Open access peer-reviewed chapter

Mesenchymal Stem/Stromal Cells in Allergic Disease Management

Written By

Leisheng Zhang, Zhongchao Han and Xiaowei Gao

Submitted: 24 May 2022 Reviewed: 08 June 2022 Published: 28 June 2022

DOI: 10.5772/intechopen.105763

From the Edited Volume

Allergic Disease - New Developments in Diagnosis and Therapy

Edited by Öner Özdemir

Chapter metrics overview

131 Chapter Downloads

View Full Metrics

Abstract

Allergic diseases are a clump of disorders caused by protective or harmful immune responses to specific exogenous stimulations. To date, the worldwide prevalence of allergic diseases has caused considerable perplex to patients and guardians physically and mentally. Despite the significant advances in preclinical investigation and clinical practice, yet the effective treatment strategies for allergic diseases are far from satisfaction. State-of-the-art renewal has highlighted the involvement of mesenchymal stem/stromal cell (MSC)-based cytotherapy for various allergic disease management including atopic dermatitis, pediatric asthma, allergic rhinitis, and urticaria, which largely attributes to the unique immunomodulatory properties and mode of action via autocrine and paracrine, direct- or trans-differentiation. In this chapter, we mainly focus on the latest updates of MSC-based investigations upon allergic disease administration as well as the concomitant prospective and challenges, which will provide overwhelming new references for MSC-based cytotherapy in regenerative medicine.

Keywords

  • mesenchymal stem/stromal cells
  • allergic diseases
  • exosome
  • immunomodulation
  • allergic rhinitis
  • cytotherapy

1. Introduction

Allergic diseases, including atopic dermatitis, pediatric asthma, allergic rhinitis, and urticaria, have been recognized as one of the most prevalent chronic diseases and affected more than 300 million individuals all over the world and thus, have garnered public health attention worldwide over the past decades [1]. To date, a variety of factors (e.g., IL-6, IL-8, IL-25, IL-33, INF-γ) and noxious stimuli (e.g., microbiota, helminths, human milk immunological composition) in the environment have been involved in the occurrence or progression of allergic diseases [2, 3]. Of note, immunomodulation has been acknowledged as the core strategy for allergic and autoimmune diseases.

Despite the diversity in the pathogenic mechanism of governing the progression, a variety of key elements involved in allergic diseases have been identified such as immune cells (e.g., mast cells, T cells), antibodies, cytokines, epigenetic and genetic determinants [1, 4, 5]. For instance, of the aforementioned pathogenic factors, mast cells with inflammatory mediator expression have been recognized playing a key role in various allergic reactions and autoimmune processes [6, 7].

For decades, integrated prevention and intervention strategies have been developed for the remission of allergic diseases. For example, the European Academy of Allergy and Clinical Immunology (EAACI) guidelines for allergen immunotherapy (AIT) have been reported in preparations for the administration of allergic disease by Dhami and colleagues [8]. Meanwhile, JAK/STAT inhibitors, together with relevant small-molecule cytokine antagonists such as CRTH2 inhibitors and PDE4 inhibitors, have been tested in a spectrum of allergic diseases [9]. Additionally, current advances have also suggested the probiotics and prebiotics in the treatment or prevention of allergic diseases during the prenatal period [10, 11, 12]. For example, Tang et al. reviewed that prebiotic-supplemented formulas might be an effective alternative for preventing atopic eczema in infants with high probability of developing allergic disease [10, 13].

Advertisement

2. The overview of MSCs

Mesenchymal stem/stromal cells (MSCs) are cell populations with unique hematopoietic-supporting and immunoregulatory properties, which are currently recognized as the uppermost counterparts for regenerative medicine in the field [14]. Since the first isolation from bone marrow, MSCs with various origins have been identified including adult tissues and perinatal tissues such as adipose-tissue-derived MSCs (AD-MSCs), dental-pulp-derived MSCs (DPSCs) [15], fetal-liver-derived MSCs (FL-MSCs), amniotic-membrane-derived MSCs (AMSCs), amniotic-fluid-derived MSCs (AF-MSCs), umbilical-cord-derived MSCs (UC-MSCs) [16, 17], placenta-derived MSCs (P-MSCs) [18], supernumerary teeth-derived apical papillary stem cells (SCAP-Ss) [15]. Meanwhile, current progress also highlighted the large-scale generation of MSCs from human pluripotent stem cells (hPSCs) including human embryonic stem cells (hESCs) and human induced PSCs (hiPSCs) as well [19, 20, 21].

2.1 Biofunctions of MSCs

As mentioned above, MSCs are heterogeneous populations with advantaged properties, which thus have been largely recognized as the dominating stromal cells in the hematopoietic microenvironment and the splendid “seed” cells for cellular therapy [22]. Not until the year of 2006, the International Society for Cellular Therapy (ISCT) released minimal guidelines for MSC definition including the fibroblast-like plastic-adherent cells, high percentage of subsets with mesenchymal-associated biomarker expression (CD73, CD90, CD105), whereas minimal expression of hematopoietic-associated (CD31, CD34) or immune-related (HLA-DR) surface markers and multi-lineage differentiation potential toward adipocytes, osteoblasts, and chondrocytes [23]. Of the biofunctions, immunomodulation is of great importance for the translational purposes of MSCs and the derivatives in tissue engineering and regenerative medicine via simultaneously inhibiting and stimulating the immune system and secreting immunosuppressors [24].

To date, MSCs have been extensively explored in multiple intractable and recurrent diseases such as acute-on-chronic liver failure (ACLF) [25], acute myocardial infarction (AMI) [26], acute myelogenous leukemia (AML) [27], refractory wounds [28], atopic dermatitis (AD), Crohn’s disease (CD) [18], graft-versus-host disease (GvHD) [16], coronavirus disease 2019 (COVID-19)-associated acute lung injury and acute respiratory distress syndrome (ALI/ARDS) [29, 30].

2.2 Regulatory mechanisms of MSCs

Generally, MSCs function mainly via serving as constructive microenvironment for hematogenesis, secretion (autocrine, paracrine), immunomodulation, and differentiation [31, 32, 33, 34]. For instance, the orchestration of multiple pathways (e.g., TGF-β, PPAR-γ2, and the Smad3-SOX9-CREB/p300 axis) in MSCs is critical for in vitro differentiation toward the mesodermal lineages [35, 36]. Instead, López-García and Castro-Manrreza verified the TNF-α and IFN-γ in mediating the immunoregulatory capacity of MSCs in the modulation of the immune response [37]. Interestingly, Montesinos and colleagues verified the regulatory effect of TNF-α and IFN-γ for the enhanced expression of ICAM-1 and microvesicle release of BM-MSCs when exposed to an inflammatory environment [38]. As to bone-marrow-derived MSCs (BM-MSCs), Zhang et al. demonstrated the hyperactivation of JAK–STAT signaling in AML patients compared with those in healthy donors [27].

Advertisement

3. MSCs for allergic disease management

As an intractable autoimmune disease with complex pathogenesis, allergic diseases have caused heavy economic and psychological burden to the patients and their families, and in particular, those with relapse and resistance against drugs. For the purpose, autogenous and allogeneic MSCs with unique bidirectional immunomodulatory property have caught the attention of pioneering investigators in the field. To date, MSCs have been involved in various subtypes of allergic disease management with considerable efficacy such as allergic rhinitis, allergic dermatitis, allergic asthma, and urticaria.

3.1 MSCs for allergic rhinitis management

Allergic rhinitis (AR), a well-described disease entity with extra-nasal manifestations, is considered as a major and increasing chronic inflammatory disease in the respiratory tract [39, 40, 41]. The pathogenesis of AR is associated with inflammatory mediators (e.g., IgE) and sensitized mast cells in the submucosa of the upper aerodigestive tract, which is also involved in various upper airway diseases including otitis media, chronic laryngitis, oral allergy syndrome, and obstructive sleep apnea [40, 42, 43, 44]. Clinically, although with certain disadvantages such as repeated attacks and adverse reaction, a series of desensitizing drugs including nasal glucocorticoids and antihistamines, together with acupuncture, are currently in use for allergic rhinitis treatment [39, 45, 46, 47].

Recently, Zheng et al. investigated the outcomes of 70 patients with allergic rhinitis with the administration of azelastine hydrochloride and montelukast sodium and found that clinical symptom score (e.g., nasal itching, runny nose, and nasal congestion) and serum levels of proinflammatory factors (e.g., hsCRP, IL-6, and IL-8) revealed preferable improvement compared with those 67 patients with azelastine hydrochloride alone [48]. Simultaneously, Xiong et al. recently reported the ameliorative effect of Chinese herbs (e.g., Guominjian) upon AR by utilizing the anti-inflammatory, anti-allergic, and immunomodulatory effects [49]. However, the spectrum of AR and the complex immunopathology further affect the efficacy of antiallergic drugs including antihistamines and mast cell stabilizers and thus, limit the treatment with the concomitant corticosteroid. Moreover, the recurrence of AR has been considered difficult to handle by current drug therapy.

Of note, pioneering clinicians have turned to MSC-based remedy for further improvement in the management of AR based on the immunomodulatory properties. For example, two interventional studies (NCT05167552, NCT05151133) have been registered according to the Clinicaltrials.gov website, and a total number of 78 participants are being enrolled for further treatment with various doses (low dose, 0.5 × 106 cells/kg; moderate dose, 1.0 × 106 cells/kg; high dose, 2.0 × 106 cells/kg) of hUC-MSC infusion (Table 1).

ConditionsNCT no.StatusPhasesEnrollLocation
Allergic rhinitisNCT05167552Not yet recruitingP1, P260
NCT05151133RecruitingP118China
Allergic dermatitisNCT02888704CompletedP113Korea
NCT03252340Active, not recruiting11Korea
NCT04179760RecruitingP1, P292Korea
NCT04137562RecruitingP2118Korea
UrticariaNCT02824393CompletedEarly P110Turkey

Table 1.

MSC-based clinical trials upon atopic diseases.

3.2 MSCs for atopic dermatitis management

Atopic dermatitis (AD), also regarded as atopic eczema, is a relapsing inflammatory skin condition and a chronic heterogeneous skin lesion worldwide among childhood, infancy, and even adulthood, and in particular, among those families with a history of allergic diseases [50, 51, 52]. To date, a variety of pathogenic factors associated with the environmental, immunologic, and genetic elements have been identified for the intrinsic and extrinsic subtypes of AD such as food allergies, respiratory diseases, autoimmune disorders, and inflammatory skin infections [50, 53]. For example, the well-established ingredients including dysbiosis of skin microbiota, epidermal barrier disruption, overactivation of the helper T cell subsets (e.g., Th1, Th2, Th17, Th22), together with increased immunoglobulin E (IgE) and eosinophils in blood, have been demonstrated in association with the pathogenesis of AD [54, 55]. Of the disease progression, the impaired skin barrier is considered as the initial step during the development of AD, which is adequate to cause further allergic sensitization and skin inflammation [56]. Simultaneously, AD is deemed to the initiation phase of relevant atopic disorders such as food allergy and allergic asthma and rhinitis, which is continuous for ages and maintains the relapsing-remitting status in numerous patients [57].

Therewith, despite the current pharmacological and nonpharmacological treatment modalities in relieving misery in patients with moderate to severe AD, the efficacy or persistence is still unsatisfactory on account of the indeterminacy and complexity of the underlying pathogenesis [1, 52]. Strikingly, Kim et al. reported the safety and certain improvements of AD inpatients with an overall response rate of 55% at week 12 with a high dose of hUC-MSC (5 × 107) administration through local subcutaneous injection [58]. Similarly, our group further reported the real cure of an elderly patient rather than partial remission by conducting a single round of intravenous injection of hUC-MSCs without recrudesce during the 14-month’s follow-up. Overall, the aforementioned proof-of-concept studies ulteriorly highlighted the feasibility upon AD patients with refractory AD-associated symptoms.

3.3 MSCs for allergic asthma management

Allergic asthma, known as a “syndrome” with over 300 million individuals worldwide, which also has become a dominating burden in Westernized societies [59].

Generally, allergic asthma is caused by a complex interplay between environmental stimulus and genetic and factors [60, 61]. As a chronic airway inflammatory disease, patients with allergic asthma reveal multifaceted clinical manifestations such as intermittent attacks of airway hyper-reactivity, breathlessness, coughing, and wheezing. As to adult asthma, an initial exposure to allergen triggers Th2 cell-dependent immune response that regulates the production of IgE and cytokines in the lungs [60]. Distinguishing from the characteristics of ILC2s in chronic allergic diseases, IgE sensitization has been considered acting as a crucial role in the progression of allergic diseases [60, 62, 63]. Collectively, the environmental and genetic factors orchestrate the complexity and challenges of allergic asthma posed for the further development of novel remedies for effective treatment and prevention of allergic asthma.

State-of-the-art updates have suggested the therapeutic effect of MSCs or MSC-derived exosomes (MSC-Exo) in the management of allergic asthma in preclinical and clinical investigations [64, 65, 66]. For instance, Boldrini-Leite et al. took advantage of the ovalbumin-induced allergic asthma mice model for the remodeling of the inflammatory process and pulmonary symptoms and confirmed the potential of BM-MSCs to modulate lung inflammatory processes and tissue repair. Recently, Huang et al. found that the mitochondrial dysfunction and asthma pathophysiology in the asthma animal model were efficiently rescued by MSC injection, and the levels of relevant gene expression were reversed as well such as interleukins (e.g., IL-4, IL-5, IL-13, IL-25, IL-33) and mitochondria genes (e.g., COX-1, COX-2, Cytb, ND-1) and inflammatory factors (e.g., INF-γ) [65]. Similarly, de Castro et al. demonstrated the efficacy of human adipose tissue–derived MSCs (hAD-MSCs) and the extracellular vesicles upon experimental allergic asthma by airway remodeling. In detail, C57BL/6 female mice with experimental allergic asthma manifested reduced eosinophils in lung tissue, collagen fiber content in lung parenchyma and airways, levels of Tgf-β in lung tissue, and CD3+CD4+ T lymphocyte counts in the thymus [67]. Interestingly, Abreu and colleagues verified the enhanced therapeutic effect of MSCs upon allergic asthma by pretreatment with eicosapentaenoic acid (EPA) [68]. Moreover, serum from asthmatic mice has been proved with potentiated efficacy of MSCs in experimental allergic asthma [69]. Taken together, MSCs of different origins alone or in combination with relevant remedies reveal rosy prospective in allergic asthma management.

3.4 MSCs for urticaria management

Urticaria, including the immunological and nonimmunological subtypes, is a series of common skin disorder occurring in 0.5–5% of the general population that affects individuals of all ages and results from many different stimuli, which compromise quality of life and affect individual performance physically and mentally [70, 71, 72, 73]. Generally, urticaria acts as a hypersensitivity reaction with mast cell activation due to the stimulation of T lymphocytes and/or antibodies. Instead, nonimmunological urticarias with mast cell activation are involved in immunomodulation (e.g., Toll-like, complement, proinflammatory factors) or toxicity of xenobiotics (e.g., haptens, drugs). Therewith, the variations in the pathophysiological mechanisms further result in the great heterogeneity of clinical symptoms and the variable remedies [72, 74].

Urticaria exhibits multifaceted clinical manifestations such as intensely pruritic wheals, edema of the interstitial or subcutaneous tissue. In details, distinguishing from the acute urticaria, chronic urticaria, including chronic spontaneous urticaria (CSU) and chronic inducible urticaria (e.g., cold urticaria), is regarded as a difficult-to-treat skin disease and results in the major impact on quality of life in patients according to the European guideline on the management of urticaria, which describes a multidisciplinary approach for urticaria administration [75, 76, 77].

Being obscure in fully elucidating the underlying etiopathogenesis as well as the limitation in urticaria management, pioneering scientists and clinicians turned to MSC-based cytotherapy for developing more efficient treatment options [73]. Of note, Özgül Özdemir and colleagues employed autologous AD-MSCs for the administration of 10 refractory CSU patients and noticed the immunomodulatory effect upon CD4+ T cell subsets and cytokine expression profiling. For instance, the Th2 subset and pro-inflammatory factors (e.g., TGF-β1, IDO, PGE2, anti-FcεRI) revealed a significant decrease in urticaria patients with MSC injection after 2 weeks [73]. Collectively, despite the minimal literatures in the field, the findings suggested that MSCs might be an alternative and effective strategy for treatment-resistant CSU patients in clinical practice [73].

Advertisement

4. Clinical trials of MSC-based remedy for allergic diseases

In the recent years, MSC-based cytotherapy has attracted the attention of a certain number of biologists and clinicians in the field for allergic disease management. According to the Clinicaltrials.gov website of National Institutes of Health (NIH), a total number of seven clinical trials have been registered worldwide (up to May 24th, 2022) including four trials in Korea (NCT02888704, NCT03252340, NCT04179760, NCT04137562), one trial in China (NCT05151133), one trial in Turkey (NCT02824393), and one trial unknowable (NCT05167552) (Table 1).

The interventional studies conducted by clinical investigators are designed to explore the safety and effectiveness of MSC-based treatment for relevant disease treatment including two trials for allergic rhinitis, four trials for allergic dermatitis, and one trial for urticaria (Table 1). Of the aforementioned clinical trials, two were not yet recruiting, three were recruiting, four were completed, and two were completed (Table 1). Meanwhile, we further noticed that all of the registered clinical trials were in the Phase 1 and Phase 2 stages (Table 1).

Advertisement

5. Conclusions

MSCs and the concomitant derivatives have emerged as advantaged and alternative sources for allergic disease management. MSC- or MSC-exo/small secretory vesicles (sEVs)-based cytotherapy has supplied overwhelming new tissue engineering platforms to sequentially ameliorate disease manifestations and improve the clinical outcomes of patients with relevant allergic diseases. However, the lack of standardized methodology and evaluation criteria (e.g., safety, effectiveness, biodistribution) in the preparation of good manufacturing practices (GMP)-grade MSCs for clinical purposes hinders the development of MSC-based tissue engineering and regenerative medicine. Therefore, further understanding of the aforementioned aspects of MSCs will benefit clinical applications and the industrialization of MSC-based cytotherapy in future.

Advertisement

Acknowledgments

The authors would like to thank the members in Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province & NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, and Institute of Biology & Hefei Institute of Physical Science, Chinese Academy of Sciences for their kind suggestions. This study was supported by grants from Science and technology projects of Guizhou Province (QKH-J-ZK[2021]-107), Natural Science Foundation of Jiangxi Province (20212BAB216073), the project Youth Fund funded by Shandong Provincial Natural Science Foundation (ZR2020QC097), the Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (2019PT320005), The 2021 Central-Guided Local Science and Technology Development Fund (ZYYDDFFZZJ-1), Gansu Key Laboratory of molecular diagnosis and precision treatment of surgical tumors (18JR2RA033), Key project funded by Department of Science and Technology of Shangrao City (2020AB002, 2020 K003, 2021F013), Jiangxi Provincial Key New Product Incubation Program Funded by Technical Innovation Guidance Program of Shangrao (2020G002), Natural Science Foundation of Gansu Province (21JR11RA186, 20JR10RA415), Key talent project of Gansu Province of the Organization Department of Gansu provincial Party committee (2020RCXM076), Fujian Provincial Ministerial Finance Special Project (2021XH018).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Notes/thanks/other declarations

Not applicable.

Advertisement

Appendices and nomenclature

MSCs

mesenchymal stem/stromal cells

EAACI

the European Academy of Allergy and Clinical Immunology

AIT

allergen immunotherapy

AR

allergic rhinitis

AD

atopic dermatitis

IgE

immunoglobulin E

hAD-MSCs

human adipose tissue–derived MSCs

EPA

eicosapentaenoic acid

CSU

chronic spontaneous urticaria

UC-MSCs

Umbilical-cord-derived MSCs

P-MSCs

placenta-derived MSCs

AD-MSCs

adipose-tissue-derived MSCs

DPSCs

dental-pulp-derived MSCs

FL-MSCs

fetal-liver-derived MSCs

AMSCs

amniotic-membrane-derived MSCs

AF-MSCs

amniotic-fluid-derived MSCs

UC-MSCs

umbilical-cord-derived MSCs

P-MSCs

placenta-derived MSCs

hPSCs

human pluripotent stem cells

SCAP-Ss

supernumerary teeth-derived apical papillary stem cells

ISCT

the International Society for Cellular Therapy

hESCs

human embryonic stem cells

hiPSCs

human induced PSCs

MSC-exo

MSC-derived exosomes

sEVs

small secretory vesicles

GMP

good manufacturing practices

AML

acute myelogenous leukemia

CD

Crohn’s disease

GvHD

graft-versus-host disease

COVID-19

coronavirus disease 2019

ALI

acute lung injury

ARDS

acute respiratory distress syndrome

AMI

acute myocardial infarction

ACLF

acute-on-chronic liver failure

References

  1. 1. DeVries A, Vercelli D. Epigenetics in allergic diseases. Current Opinion in Pediatrics. 2015;27(6):719-723
  2. 2. Huang Y, Wang G, Wang C. Dietary intervention for allergic disease. Current Pharmaceutical Design. 2014;20(6):988-995
  3. 3. Nakao A. Circadian regulation of the biology of allergic disease: Clock disruption can promote allergy. Frontiers in Immunology. 2020;11:1237
  4. 4. Rottem M, Gershwin ME, Shoenfeld Y. Allergic disease and autoimmune effectors pathways. Developmental Immunology. 2002;9(3):161-167
  5. 5. Agache I, Cojanu C, Laculiceanu A, Rogozea L. Genetics and epigenetics of allergy. Current Opinion in Allergy and Clinical Immunology. 2020;20(3):223-232
  6. 6. Costela-Ruiz VJ, Illescas-Montes R, Pavon-Martinez R, Ruiz C, Melguizo-Rodriguez L. Role of mast cells in autoimmunity. Life Sciences. 2018;209:52-56
  7. 7. Robbie-Ryan M, Brown M. The role of mast cells in allergy and autoimmunity. Current Opinion in Immunology. 2002;14(6):728-733
  8. 8. Dhami S, Nurmatov U, Halken S, Calderon MA, Muraro A, Roberts G, et al. Allergen immunotherapy for the prevention of allergic disease: Protocol for a systematic review. Pediatric Allergy and Immunology. 2016;27(3):236-241
  9. 9. Howell MD, Fitzsimons C, Smith PA. JAK/STAT inhibitors and other small molecule cytokine antagonists for the treatment of allergic disease. Annals of Allergy, Asthma & Immunology. 2018;120(4):367-375
  10. 10. Tang ML, Lahtinen SJ, Boyle RJ. Probiotics and prebiotics: Clinical effects in allergic disease. Current Opinion in Pediatrics. 2010;22(5):626-634
  11. 11. Kukkonen K, Savilahti E, Haahtela T, Juntunen-Backman K, Korpela R, Poussa T, et al. Probiotics and prebiotic galacto-oligosaccharides in the prevention of allergic diseases: A randomized, double-blind, placebo-controlled trial. The Journal of Allergy and Clinical Immunology. 2007;119(1):192-198
  12. 12. Lopez-Santamarina A, Gonzalez EG, Lamas A, Mondragon ADC, Regal P, Miranda JM. Probiotics as a possible strategy for the prevention and treatment of allergies. A narrative review. Foods. 2021;10(4):701
  13. 13. Ismail IH, Licciardi PV, Tang ML. Probiotic effects in allergic disease. Journal of Paediatrics and Child Health. 2013;49(9):709-715
  14. 14. Kulus M, Sibiak R, Stefańska K, Zdun M, Wieczorkiewicz M, Piotrowska-Kempisty H, et al. Mesenchymal stem/stromal cells derived from human and animal perinatal tissues-origins, characteristics, signaling pathways, and clinical trials. Cells. 2021;10(12):3278
  15. 15. Yao J, Chen N, Wang X, Zhang L, Huo J, Chi Y, et al. Human supernumerary teeth-derived apical papillary stem cells possess preferable characteristics and efficacy on hepatic fibrosis in mice. Stem Cells International. 2020;2020:6489396
  16. 16. Zhao Q , Zhang L, Wei Y, Yu H, Zou L, Huo J, et al. Systematic comparison of hUC-MSCs at various passages reveals the variations of signatures and therapeutic effect on acute graft-versus-host disease. Stem Cell Research & Therapy. 2019;10(1):354
  17. 17. Zhang Y, Li Y, Li W, Cai J, Yue M, Jiang L, et al. Therapeutic effect of human umbilical cord mesenchymal stem cells at various passages on acute liver failure in rats. Stem Cells International. 2018;2018:7159465
  18. 18. Hou H, Zhang L, Duan L, Liu Y, Han Z, Li Z, et al. Spatio-temporal metabolokinetics and efficacy of human placenta-derived mesenchymal stem/stromal cells on mice with refractory Crohn’s-like Enterocutaneous Fistula. Stem Cell Reviews and Reports. 2020;16(6):1292-1304
  19. 19. Zhang L, Wei Y, Chi Y, Liu D, Yang S, Han Z, et al. Two-step generation of mesenchymal stem/stromal cells from human pluripotent stem cells with reinforced efficacy upon osteoarthritis rabbits by HA hydrogel. Cell & Bioscience. 2021;11(1):6
  20. 20. Wei Y, Hou H, Zhang L, Zhao N, Li C, Huo J, et al. JNKi- and DAC-programmed mesenchymal stem/stromal cells from hESCs facilitate hematopoiesis and alleviate hind limb ischemia. Stem Cell Research & Therapy. 2019;10(1):186
  21. 21. Zhang L, Wang H, Liu C, Wu Q , Su P, Wu D, et al. MSX2 initiates and accelerates mesenchymal stem/stromal cell specification of hPSCs by regulating TWIST1 and PRAME. Stem Cell Reports. 2018;11(2):497-513
  22. 22. Barilani M, Banfi F, Sironi S, Ragni E, Guillaumin S, Polveraccio F, et al. Low-affinity nerve growth factor receptor (CD271) heterogeneous expression in adult and fetal mesenchymal stromal cells. Scientific Reports. 2018;8(1):9321
  23. 23. Horwitz EM, Le Blanc K, Dominici M, Mueller I, Slaper-Cortenbach I, Marini FC, et al. International Society for Cellular T: Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement. Cytotherapy. 2005;7(5):393-395
  24. 24. Jiang W, Xu J. Immune modulation by mesenchymal stem cells. Cell Proliferation. 2020;53(1):e12712
  25. 25. Lin BL, Chen JF, Qiu WH, Wang KW, Xie DY, Chen XY, et al. Allogeneic bone marrow-derived mesenchymal stromal cells for hepatitis B virus-related acute-on-chronic liver failure: A randomized controlled trial. Hepatology. 2017;66(1):209-219
  26. 26. Lim M, Wang W, Liang L, Han ZB, Li Z, Geng J, et al. Intravenous injection of allogeneic umbilical cord-derived multipotent mesenchymal stromal cells reduces the infarct area and ameliorates cardiac function in a porcine model of acute myocardial infarction. Stem Cell Research & Therapy. 2018;9(1):129
  27. 27. Zhang L, Chi Y, Wei Y, Zhang W, Wang F, Zhang L, et al. Bone marrow-derived mesenchymal stem/stromal cells in patients with acute myeloid leukemia reveal transcriptome alterations and deficiency in cellular vitality. Stem Cell Research & Therapy. 2021;12(1):365
  28. 28. Hocking AM. Mesenchymal stem cell therapy for cutaneous wounds. Advanced Wound Care (New Rochelle). 2012;1(4):166-171
  29. 29. Zhang LS, Yu Y, Yu H, Han ZC. Therapeutic prospects of mesenchymal stem/stromal cells in COVID-19 associated pulmonary diseases: From bench to bedside. World Journal of Stem Cells. 2021;13(8):1058-1071
  30. 30. Aitong W, Leisheng Z, Hao Y. Visualized analyses of investigations upon mesenchymal stem/stromal cell-based cytotherapy and underlying mechanisms for COVID-19 associated ARDS. Current Stem Cell Research & Therapy. 2022;17(1):2-12
  31. 31. Wang X, Lazorchak AS, Song L, Li E, Zhang Z, Jiang B, et al. Immune modulatory mesenchymal stem cells derived from human embryonic stem cells through a trophoblast-like stage. Stem Cells. 2016;34(2):380-391
  32. 32. Wei Y, Zhang L, Chi Y, Ren X, Gao Y, Song B, et al. High-efficient generation of VCAM-1(+) mesenchymal stem cells with multidimensional superiorities in signatures and efficacy on aplastic anaemia mice. Cell Proliferation. 2020;53(8):e12862
  33. 33. Wu Y, Wang Z, Cao Y, Xu L, Li X, Liu P, et al. Cotransplantation of haploidentical hematopoietic and umbilical cord mesenchymal stem cells with a myeloablative regimen for refractory/relapsed hematologic malignancy. Annals of Hematology. 2013;92(12):1675-1684
  34. 34. Zhao M, Liu S, Wang C, Wang Y, Wan M, Liu F, et al. Mesenchymal stem cell-derived extracellular vesicles attenuate mitochondrial damage and inflammation by stabilizing mitochondrial DNA. ACS Nano. 2021;15(1):1519-1538
  35. 35. Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid-activated transcription factor. Cell. 1994;79(7):1147-1156
  36. 36. Furumatsu T, Tsuda M, Taniguchi N, Tajima Y, Asahara H. Smad3 induces chondrogenesis through the activation of SOX9 via CREB-binding protein/p300 recruitment. The Journal of Biological Chemistry. 2005;280(9):8343-8350
  37. 37. Lopez-Garcia L, Castro-Manrreza ME. TNF-alpha and IFN-gamma participate in improving the immunoregulatory capacity of mesenchymal stem/stromal cells: Importance of cell-cell contact and extracellular vesicles. International Journal of Molecular Sciences. 2021;22(17)
  38. 38. Montesinos JJ, Lopez-Garcia L, Cortes-Morales VA, Arriaga-Pizano L, Valle-Rios R, Fajardo-Orduna GR, et al. Human bone marrow mesenchymal stem/stromal cells exposed to an inflammatory environment increase the expression of ICAM-1 and release microvesicles enriched in this adhesive molecule: Analysis of the Participation of TNF-alpha and IFN-gamma. Journal of Immunology Research. 2020;2020:8839625
  39. 39. Bao H, Si D, Gao L, Sun H, Shi Q , Yan Y, et al. Acupuncture for the treatment of allergic rhinitis: A systematic review protocol. Medicine (Baltimore). 2018;97(51):e13772
  40. 40. Mur T, Brook C, Platt M. Extranasal manifestations of allergy in the head and neck. Current Allergy and Asthma Reports. 2020;20(7):21
  41. 41. Cassano M, Maselli A, Mora F, Cassano P. Rhinobronchial syndrome: Pathogenesis and correlation with allergic rhinitis in children. International Journal of Pediatric Otorhinolaryngology. 2008;72(7):1053-1058
  42. 42. Martines F, Salvago P, Ferrara S, Messina G, Mucia M, Plescia F, et al. Factors influencing the development of otitis media among Sicilian children affected by upper respiratory tract infections. Brazilian Journal of Otorhinolaryngology. 2016;82(2):215-222
  43. 43. Fireman P. Otitis media and eustachian tube dysfunction: Connection to allergic rhinitis. The Journal of Allergy and Clinical Immunology. 1997;99(2):S787-S797
  44. 44. Ciprandi G, Tosca MA. Turbinate hypertrophy, allergic rhinitis, and otitis media. Current Allergy and Asthma Reports. 2021;21(9):44
  45. 45. Li J, Liu L, Jiao L, Liao K, Xu L, Zhou X, et al. Clinical acupuncture therapy for children with allergic rhinitis: A protocol for systematic review and meta analysis. Medicine (Baltimore). 2021;100(3):e24086
  46. 46. Feng S, Han M, Fan Y, Yang G, Liao Z, Liao W, et al. Acupuncture for the treatment of allergic rhinitis: A systematic review and meta-analysis. American Journal of Rhinology & Allergy. 2015;29(1):57-62
  47. 47. Bai H, Xu S, Wu Q , Xu S, Sun K, Wu J, et al. Clinical events associated with acupuncture intervention for the treatment of chronic inflammation associated disorders. Mediators of Inflammation. 2020;2020:2675785
  48. 48. Zheng Q , Ma D, Zhu Q , Tang S, Chen C. Effect of azelastine hydrochloride combined with montelukast sodium in the treatment of patients with allergic rhinitis. American Journal of Translational Research. 2021;13(8):9570-9577
  49. 49. Xiong Y, Li H, Zhang SN. Guominjian for allergic rhinitis: A protocol for systematic review and meta-analysis of randomized clinical trials. Medicine (Baltimore). 2020;99(44):e22854
  50. 50. Torres T, Ferreira EO, Goncalo M, Mendes-Bastos P, Selores M, Filipe P. Update on atopic dermatitis. Acta Médica Portuguesa. 2019;32(9):606-613
  51. 51. Cabanillas B, Brehler AC, Novak N. Atopic dermatitis phenotypes and the need for personalized medicine. Current Opinion in Allergy and Clinical Immunology. 2017;17(4):309-315
  52. 52. Stander S. Atopic dermatitis. The New England Journal of Medicine. 2021;384(12):1136-1143
  53. 53. Leung DY, Nicklas RA, Li JT, Bernstein IL, Blessing-Moore J, Boguniewicz M, et al. Disease management of atopic dermatitis: An updated practice parameter. Joint Task Force on Practice Parameters. Annals of Allergy, Asthma & Immunology. 2004;93(3 Suppl 2):S1-S21
  54. 54. Guttman-Yassky E, Nograles KE, Krueger JG. Contrasting pathogenesis of atopic dermatitis and psoriasis—Part II: Immune cell subsets and therapeutic concepts. The Journal of Allergy and Clinical Immunology. 2011;127(6):1420-1432
  55. 55. Suarez-Farinas M, Dhingra N, Gittler J, Shemer A, Cardinale I, de Guzman SC, et al. Intrinsic atopic dermatitis shows similar TH2 and higher TH17 immune activation compared with extrinsic atopic dermatitis. The Journal of Allergy and Clinical Immunology. 2013;132(2):361-370
  56. 56. Kim J, Kim BE, Leung DYM. Pathophysiology of atopic dermatitis: Clinical implications. Allergy and Asthma Proceedings. 2019;40(2):84-92
  57. 57. Schneider L, Tilles S, Lio P, Boguniewicz M, Beck L, LeBovidge J, et al. Atopic dermatitis: A practice parameter update 2012. The Journal of Allergy and Clinical Immunology. 2013;131(2):295-299
  58. 58. Kim HS, Yun JW, Shin TH, Lee SH, Lee BC, Yu KR, et al. Human umbilical cord blood mesenchymal stem cell-derived PGE2 and TGF-beta1 alleviate atopic dermatitis by reducing mast cell degranulation. Stem Cells. 2015;33(4):1254-1266
  59. 59. Eigenmann PA. Diagnosis of allergy syndromes: Do symptoms always mean allergy? Allergy. 2005;60(Suppl 79):6-9
  60. 60. Mukherjee AB, Zhang Z. Allergic asthma: Influence of genetic and environmental factors. The Journal of Biological Chemistry. 2011;286(38):32883-32889
  61. 61. Gaber T, Chen Y, Krauss PL, Buttgereit F. Metabolism of T lymphocytes in health and disease. International Review of Cell and Molecular Biology. 2019;342:95-148
  62. 62. Leffler J, Stumbles PA, Strickland DH. Immunological processes driving IgE sensitisation and disease development in males and females. International Journal of Molecular Sciences. 2018;19(6)
  63. 63. Doherty TA. At the bench: Understanding group 2 innate lymphoid cells in disease. Journal of Leukocyte Biology. 2015;97(3):455-467
  64. 64. Ren J, Liu Y, Yao Y, Feng L, Zhao X, Li Z, et al. Intranasal delivery of MSC-derived exosomes attenuates allergic asthma via expanding IL-10 producing lung interstitial macrophages in mice. International Immunopharmacology. 2021;91:107288
  65. 65. Huang M, Mehrabi Nasab E, Athari SS. Immunoregulatory effect of mesenchymal stem cell via mitochondria signaling pathways in allergic asthma. Saudi Journal of Biological Science. 2021;28(12):6957-6962
  66. 66. Abreu SC, Antunes MA, de Castro JC, de Oliveira MV, Bandeira E, Ornellas DS, et al. Bone marrow-derived mononuclear cells vs. mesenchymal stromal cells in experimental allergic asthma. Respiratory Physiology & Neurobiology. 2013;187(2):190-198
  67. 67. de Castro LL, Xisto DG, Kitoko JZ, Cruz FF, Olsen PC, Redondo PAG, et al. Human adipose tissue mesenchymal stromal cells and their extracellular vesicles act differentially on lung mechanics and inflammation in experimental allergic asthma. Stem Cell Research & Therapy. 2017;8(1):151
  68. 68. Abreu SC, Lopes-Pacheco M, da Silva AL, Xisto DG, de Oliveira TB, Kitoko JZ, et al. Eicosapentaenoic acid enhances the effects of mesenchymal stromal cell therapy in experimental allergic asthma. Frontiers in Immunology. 2018;9:1147
  69. 69. Abreu SC, Xisto DG, de Oliveira TB, Blanco NG, de Castro LL, Kitoko JZ, et al. Serum from asthmatic mice potentiates the therapeutic effects of mesenchymal stromal cells in experimental allergic asthma. Stem Cells Translational Medicine. 2019;8(3):301-312
  70. 70. Kulthanan K, Tuchinda P, Chularojanamontri L, Chanyachailert P, Korkij W, Chunharas A, et al. Clinical practice guideline for diagnosis and management of urticaria. Asian Pacific Journal of Allergy and Immunology. 2016;34(3):190-200
  71. 71. Monroe EW, Jones HE. Urticaria. An updated review. Archives in Dermatology. 1977;113(1):80-90
  72. 72. Hennino A, Berard F, Guillot I, Saad N, Rozieres A, Nicolas JF. Pathophysiology of urticaria. Clinical Reviews in Allergy and Immunology. 2006;30(1):3-11
  73. 73. Ozgul Ozdemir RB, Ozdemir AT, Kirmaz C, Ovali E, Olmez E, Kerem H, et al. Mesenchymal stem cells: A potential treatment approach for refractory chronic spontaneous urticaria. Stem Cell Reviews and Reports. 2021;17(3):911-922
  74. 74. Saini SS. Chronic spontaneous urticaria: Etiology and pathogenesis. Immunology and Allergy Clinics of North America. 2014;34(1):33-52
  75. 75. Sabroe RA. Acute urticaria. Immunology and Allergy Clinics of North America. 2014;34(1):11-21
  76. 76. Alcantara Villar M, Armario Hita JC, Cimbollek S, Fernandez Ballesteros MD, Galan Gutierrez M, Hernandez Montoya C, et al. A review of the latest recommendations on the management of chronic urticaria: A Multidisciplinary Consensus Statement from Andalusia, Spain. Actas Dermosifiliogr (Engl Ed). 2020;111(3):222-228
  77. 77. Maltseva N, Borzova E, Fomina D, Bizjak M, Terhorst-Molawi D, Kosnik M, et al. Cold urticaria—What we know and what we do not know. Allergy. 2021;76(4):1077-1094

Written By

Leisheng Zhang, Zhongchao Han and Xiaowei Gao

Submitted: 24 May 2022 Reviewed: 08 June 2022 Published: 28 June 2022