secretory IgA and systemic IgG are known to act synergistically in defense against gut microbiota. High serum levels of IgG directed against gut microbiota have been detected in animal models of IEI diseases. Furthermore, IgG antibodies to E. coli have been detected in IBD cases and secretory IgA-deficient mice[23]. Fadlallah et al[24] have also shown that serum anti-microbiota IgG is increased in cases with selective IgA deficiency compared to controls[24]. These circulating IgGs are formed against preserved antigenic motifs and protect against commensal bacteria. It is thought that intravenous immunoglobulin products consisting of selective IgA-deficient individuals in the pool may increase protection against dysbiosis and microbial displacement in CVID patients[4,16,24].

FMT involves the administration of healthy donor feces to the patient to restore spoiled intestinal microbiota and provide therapeutic profits. FMT, which is the standard for the treatment of recurrent Clostridium difficile infections, may not be used as a single treatment modality but as likely management of immune dysregulation in IEI, considering its side effects and the complexity of IEI[2]. A case with immune dysregulation, polyendocrinopathy, enteropathy, X-linked disease, severe enteropathy, and chronic diarrhea was reported to be efficaciously treated with FMT[16,25]. The disadvantage of this type of therapeutic management includes procedure-related risks, such as insufficient prior screening of FMT for multidrug-resistant microorganisms, especially for immunocompromised recipients[2,26]. Gut dysbiosis has been observed in germ-free mice transplanted with feces from CVID cases with non-infectious problems. No dysbiosis was observed with FMT using only feces from CVID cases with an infectious phenotype or from household contacts. Intestinal dysbiosis in non-infectious CVID cases is seen with augmented richness of Dysgonomonas mossii and Bacillus massiliensis[2,27,28].

CONCLUSION

The presence of dysbiosis in different types of IEI patients has been demonstrated in different clinical studies. Studies are ongoing to understand the role of this dysbiosis on the phenotype of the IEI diseases and in their treatment.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Medical laboratory technology

Country of origin: Türkiye

Peer-review report’s classification

Scientific Quality: Grade D

Novelty: Grade C

Creativity or Innovation: Grade C

Scientific Significance: Grade C

P-Reviewer: Belkova N S-Editor: Liu H L-Editor: Filipodia P-Editor: Zhang XD

References
1.  Bousfiha A, Moundir A, Tangye SG, Picard C, Jeddane L, Al-Herz W, Rundles CC, Franco JL, Holland SM, Klein C, Morio T, Oksenhendler E, Puel A, Puck J, Seppänen MRJ, Somech R, Su HC, Sullivan KE, Torgerson TR, Meyts I. The 2022 Update of IUIS Phenotypical Classification for Human Inborn Errors of Immunity. J Clin Immunol. 2022;42:1508-1520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 150]  [Reference Citation Analysis (0)]
2.  Al-Nesf MA, Morgan D, Mohamed-Ali V. Primary immunodeficiency and the microbiome. Curr Opin Pediatr. 2021;33:633-638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
3.  Cabanero-Navalon MD, Garcia-Bustos V, Mira A, Moral Moral P, Salavert-Lleti M, Forner Giner MJ, Núñez Beltrán M, Todolí Parra J, Bracke C, Carda-Diéguez M. Dysimmunity in common variable immunodeficiency is associated with alterations in oral, respiratory, and intestinal microbiota. Clin Immunol. 2023;256:109796.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
4.  Sharma M, Dhaliwal M, Tyagi R, Goyal T, Sharma S, Rawat A. Microbiome and Its Dysbiosis in Inborn Errors of Immunity. Pathogens. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
5.  Cunningham-Rundles C. How I treat common variable immune deficiency. Blood. 2010;116:7-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 196]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
6.  Pellicciotta M, Rigoni R, Falcone EL, Holland SM, Villa A, Cassani B. The microbiome and immunodeficiencies: Lessons from rare diseases. J Autoimmun. 2019;98:132-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 29]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
7.  Bosák J, Lexa M, Fiedorová K, Gadara DC, Micenková L, Spacil Z, Litzman J, Freiberger T, Šmajs D. Patients With Common Variable Immunodeficiency (CVID) Show Higher Gut Bacterial Diversity and Levels of Low-Abundance Genes Than the Healthy Housemates. Front Immunol. 2021;12:671239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
8.  van Schewick CM, Nöltner C, Abel S, Burns SO, Workman S, Symes A, Guzman D, Proietti M, Bulashevska A, Moreira F, Soetedjo V, Lowe DM, Grimbacher B. Altered Microbiota, Impaired Quality of Life, Malabsorption, Infection, and Inflammation in CVID Patients With Diarrhoea. Front Immunol. 2020;11:1654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
9.  Kawamoto S, Maruya M, Kato LM, Suda W, Atarashi K, Doi Y, Tsutsui Y, Qin H, Honda K, Okada T, Hattori M, Fagarasan S. Foxp3(+) T cells regulate immunoglobulin a selection and facilitate diversification of bacterial species responsible for immune homeostasis. Immunity. 2014;41:152-165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 344]  [Cited by in F6Publishing: 283]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
10.  Jørgensen SF, Trøseid M, Kummen M, Anmarkrud JA, Michelsen AE, Osnes LT, Holm K, Høivik ML, Rashidi A, Dahl CP, Vesterhus M, Halvorsen B, Mollnes TE, Berge RK, Moum B, Lundin KE, Fevang B, Ueland T, Karlsen TH, Aukrust P, Hov JR. Altered gut microbiota profile in common variable immunodeficiency associates with levels of lipopolysaccharide and markers of systemic immune activation. Mucosal Immunol. 2016;9:1455-1465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 101]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
11.  Skarpengland T, Macpherson ME, Hov JR, Kong XY, Bohov P, Halvorsen B, Fevang B, Berge RK, Aukrust P, Jørgensen SF. Altered Plasma Fatty Acids Associate with Gut Microbial Composition in Common Variable Immunodeficiency. J Clin Immunol. 2022;42:146-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
12.  Tam JS, Routes JM. Common variable immunodeficiency. Am J Rhinol Allergy. 2013;27:260-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
13.  Franco-Esquivias AP, Peña CG, Torres-Lozano C, Vaca-Paniagua F, Díaz-Velásquez C, Ortega-Cisneros M, Quintero-Ramos A. Gut microbiota in Mexican patients with common variable immunodeficiency. Gac Med Mex. 2019;155:447-452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
14.  Fiedorová K, Radvanský M, Bosák J, Grombiříková H, Němcová E, Králíčková P, Černochová M, Kotásková I, Lexa M, Litzman J, Šmajs D, Freiberger T. Bacterial but Not Fungal Gut Microbiota Alterations Are Associated With Common Variable Immunodeficiency (CVID) Phenotype. Front Immunol. 2019;10:1914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
15.  Fadlallah J, El Kafsi H, Sterlin D, Juste C, Parizot C, Dorgham K, Autaa G, Gouas D, Almeida M, Lepage P, Pons N, Le Chatelier E, Levenez F, Kennedy S, Galleron N, de Barros JP, Malphettes M, Galicier L, Boutboul D, Mathian A, Miyara M, Oksenhendler E, Amoura Z, Doré J, Fieschi C, Ehrlich SD, Larsen M, Gorochov G. Microbial ecology perturbation in human IgA deficiency. Sci Transl Med. 2018;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 175]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
16.  Castagnoli R, Pala F, Bosticardo M, Licari A, Delmonte OM, Villa A, Marseglia GL, Notarangelo LD. Gut Microbiota-Host Interactions in Inborn Errors of Immunity. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
17.  Lane JP, Stewart CJ, Cummings SP, Gennery AR. Gut microbiome variations during hematopoietic stem cell transplant in severe combined immunodeficiency. J Allergy Clin Immunol. 2015;135:1654-1656.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
18.  Hazime R, Eddehbi FE, El Mojadili S, Lakhouaja N, Souli I, Salami A, M'Raouni B, Brahim I, Oujidi M, Guennouni M, Bousfiha AA, Admou B. Inborn errors of immunity and related microbiome. Front Immunol. 2022;13:982772.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
19.  Zhang L, Li YY, Tang X, Zhao X. Faecal microbial dysbiosis in children with Wiskott-Aldrich syndrome. Scand J Immunol. 2020;91:e12805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
20.  Abusleme L, Diaz PI, Freeman AF, Greenwell-Wild T, Brenchley L, Desai JV, Ng WI, Holland SM, Lionakis MS, Segre JA, Kong HH, Moutsopoulos NM. Human defects in STAT3 promote oral mucosal fungal and bacterial dysbiosis. JCI Insight. 2018;3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 40]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
21.  Jagessar SAR, Long C, Cui B, Zhang F. Improvement of Good's syndrome by fecal microbiota transplantation: the first case report. J Int Med Res. 2019;47:3408-3415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
22.  Poto R, Laniro G, de Paulis A, Spadaro G, Marone G, Gasbarrini A, Varricchi G. Is there a role for microbiome-based approach in common variable immunodeficiency? Clin Exp Med. 2023;23:1981-1998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
23.  Castro-Dopico T, Clatworthy MR. Mucosal IgG in inflammatory bowel disease - a question of (sub)class? Gut Microbes. 2020;12:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
24.  Fadlallah J, Sterlin D, Fieschi C, Parizot C, Dorgham K, El Kafsi H, Autaa G, Ghillani-Dalbin P, Juste C, Lepage P, Malphettes M, Galicier L, Boutboul D, Clément K, André S, Marquet F, Tresallet C, Mathian A, Miyara M, Oksenhendler E, Amoura Z, Yssel H, Larsen M, Gorochov G. Synergistic convergence of microbiota-specific systemic IgG and secretory IgA. J Allergy Clin Immunol. 2019;143:1575-1585.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 61]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
25.  Wu W, Shen N, Luo L, Deng Z, Chen J, Tao Y, Mo X, Cao Q. Fecal microbiota transplantation before hematopoietic stem cell transplantation in a pediatric case of chronic diarrhea with a FOXP3 mutation. Pediatr Neonatol. 2021;62:172-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
26.  Gupta S, Mullish BH, Allegretti JR. Fecal Microbiota Transplantation: The Evolving Risk Landscape. Am J Gastroenterol. 2021;116:647-656.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 30]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
27.  Hajjar J, Voigt A, Conner M, Swennes A, Fowler S, Calarge C, Mendonca D, Armstrong D, Chang CY, Walter J, Butte M, Savidge T, Oh J, Kheradmand F, Petrosino J. Common Variable Immunodeficiency Patient Fecal Microbiota Transplant Recapitulates Gut Dysbiosis. Res Sq. 2023;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
28.  Khoury ZH, Vila T, Puthran TR, Sultan AS, Montelongo-Jauregui D, Melo MAS, Jabra-Rizk MA. The Role of Candida albicans Secreted Polysaccharides in Augmenting Streptococcus mutans Adherence and Mixed Biofilm Formation: In vitro and in vivo Studies. Front Microbiol. 2020;11:307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 52]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]