Next Article in Journal
Metabolic Changes Induced by Cerebral Ischemia, the Effect of Ischemic Preconditioning, and Hyperhomocysteinemia
Next Article in Special Issue
Sex Plays a Multifaceted Role in Asthma Pathogenesis
Previous Article in Journal
Osteoprotegerin Is a Better Predictor for Cardiovascular and All-Cause Mortality than Vascular Calcifications in a Multicenter Cohort of Patients on Peritoneal Dialysis
Previous Article in Special Issue
Sequential Biotherapy Targeting IL-5 and IL-4/13 in Patients with Eosinophilic Asthma with Sinusitis and Otitis Media
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential for Intractable Asthma by Targeting L-Type Amino Acid Transporter 1

1
Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Tochigi 321-0293, Japan
2
Department of Disease Model, Research Institute of Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
*
Author to whom correspondence should be addressed.
Biomolecules 2022, 12(4), 553; https://doi.org/10.3390/biom12040553
Submission received: 18 March 2022 / Revised: 6 April 2022 / Accepted: 6 April 2022 / Published: 8 April 2022

Abstract

:
Bronchial asthma is a chronic disease characterized by airway inflammation, obstruction, and hyperresponsiveness. CD4+ T cells, particularly T helper (Th) 2 cells, and their specific cytokines are important mediators in asthma pathogenesis. However, it has been established that Th subsets, other than Th2, as well as various cell types, including innate lymphoid cells (ILCs), significantly contribute to the development of allergic inflammation. These cells require facilitated amino acid uptake to ensure their full function upon activation. Emerging studies have suggested the potential of pharmacological inhibition of amino acid transporters to inhibit T cell activation and the application of this strategy for treating immunological and inflammatory disorders. In the present review, we explore the possibility of targeting L-type amino acid transporter (LAT) as a novel therapeutic approach for bronchial asthma, including its steroid-resistant endotypes.

1. Introduction

It is estimated that more than 300 million people suffer from bronchial asthma worldwide, and the number of patients continues growing [1]. Among the various implicated immunological and inflammatory cells, T helper (Th) 2 cells have been recognized as major players in asthma pathogenesis. The identification of CD4+ T cells expressing type 2 cytokines, such as interleukin (IL)-4, IL-5, and IL-13, in bronchial biopsy specimens and bronchial lavage fluids [2,3], as well as the recent clinical application of biomedicines against type 2 cytokines [4,5,6], further emphasizes the importance of Th2 cells in the development of asthma.
Glucocorticoids (steroids) are principally employed in all steps of asthma treatment. Consistent with the steroid-mediated suppression of Th2 cell cytokine production, bronchial asthma is now considered a controllable disease, given the efficacy and safety of inhaled corticosteroid (ICS)-based therapies. However, approximately 10% of patients with asthma fail to adequately respond to steroids and other existing therapeutics [7].
Targeting activated cells refractory to steroid treatment is a promising strategy for resolving issues that continue to hinder asthma therapy. Recently, we discovered the enhanced expression of L-type amino acid transporter (LAT) 1 in activated T cells [8,9]. LAT1 regulates the incorporation of various essential amino acids into cells, potentially contributing to the development of T cell-mediated allergic inflammation. Herein, we describe the possible management of bronchial asthma, including steroid-resistant endotypes by targeting LAT1, summarizing the recent establishment of asthma pathogenesis, including the contribution of LAT1, and efforts to generate a novel LAT1 inhibitor.

2. Classification of Asthma Endotypes

2.1. Type 2-High (T2) Asthma

Bronchial asthma can be classified into two major categories based on the key biological pathway: T2 and type 2-low (non-T2) [10,11,12]. Naïve CD4+ T cells differentiate into Th cell subpopulations such as Th1, Th2, Th9, and Th17 cells, exhibiting characteristic cytokine-producing capacities according to their surrounding environment. Among them, Th2 cells and type 2 cytokines are predominantly involved in mechanisms underlying T2 asthma. Type 2 cytokines, including IL-4, IL-5, and IL-13, drive IgE class switch, eosinophil recruitment into inflammatory sites, and mucus production [13]. In murine models of asthma, diminished airway inflammation was observed in several lines of eosinophil-deficient mice [14,15,16], indicating a considerable contribution of eosinophils to T2 asthma [17,18].
Consistent with the clinical effectiveness of an anti-IgE antibody [19,20], IgE-mediated mast cell activation is a crucial cascade that induces allergic reactions. However, in an allergen-induced airway inflammation model, substantial airway hyperresponsiveness, accompanied by eosinophil infiltration, was induced in mast cell-deficient mice [21]. CD4+ T cell depletion following administration of an anti-CD4 antibody suppressed eosinophilic inflammation. Furthermore, the adoptive transfer of in vitro-differentiated Th2 cells to normal mice could induce airway eosinophilic inflammation and hyperresponsiveness following allergen provocation [22]. Given that this procedure is free from allergen-specific IgE synthesis, it is strongly suggested that Th2 cells directly contribute to asthma pathogenesis, independent of the IgE/mast cell cascade.
Stimulation-induced type 2 cytokine production by Th2 cells is strongly suppressed by steroid application in vitro [23]. Accordingly, most patients with T2 asthma are likely to be steroid-sensitive; thus, decreased serum type 2 cytokine levels can be observed in parallel with symptomatic remission following steroid treatment [24].
In addition to Th2 cells, another cellular source of type 2 cytokines has been identified. Allergens, pollutants, and microorganisms stimulate bronchial epithelial cells to release IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) [25,26,27], which stimulate group 2 innate lymphoid cells (ILC2), and, in turn, release type 2 cytokines [28,29]. In contrast to Th2 cells, steroids do not potently suppress ILC2-mediated type 2 cytokine production [30]. These findings are consistent with the substantial efficacy of biomedicines against type 2 cytokines in patients with steroid-resistant asthma [31].

2.2. Non-T2 Asthma

It has been highlighted that Th cell subpopulations that preferentially produce non-type 2 cytokines can also contribute to the development of allergic inflammation without eosinophil accumulation. Th17 cells have been implicated in non-T2 asthma pathology via the production of IL-17A, IL-17E, and IL-22 [32]. IL-17A was shown to be upregulated in airway biopsy specimens and sputa from patients with moderate-to-severe asthma [33,34,35]. Peripheral blood mononuclear cells from patients with severe asthma reportedly exhibit an enhanced capacity to synthesize IL-17A and IL-22 following in vitro activation [36]. In addition, IL-17A stimulated human bronchial fibroblasts to produce pro-fibrotic cytokines [35] and directly increased airway smooth muscle contractility in humans and mice [37]. The adoptive transfer of Th17 cells into normal mice could induce airway hyperresponsiveness, along with neutrophil infiltration following allergen challenge [38]. Notably, steroid treatment failed to effectively suppress Th17-mediated responses [38].
Therefore, as a new strategy to treat steroid-resistant asthma, the potential use of antibodies against IL-17A and its receptor has been investigated. Administration of an anti-IL-17A antibody attenuated house dust mite-induced airway inflammation and hyperresponsiveness in mice [39]. Human anti-IL-17A antibodies (secukinumab and ixekizumab) are currently approved to treat inflammatory diseases other than asthma, such as plaque psoriasis. Notably, brodalumab, a monoclonal antibody against human IL-17 receptor approved for moderate-to-severe plaque psoriasis, failed to demonstrate the potent efficacy in patients with moderate-to-severe asthma in the first clinical trial [40]; however, further investigations with adequate recruitment of Th17 cell-dependent endotype patients should be considered.
In addition, the involvement of immunological and inflammatory cells other than Th17 cells has been suggested in the development of non-T2 and steroid-resistant asthma. Along with Th2 and Th17 cells, an increased number of other T cells, such as Th1 and Th9 cells, was detected in the lungs of patients with asthma [41,42]. Both Th1 and Th9 cells could potentially induce significant airway hyperresponsiveness in normal mice following adoptive transfer and allergen challenge [22,23,43,44,45]. Regardless of differences in the dominant cellular populations accumulated in the lungs, e.g., neutrophils induced by Th1 and Th17 and eosinophils and neutrophils induced by Th9 cells, most non-T2 responses tend to exhibit steroid resistance [23,38].

3. LAT1 and Its Inhibitors

Amino acid delivery into cells is regulated via transporters, termed solute carriers (SLCs). Among >60 amino acid transporters with differential specificity and potency in humans [46], LAT1 (SLC7A5) transports large neutral amino acids, mostly classified as essential amino acids, including branched-chain amino acids (Leu, Ile, Val), aromatic amino acids (Phe, Tyr, Trp, His), and methionine in a sodium-independent manner [47]. LAT1 forms a complex with CD98 (SLC3A2/4F2 heavy chain) to achieve its stable localization at the plasma membrane [48]. The biological significance of LAT1 has been reported, particularly its gene targeting strategy. Reportedly, Slc7a5-null mouse embryos display neural and limb bud outgrowth defects [49]. Additionally, Slc7a5-deficient mice displayed enhanced osteoclastogenesis and bone loss [50]. Notably, LAT1 is preferentially expressed in diverse human cancer cells [51,52]. Accumulating evidence suggests that LAT1 inhibition can effectively regulate the growth of various tumor cells [51]. Knockdown of human LAT1 using small interfering RNAs markedly downregulated the proliferation of MIA Paca-2 (pancreatic cancer), LNCaP and C4-2 (prostate cancer), HEC1A (endometrial cancer), and MKN-45 (gastric cancer) cells in vitro [53,54,55,56]. In addition, substantial in vivo tumor growth inhibition was observed following inoculation with anti-human LAT1 antibody-generating hybridoma cells [57]. Furthermore, oncogenic KRAS gene mutant-mediated colorectal tumor formation was alleviated in Slc7a5-knockout mice [58]. Given these findings, LAT1 has attracted worldwide attention as a novel target for cancer treatment.
For the clinical application of a LAT1-targeting strategy, researchers have attempted to produce LAT1 inhibitors (Figure 1). In the late 1960s, 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH) was identified as an amino acid transporter inhibitor. BCH was shown to inhibit LAT1; however, a markedly high concentration is required to mediate this action. Furthermore, BCH lacked selectivity toward LAT1; thus, all isoforms of system L transporters (LAT1, LAT2 LAT3 and LAT4) could be suppressed. Geier et al. identified 3-iodo-L-tyrosine, 3,5-diiodo-L-tyrosine, fenclonine, and acivicin as potential LAT1 inhibitors using comparative modeling and computational screening [59]. Triiodothyronine, a thyroid hormone, exerts an inhibitory effect on LAT1 with a relatively high affinity and selectivity [60]. Based on the T3 structure, Endou et al. generated a series of compounds exhibiting high LAT1 affinity. Among them, JPH203 (KYT-0353: (S)-2-amino-3-(4-((5-amino-2-phenylbenzo[d]oxazol-7-yl) methoxy)-3,5-dichlorophenyl) propanoic acid) showed potent and selective inhibitory effects against LAT1 [61] (Figure 1). In HT-29 cells, the half-maximal inhibitory concentrations (IC50s) of JPH203 were 0.06 and 4.1 µM for inhibiting leucine uptake and cell growth, respectively; however, at nearly the same dose range, JPH203 did not impact LAT2, indicating the substantial selectivity of JPH203 for LAT1. The significant efficacy of JPH203 has been demonstrated in cancer cell transplantation experiments in vivo [61]. Based on these findings, a first-in-human study for treating advanced solid tumors was completed, which confirmed the efficacy and safety of this compound [62].

4. LAT1 in T Cells

Activation of T cells following the recognition of allergen epitopes through T cell receptors (TCRs), accompanied by the participation of co-stimulatory molecules, facilitates nutrient uptake from the extracellular environment [63]. Targeted disruption of the Slc7a5 gene in CD4+ T cells did not affect T cell selection and homeostasis of the naïve T cell pool in the thymus [64], which is consistent with negligible LAT1 expression in naïve T cells [8]. However, LAT1 plays a substantial role in activated T cells (Figure 2). Stimulation of TCRs induced marked LAT1 expression in human T cells [8]. Given the suppression of leucine uptake, JPH203 could inhibit stimulation-induced production of IL-4, IL-5, IL-17, and interferon (IFN)-γ, as well as the proliferation of human and/or mouse T cells [8,9]. The blockade of amino acid incorporation into T cells downregulated the mechanistic target of rapamycin (mTOR)-dependent signaling pathway, oxidative phosphorylation (OXPHOS), glycolysis, and expression of cyclin and cyclin-dependent kinases [9]. Furthermore, upregulation of ChaC glutathione-specific gamma-glutamylcyclotransferase 1 and DNA damage-inducible transcript 3 by activating transcription factor 4, an amino acid starvation-inducible transcription factor, contributed to the suppression of T cell activation following JPH203 treatment [8,65] (Figure 2).

5. LAT1 in Allergic and Inflammatory Diseases

We hypothesized that the LAT1 inhibitor has a beneficial effect for treating allergic diseases in which activated T cells play a crucial role. We have developed murine models of allergic inflammation in various target tissues and demonstrated the essential contribution of allergen-specific T cells to their pathogenesis [66]. Consistent with the enhanced expression of LAT1 in skin-accumulated CD4+ T cells in patients with atopic dermatitis, JPH203 treatment could suppress allergen-induced skin inflammation in both ovalbumin (OVA)-immunized and OVA-specific Th2 cell-transferred mice [9]. Upregulation of LAT1 in CD4+ T cells was observed in imiquimod-induced psoriasis, another skin inflammation model [67]. JPH203 treatment and Slc7a5 gene disruption in CD4+ T cells alleviated disease progression with cytokine production in a psoriasis mouse model [67]. In addition, JPH203 could effectively suppress allergen-induced airway and nasal hyperresponsiveness in immunized and/or Th2-transferred mice [65,68]. Interestingly, JPH203 treatment ameliorated eosinophil accumulation in the nasal mucosa but not in the lungs [65,68]. Pharmacological inhibition of LAT1 is a novel promising strategy to control allergic and inflammatory disorders.

6. Possible Management of Steroid-Resistant Asthma by LAT1 Inhibitors

In addition to improving Th2-driven asthma phenotypes in mice, we observed that JPH203 could effectively suppress IL-17A production by human T cells [8]. Therefore, pharmacological LAT1 inhibition may afford therapeutic benefits against steroid-resistant asthma, at least when mediated via Th17 cells. We are currently investigating the effect of JPH203 on allergic airway inflammation in mice transferred with in vitro-differentiated Th17 cells. Preliminary findings indicated that JPH203 administration suppressed allergen-induced airway hyperresponsiveness, neutrophil accumulation, and IL-17 production in the lungs, whereas dexamethasone treatment failed to demonstrate any significant improvement (manuscript in preparation). On the other hand, targeted deletion of LAT1 in skin-resident RORγt-expressing cells containing Th17 cells and ILCs significantly improved imiquimod-induced psoriasis in mice [67]. JPH203 also suppressed IFN-γ production by activated human T cells [8], suggesting the involvement of LAT1 in other steroid-resistant mechanisms (Figure 3).

7. Conclusions

Despite efforts to develop effective treatments, a suitable strategy to control steroid-resistant asthma has not been successfully established. The technique of depleting the nutrients in T cells, distinct from current treatment strategies, could potentially modulate steroid-resistant mechanisms. Phase II clinical trials assessing JPH203 for cancer treatment are ongoing, highlighting its safety and usefulness. Further in-depth investigations evaluating JPH203, including its efficacy, pharmacokinetics, and formulations, are warranted to establish its potential application for managing intractable asthma.

Author Contributions

Conceptualization, K.H. and O.K.; Funding acquisition, K.H. and O.K.; Project administration, K.H. and O.K.; Visualization, K.H. and O.K.; Writing—Original draft, K.H.; Writing—Review and editing, O.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Grant-in-Aid for JSPS KAKENHI to Keitaro Hayashi (No. 21K06581) and to Osamu Kaminuma (No. 18K19561), Joint Research Grant of the Research Center for Radiation Disaster Medical Science to Keitaro Hayashi, Triangle Project of the Research Center for Radiation Disaster Medical Science to Osamu Kaminuma, and funding from the OTC Self-Medication Promotion Foundation and Smoking Research Foundation to Osamu Kaminuma.

Acknowledgments

The authors thank Hitoshi Endou and J-Pharma Co., Ltd. for valuable advice and providing JPH203.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Moldaver, D.M.; Larché, M.; Rudulier, C.D. An Update on Lymphocyte Subtypes in Asthma and Airway Disease. Chest 2017, 151, 1122–1130. [Google Scholar] [CrossRef] [PubMed]
  2. Hamid, Q.; Azzawi, M.; Ying, S.; Moqbel, R.; Wardlaw, A.J.; Corrigan, C.J.; Bradley, B.; Durham, S.R.; Collins, J.V.; Jeffery, P.K. Expression of mRNA for interleukin-5 in mucosal bronchial biopsies from asthma. J. Clin. Investig. 1991, 87, 1541–1546. [Google Scholar] [CrossRef] [PubMed]
  3. Walker, C.; Virchow, J.C., Jr.; Bruijnzeel, P.L.; Blaser, K. T cell subsets and their soluble products regulate eosinophilia in allergic and nonallergic asthma. J. Immunol. 1991, 146, 1829–1835. [Google Scholar] [PubMed]
  4. Pelaia, C.; Crimi, C.; Vatrella, A.; Tinello, C.; Terracciano, R.; Pelaia, G. Molecular Targets for Biological Therapies of Severe Asthma. Front. Immunol. 2020, 11, 603312. [Google Scholar] [CrossRef]
  5. Eyerich, S.; Metz, M.; Bossios, A.; Eyerich, K. New biological treatments for asthma and skin allergies. Allergy 2020, 75, 546–560. [Google Scholar] [CrossRef] [Green Version]
  6. Han, X.; Krempski, J.W.; Nadeau, K. Advances and novel developments in mechanisms of allergic inflammation. Allergy 2020, 75, 3100–3111. [Google Scholar] [CrossRef]
  7. Henderson, I.; Caiazzo, E.; McSharry, C.; Guzik, T.J.; Maffia, P. Why do some asthma patients respond poorly to glucocorticoid therapy? Pharmacol. Res. 2020, 160, 105189. [Google Scholar] [CrossRef]
  8. Hayashi, K.; Jutabha, P.; Endou, H.; Sagara, H.; Anzai, N. LAT1 is a critical transporter of essential amino acids for immune reactions in activated human T cells. J. Immunol. 2013, 191, 4080–4085. [Google Scholar] [CrossRef]
  9. Hayashi, K.; Kaminuma, O.; Nishimura, T.; Saeki, M.; Matsuoka, K.; Hiroi, T.; Jutabha, P.; Iwata, Y.; Sugiura, K.; Owada, T.; et al. LAT1-specific inhibitor is effective against T cell-mediated allergic skin inflammation. Allergy 2020, 75, 463–467. [Google Scholar] [CrossRef]
  10. Sze, E.; Bhalla, A.; Nair, P. Mechanisms and therapeutic strategies for non-T2 asthma. Allergy 2020, 75, 311–325. [Google Scholar] [CrossRef] [Green Version]
  11. Agache, I.; Eguiluz-Gracia, I.; Cojanu, C.; Laculiceanu, A.; Del Giacco, S.; Zemelka-Wiacek, M.; Kosowska, A.; Akdis, C.A.; Jutel, M. Advances and highlights in asthma in 2021. Allergy 2021, 76, 3390–3407. [Google Scholar] [CrossRef] [PubMed]
  12. McDowell, P.J.; Heaney, L.G. Different endotypes and phenotypes drive the heterogeneity in severe asthma. Allergy 2020, 75, 302–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Nabe, T. Steroid-Resistant Asthma and Neutrophils. Biol. Pharm. Bull. 2020, 43, 31–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Walsh, E.R.; Sahu, N.; Kearley, J.; Benjamin, E.; Kang, B.H.; Humbles, A.; August, A. Strain-specific requirement for eosinophils in the recruitment of T cells to the lung during the development of allergic asthma. J. Exp. Med. 2008, 205, 1285–1292. [Google Scholar] [CrossRef] [PubMed]
  15. Lee, J.J.; Dimina, D.; Macias, M.P.; Ochkur, S.I.; McGarry, M.P.; O’Neill, K.R.; Protheroe, C.; Pero, R.; Nguyen, T.; Cormier, S.A.; et al. Defining a link with asthma in mice congenitally deficient in eosinophils. Science 2004, 305, 1773–1776. [Google Scholar] [CrossRef]
  16. Ohtomo, T.; Kaminuma, O.; Yamada, J.; Kitamura, N.; Abe, A.; Kobayashi, N.; Suko, M.; Mori, A. Eosinophils are required for the induction of bronchial hyperresponsiveness in a Th transfer model of BALB/c background. Int. Arch. Allergy Immunol. 2010, 152 (Suppl. S1), 79–82. [Google Scholar] [CrossRef]
  17. Asano, K.; Ueki, S.; Tamari, M.; Imoto, Y.; Fujieda, S.; Taniguchi, M. Adult-onset eosinophilic airway diseases. Allergy 2020, 75, 3087–3099. [Google Scholar] [CrossRef]
  18. Lee, L.Y.; Hew, G.S.Y.; Mehta, M.; Shukla, S.D.; Satija, S.; Khurana, N.; Anand, K.; Dureja, H.; Singh, S.K.; Mishra, V.; et al. Targeting eosinophils in respiratory diseases: Biological axis, emerging therapeutics and treatment modalities. Life Sci. 2021, 267, 118973. [Google Scholar] [CrossRef]
  19. Guntern, P.; Eggel, A. Past, present, and future of anti-IgE biologics. Allergy 2020, 75, 2491–2502. [Google Scholar] [CrossRef] [Green Version]
  20. Okayama, Y.; Matsumoto, H.; Odajima, H.; Takahagi, S.; Hide, M.; Okubo, K. Roles of omalizumab in various allergic diseases. Allergol. Int. 2020, 69, 167–177. [Google Scholar] [CrossRef]
  21. Ogawa, K.; Kaminuma, O.; Kikkawa, H.; Nakata, A.; Asahina, M.; Egan, R.W.; Akiyama, K.; Mori, A. Transient contribution of mast cells to pulmonary eosinophilia but not to hyper-responsiveness. Clin. Exp. Allergy 2002, 32, 140–148. [Google Scholar] [CrossRef] [PubMed]
  22. Kaminuma, O.; Ohtomo, T.; Mori, A.; Nagakubo, D.; Hieshima, K.; Ohmachi, Y.; Noda, Y.; Katayama, K.; Suzuki, K.; Motoi, Y.; et al. Selective down-regulation of Th2 cell-mediated airway inflammation in mice by pharmacological intervention of CCR4. Clin. Exp. Allergy 2012, 42, 315–325. [Google Scholar] [CrossRef] [PubMed]
  23. Saeki, M.; Kaminuma, O.; Nishimura, T.; Kitamura, N.; Mori, A.; Hiroi, T. Th9 cells induce steroid-resistant bronchial hyperresponsiveness in mice. Allergol. Int. 2017, 66S, S35–S40. [Google Scholar] [CrossRef]
  24. Corrigan, C.J.; Haczku, A.; Gemou-Engesaeth, V.; Doi, S.; Kikuchi, Y.; Takatsu, K.; Durham, S.R.; Kay, A.B. CD4 T-lymphocyte activation in asthma is accompanied by increased serum concentrations of interleukin-5. Effect of glucocorticoid therapy. Am. Rev. Respir. Dis. 1993, 147, 540–547. [Google Scholar] [CrossRef]
  25. Akdis, C.A.; Arkwright, P.D.; Brüggen, M.C.; Busse, W.; Gadina, M.; Guttman-Yassky, E.; Kabashima, K.; Mitamura, Y.; Vian, L.; Wu, J.; et al. Type 2 immunity in the skin and lungs. Allergy 2020, 75, 1582–1605. [Google Scholar] [CrossRef] [PubMed]
  26. Hong, H.; Liao, S.; Chen, F.; Yang, Q.; Wang, D.Y. Role of IL-25, IL-33, and TSLP in triggering united airway diseases toward type 2 inflammation. Allergy 2020, 75, 2794–2804. [Google Scholar] [CrossRef] [PubMed]
  27. Maizels, R.M. Regulation of immunity and allergy by helminth parasites. Allergy 2020, 75, 524–534. [Google Scholar] [CrossRef] [Green Version]
  28. De Lucía Finkel, P.; Xia, W.; Jefferies, W.A. Beyond Unconventional: What Do We Really Know about Group 2 Innate Lymphoid Cells? J. Immunol. 2021, 206, 1409–1417. [Google Scholar] [CrossRef]
  29. Nakajima, S.; Kabata, H.; Kabashima, K.; Asano, K. Anti-TSLP antibodies: Targeting a master regulator of type 2 immune responses. Allergol. Int. 2020, 69, 197–203. [Google Scholar] [CrossRef]
  30. Kabata, H.; Moro, K.; Fukunaga, K.; Suzuki, Y.; Miyata, J.; Masaki, K.; Betsuyaku, T.; Koyasu, S.; Asano, K. Thymic stromal lymphopoietin induces corticosteroid resistance in natural helper cells during airway inflammation. Nat. Commun. 2013, 4, 2675. [Google Scholar] [CrossRef] [Green Version]
  31. Papadopoulos, N.G.; Barnes, P.; Canonica, G.W.; Gaga, M.; Heaney, L.; Menzies-Gow, A.; Kritikos, V.; Fitzgerald, M. The evolving algorithm of biological selection in severe asthma. Allergy 2020, 75, 1555–1563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Cosmi, L.; Liotta, F.; Maggi, E.; Romagnani, S.; Annunziato, F. Th17 cells: New players in asthma pathogenesis. Allergy 2011, 66, 989–998. [Google Scholar] [CrossRef] [PubMed]
  33. Chakir, J.; Shannon, J.; Molet, S.; Fukakusa, M.; Elias, J.; Laviolette, M.; Boulet, L.P.; Hamid, Q. Airway remodeling-associated mediators in moderate to severe asthma: Effect of steroids on TGF-beta, IL-11, IL-17, and type I and type III collagen expression. J. Allergy Clin. Immunol. 2003, 111, 1293–1298. [Google Scholar] [CrossRef] [PubMed]
  34. Barczyk, A.; Pierzchala, W.; Sozañska, E. Interleukin-17 in sputum correlates with airway hyperresponsiveness to methacholine. Respir. Med. 2003, 97, 726–733. [Google Scholar] [CrossRef] [Green Version]
  35. Molet, S.; Hamid, Q.; Davoine, F.; Nutku, E.; Taha, R.; Pagé, N.; Olivenstein, R.; Elias, J.; Chakir, J. IL-17 is increased in asthmatic airways and induces human bronchial fibroblasts to produce cytokines. J. Allergy Clin. Immunol. 2001, 108, 430–438. [Google Scholar] [CrossRef]
  36. Nanzer, A.M.; Chambers, E.S.; Ryanna, K.; Richards, D.F.; Black, C.; Timms, P.M.; Martineau, A.R.; Griffiths, C.J.; Corrigan, C.J.; Hawrylowicz, C.M. Enhanced production of IL-17A in patients with severe asthma is inhibited by 1α,25-dihydroxyvitamin D3 in a glucocorticoid-independent fashion. J. Allergy Clin. Immunol. 2013, 132, 297–304.e3. [Google Scholar] [CrossRef]
  37. Kudo, M.; Melton, A.C.; Chen, C.; Engler, M.B.; Huang, K.E.; Ren, X.; Wang, Y.; Bernstein, X.; Li, J.T.; Atabai, K.; et al. IL-17A produced by αβ T cells drives airway hyper-responsiveness in mice and enhances mouse and human airway smooth muscle contraction. Nat. Med. 2012, 18, 547–554. [Google Scholar] [CrossRef] [Green Version]
  38. McKinley, L.; Alcorn, J.F.; Peterson, A.; Dupont, R.B.; Kapadia, S.; Logar, A.; Henry, A.; Irvin, C.G.; Piganelli, J.D.; Ray, A.; et al. TH17 cells mediate steroid-resistant airway inflammation and airway hyperresponsiveness in mice. J. Immunol. 2008, 181, 4089–4097. [Google Scholar] [CrossRef] [Green Version]
  39. Lajoie, S.; Lewkowich, I.P.; Suzuki, Y.; Clark, J.R.; Sproles, A.A.; Dienger, K.; Budelsky, A.L.; Wills-Karp, M. Complement-mediated regulation of the IL-17A axis is a central genetic determinant of the severity of experimental allergic asthma. Nat. Immunol. 2010, 11, 928–935. [Google Scholar] [CrossRef] [Green Version]
  40. Busse, W.W.; Holgate, S.; Kerwin, E.; Chon, Y.; Feng, J.; Lin, J.; Lin, S.L. Randomized, double-blind, placebo-controlled study of brodalumab, a human anti-IL-17 receptor monoclonal antibody, in moderate to severe asthma. Am. J. Respir. Crit. Care Med. 2013, 188, 1294–1302. [Google Scholar] [CrossRef]
  41. Raundhal, M.; Morse, C.; Khare, A.; Oriss, T.B.; Milosevic, J.; Trudeau, J.; Huff, R.; Pilewski, J.; Holguin, F.; Kolls, J.; et al. High IFN-γ and low SLPI mark severe asthma in mice and humans. J. Clin. Investig. 2015, 125, 3037–3050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Erpenbeck, V.J.; Hohlfeld, J.M.; Volkmann, B.; Hagenberg, A.; Geldmacher, H.; Braun, A.; Krug, N. Segmental allergen challenge in patients with atopic asthma leads to increased IL-9 expression in bronchoalveolar lavage fluid lymphocytes. J. Allergy Clin. Immunol. 2003, 111, 1319–1327. [Google Scholar] [CrossRef] [PubMed]
  43. Staudt, V.; Bothur, E.; Klein, M.; Lingnau, K.; Reuter, S.; Grebe, N.; Gerlitzki, B.; Hoffmann, M.; Ulges, A.; Taube, C.; et al. Interferon-Regulatory Factor 4 Is Essential for the Developmental Program of T Helper 9 Cells. Immunity 2010, 33, 192–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Cui, J.; Pazdziorko, S.; Miyashiro, J.S.; Thakker, P.; Pelker, J.W.; Declercq, C.; Jiao, A.; Gunn, J.; Mason, L.; Leonard, J.P.; et al. TH1-mediated airway hyperresponsiveness independent of neutrophilic inflammation. J. Allergy Clin. Immunol. 2005, 115, 309–315. [Google Scholar] [CrossRef]
  45. Takaoka, A.; Tanaka, Y.; Tsuji, T.; Jinushi, T.; Hoshino, A.; Asakura, Y.; Mita, Y.; Watanabe, K.; Nakaike, S.; Togashi, Y.; et al. A critical role for mouse CXC chemokine(s) in pulmonary neutrophilia during Th type 1-dependent airway inflammation. J. Immunol. 2001, 167, 2349–2353. [Google Scholar] [CrossRef] [Green Version]
  46. Kandasamy, P.; Gyimesi, G.; Kanai, Y.; Hediger, M.A. Amino acid transporters revisited: New views in health and disease. Trends Biochem. Sci. 2018, 43, 752–789. [Google Scholar] [CrossRef]
  47. Kanai, Y.; Segawa, H.; Miyamoto, K.i.; Uchino, H.; Takeda, E.; Endou, H. Expression cloning and characterization of a transporter for large neutral amino acids activated by the heavy chain of 4F2 antigen (CD98). J. Biol. Chem. 1998, 273, 23629–23632. [Google Scholar] [CrossRef] [Green Version]
  48. Nakamura, E.; Sato, M.; Yang, H.; Miyagawa, F.; Harasaki, M.; Tomita, K.; Matsuoka, S.; Noma, A.; Iwai, K.; Minato, N. 4F2 (CD98) heavy chain is associated covalently with an amino acid transporter and controls intracellular trafficking and membrane topology of 4F2 heterodimer. J. Biol. Chem. 1999, 274, 3009–3016. [Google Scholar] [CrossRef] [Green Version]
  49. Poncet, N.; Halley, P.A.; Lipina, C.; Gierliński, M.; Dady, A.; Singer, G.A.; Febrer, M.; Shi, Y.B.; Yamaguchi, T.P.; Taylor, P.M.; et al. Wnt regulates amino acid transporter Slc7a5 and so constrains the integrated stress response in mouse embryos. EMBO Rep. 2020, 21, e48469. [Google Scholar] [CrossRef]
  50. Ozaki, K.; Yamada, T.; Horie, T.; Ishizaki, A.; Hiraiwa, M.; Iezaki, T.; Park, G.; Fukasawa, K.; Kamada, H.; Tokumura, K.; et al. The L-type amino acid transporter LAT1 inhibits osteoclastogenesis and maintains bone homeostasis through the mTORC1 pathway. Sci. Signal. 2019, 12, eaaw3921. [Google Scholar] [CrossRef]
  51. Kanai, Y. Amino acid transporter LAT1 (SLC7A5) as a molecular target for cancer diagnosis and therapeutics. Pharmacol. Ther. 2021, 230, 107964. [Google Scholar] [CrossRef] [PubMed]
  52. Hayashi, K.; Anzai, N. Novel therapeutic approaches targeting L-type amino acid transporters for cancer treatment. World J. Gastrointest. Oncol. 2017, 9, 21–29. [Google Scholar] [CrossRef] [PubMed]
  53. Hayashi, K.; Jutabha, P.; Endou, H.; Anzai, N. c-Myc is crucial for the expression of LAT1 in MIA Paca-2 human pancreatic cancer cells. Oncol. Rep. 2012, 28, 862–866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Xu, M.; Sakamoto, S.; Matsushima, J.; Kimura, T.; Ueda, T.; Mizokami, A.; Kanai, Y.; Ichikawa, T. Up-Regulation of LAT1 during Antiandrogen Therapy Contributes to Progression in Prostate Cancer Cells. J. Urol. 2016, 195, 1588–1597. [Google Scholar] [CrossRef] [PubMed]
  55. Marshall, A.D.; van Geldermalsen, M.; Otte, N.J.; Anderson, L.A.; Lum, T.; Vellozzi, M.A.; Zhang, B.K.; Thoeng, A.; Wang, Q.; Rasko, J.E.J.; et al. LAT1 is a putative therapeutic target in endometrioid endometrial carcinoma. Int. J. Cancer. 2016, 139, 2529–2539. [Google Scholar] [CrossRef]
  56. Wang, J.; Chen, X.; Su, L.; Li, P.; Liu, B.; Zhu, Z. LAT-1 functions as a promotor in gastric cancer associated with clinicopathologic features. Biomed. Pharmacother. 2013, 67, 693–699. [Google Scholar] [CrossRef]
  57. Ohkawa, M.; Ohno, Y.; Masuko, K.; Takeuchi, A.; Suda, K.; Kubo, A.; Kawahara, R.; Okazaki, S.; Tanaka, T.; Saya, H.; et al. Oncogenicity of L-type amino-acid transporter 1 (LAT1) revealed by targeted gene disruption in chicken DT40 cells: LAT1 is a promising molecular target for human cancer therapy. Biochem. Biophys. Res. Commun. 2011, 406, 649–655. [Google Scholar] [CrossRef]
  58. Najumudeen, A.K.; Ceteci, F.; Fey, S.K.; Hamm, G.; Steven, R.T.; Hall, H.; Nikula, C.J.; Dexter, A.; Murta, T.; Race, A.M.; et al. The amino acid transporter SLC7A5 is required for efficient growth of KRAS-mutant colorectal cancer. Nat. Genet. 2021, 53, 16–26. [Google Scholar] [CrossRef]
  59. Geier, E.G.; Schlessinger, A.; Fan, H.; Gable, J.E.; Irwin, J.J.; Sali, A.; Giacomini, K.M. Structure-based ligand discovery for the Large-neutral Amino Acid Transporter 1, LAT-1. Proc. Natl. Acad. Sci. USA 2013, 110, 5480–5485. [Google Scholar] [CrossRef] [Green Version]
  60. Uchino, H.; Kanai, Y.; Kim, D.K.; Wempe, M.F.; Chairoungdua, A.; Morimoto, E.; Anders, M.W.; Endou, H. Transport of amino acid-related compounds mediated by L-type amino acid transporter 1 (LAT1): Insights into the mechanisms of substrate recognition. Mol. Pharmacol. 2002, 61, 729–737. [Google Scholar] [CrossRef] [Green Version]
  61. Oda, K.; Hosoda, N.; Endo, H.; Saito, K.; Tsujihara, K.; Yamamura, M.; Sakata, T.; Anzai, N.; Wempe, M.F.; Kanai, Y.; et al. L-type amino acid transporter 1 inhibitors inhibit tumor cell growth. Cancer Sci. 2010, 101, 173–179. [Google Scholar] [CrossRef]
  62. Okano, N.; Naruge, D.; Kawai, K.; Kobayashi, T.; Nagashima, F.; Endou, H.; Furuse, J. First-in-human phase I study of JPH203, an L-type amino acid transporter 1 inhibitor, in patients with advanced solid tumors. Investig. New Drugs. 2020, 38, 1495–1506. [Google Scholar] [CrossRef]
  63. Hayashi, K.; Ouchi, M.; Endou, H.; Anzai, N. HOXB9 acts as a negative regulator of activated human T cells in response to amino acid deficiency. Immunol. Cell Biol. 2016, 94, 612–617. [Google Scholar] [CrossRef]
  64. Sinclair, L.V.; Rolf, J.; Emslie, E.; Shi, Y.B.; Taylor, P.M.; Cantrell, D.A. Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation. Nat. Immunol. 2013, 14, 500–508. [Google Scholar] [CrossRef] [Green Version]
  65. Kaminuma, O.; Nishimura, T.; Saeki, M.; Yamasaki, N.; Ogata, S.; Fujita, T.; Endou, H.; Hayashi, K. L-type amino acid transporter 1 (LAT1)-specific inhibitor is effective against T cell-mediated nasal hyperresponsiveness. Allergol. Int. 2020, 69, 455–458. [Google Scholar] [CrossRef]
  66. Miura, K.; Inoue, K.; Ogura, A.; Kaminuma, O. Role of CD4+ T Cells in Allergic Airway Diseases: Learning from Murine Models. Int. J. Mol. Sci. 2020, 21, 7480. [Google Scholar] [CrossRef]
  67. Cibrian, D.; Castillo-González, R.; Fernández-Gallego, N.; de la Fuente, H.; Jorge, I.; Saiz, M.L.; Punzón, C.; Ramírez-Huesca, M.; Vicente-Manzanares, M.; Fresno, M.; et al. Targeting L-type amino acid transporter 1 in innate and adaptive T cells efficiently controls skin inflammation. J. Allergy Clin. Immunol. 2020, 145, 199–214.e11. [Google Scholar] [CrossRef] [Green Version]
  68. Ito, D.; Miura, K.; Saeki, M.; Yamasaki, N.; Ogata, S.; Koyama, T.; Hiroi, T.; Mori, A.; Endou, H.; Hayashi, K.; et al. L-type amino acid transporter 1 inhibitor suppresses murine Th2 cell-mediated bronchial hyperresponsiveness independently of eosinophil accumulation. Asia Pac. Allergy 2021, 11, e33. [Google Scholar] [CrossRef]
Figure 1. Structures of LAT1 inhibitors. BCH (2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid) inhibits all L-type amino acid transporters. Triiodothyronine is a thyroid hormone that exerts an inhibitory effect on LAT1. Additionally, 3-iodo-L-tyrosine was identified as a LAT1 inhibitor by comparative modeling and computational screening. JPH203 is a high-affinity LAT1-specific inhibitor.
Figure 1. Structures of LAT1 inhibitors. BCH (2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid) inhibits all L-type amino acid transporters. Triiodothyronine is a thyroid hormone that exerts an inhibitory effect on LAT1. Additionally, 3-iodo-L-tyrosine was identified as a LAT1 inhibitor by comparative modeling and computational screening. JPH203 is a high-affinity LAT1-specific inhibitor.
Biomolecules 12 00553 g001
Figure 2. Regulation of T cell activation by L-type amino acid transporter (LAT) 1. In activated T cells, LAT1 is upregulated to facilitate amino acid uptake. JPH203, a LAT1-specific inhibitor, attenuates mechanistic target of rapamycin (mTOR) and metabolic reaction, and contrary, activates activating transcription factor (ATF) 4, leading to inhibition of T cell activation. CHAC1: ChaC glutathione specific gamma-glutamylcyclotransferase 1, CDK: cyclin-dependent kinase, DDIT3: DNA damage-inducible transcript 3, GCN2: general control nonderepressible 2, OXPHOS: oxidative phosphorylation, TCR: T cell receptor.
Figure 2. Regulation of T cell activation by L-type amino acid transporter (LAT) 1. In activated T cells, LAT1 is upregulated to facilitate amino acid uptake. JPH203, a LAT1-specific inhibitor, attenuates mechanistic target of rapamycin (mTOR) and metabolic reaction, and contrary, activates activating transcription factor (ATF) 4, leading to inhibition of T cell activation. CHAC1: ChaC glutathione specific gamma-glutamylcyclotransferase 1, CDK: cyclin-dependent kinase, DDIT3: DNA damage-inducible transcript 3, GCN2: general control nonderepressible 2, OXPHOS: oxidative phosphorylation, TCR: T cell receptor.
Biomolecules 12 00553 g002
Figure 3. Possible management of steroid-resistant asthma by JPH203. T helper (Th) 17 and Th1 cells activate neutrophils, which causes progression of steroid-resistant asthma. JPH203 potentially terminates the disease cascade by starving amino acids in those cells. LAT1: L-type amino acid transporter 1, ROS: reactive oxygen species, NEU: neutrophil.
Figure 3. Possible management of steroid-resistant asthma by JPH203. T helper (Th) 17 and Th1 cells activate neutrophils, which causes progression of steroid-resistant asthma. JPH203 potentially terminates the disease cascade by starving amino acids in those cells. LAT1: L-type amino acid transporter 1, ROS: reactive oxygen species, NEU: neutrophil.
Biomolecules 12 00553 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hayashi, K.; Kaminuma, O. Therapeutic Potential for Intractable Asthma by Targeting L-Type Amino Acid Transporter 1. Biomolecules 2022, 12, 553. https://doi.org/10.3390/biom12040553

AMA Style

Hayashi K, Kaminuma O. Therapeutic Potential for Intractable Asthma by Targeting L-Type Amino Acid Transporter 1. Biomolecules. 2022; 12(4):553. https://doi.org/10.3390/biom12040553

Chicago/Turabian Style

Hayashi, Keitaro, and Osamu Kaminuma. 2022. "Therapeutic Potential for Intractable Asthma by Targeting L-Type Amino Acid Transporter 1" Biomolecules 12, no. 4: 553. https://doi.org/10.3390/biom12040553

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop