Search results

Filters

  • Journals
  • Authors
  • Keywords
  • Date
  • Type

Search results

Number of results: 5
items per page: 25 50 75
Sort by:
Download PDF Download RIS Download Bibtex

Abstract

The main purpose of the study was to determine the safety of oclacitinib (OCL), a Janus kinase inhibitor, with respect of its effect on CD4 + and CD8 + T cells as well as B cells in the lymphoid tissue. The mice were treated orally with OCL at a dose of 2.7 mg/kg for 14 days and peripheral blood, head and neck lymph nodes (HNLNs), mediastinal lymph nodes (MLNs) and spleen were collected. The study found that OCL induced depletion of CD4 + T cells in the HNLNs and MLNs, while it did not affect the absolute count of CD8 + T cells in these tissues. Also OCL caused a loss of B cells in the HNLNs, although not in the MLNs. Moreover, OCL depleted B cells in the peripheral blood, but did not affect the absolute count of CD4 + and CD8 + T cells. Thus, it can be concluded that OCL may induce a depletive effect on CD4 + and CD8 + T cells as well as B cells in the lymphoid tissue. This effect should be seen as an unfavorable one, especially in patients with infections. Therefore, a clinical implication is that in such patients, the benefit/risk ratio should be thoroughly considered by clinicians. Moreover, OCL reduced the absolute count of eosinophils, basophils, neutrophils and monocytes. However, it is uncertain whether this effect should be considered to be of clinical importance because the levels of these cells were within the physiological range. It is possible that the depletive effect of OCL toward T and B cells, as well as eosinophils and basophils may contribute to the beneficial effects of the drug in the treatment of skin allergic diseases.
Go to article

Bibliography

  1. Apoquel® Summary of Product Characteristics available here: www.ema.europa.eu/en/documents/product-information/apoquel-epar-product-information_en.pdf.
  2. Banovic F, Tarigo J, Gordon H, Barber JP, Gogal RM Jr (2019) Immunomodulatory in vitro effects of oclacitinib on canine T-cell proliferation and cytokine production. Vet Dermatol 30: 17-e6.
  3. Barry M, Bleackley RC (2002) Cytotoxic T lymphocytes: all roads lead to death. Nat Rev Immunol 2: 401-409.
  4. Benucci M, Bernardini P, Coccia C, De Luca R, Levani J, Economou A, Damiani A, Russo E, Amedei A, Guiducci S, Bartoloni E, Manfredi M, Grossi V, Infantino M, Perricone C (2023) JAK inhibitors and autoimmune rheumatic diseases. Autoimmun Rev 22: 103276.
  5. Cetkovic-Cvrlje M, Olson M, Ghate K (2012) Targeting Janus tyrosine kinase 3 (JAK3) with an inhibitor induces secretion of TGF-β by CD4+ T cells. Cell Mol Immunol 9: 350-360.
  6. De Caro Martins G, da Costa-Val AP, Coura FM, Diamantino GM, Nogueira MM, de Oliveira Melo-Junior OA, Giunchetti RC, da Silveira-Lemos D, Melo MM (2022) Immunomodulatory effect of long-term oclacitinib maleate therapy in dogs with atopic dermatitis. Vet Dermatol 33: 142-e40.
  7. Denti D, Caldin M, Ventura L, De Lucia M (2022) Prolonged twice-daily administration of oclacitinib for the control of canine atopic dermatitis: a retrospective study of 53 client-owned atopic dogs. Vet Dermatol 33: 149-e42.
  8. Gonzales AJ, Bowman JW, Fici GJ, Zhang M, Mann DW, Mitton-Fry M (2014) Oclacitinib (APOQUEL(®)) is a novel Janus kinase inhibitor with activity against cytokines involved in allergy. J Vet Pharmacol Ther 37: 317-324.
  9. Gottlieb SL, Martin DH, Xu F, Byrne GI, Brunham RC (2010) Summary: The natural history and immunobiology of Chlamydia trachomatis genital infection and implications for Chlamydia control. J Infect Dis 201 Suppl 2: S190-204.
  10. Hamann D, Baars PA, Rep MH, Hooibrink B, Kerkhof-Garde SR, Klein MR, van Lier RA (1997) Phenotypic and functional separation of memory and effector human CD8+ T cells. J Exp Med 186: 1407-1418.
  11. Hashimoto T, Yokozeki H, Karasuyama H, Satoh T (2023) IL-31–generating network in atopic dermatitis comprising macrophages, basophils, thymic stromal lymphopoietin, and periostin. J Allergy Clin Immunol 151: 737-746.
  12. James JM, Kagey-Sobotka A, Sampson HA (1993) Patients with severe atopic dermatitis have activated circulating basophils. J Allergy Clin Immunol 91: 1155-1162.
  13. Jasiecka-Mikołajczyk A, Jaroszewski JJ, Maślanka T (2018) Oclacitinib depletes canine CD4+ and CD8+ T cells in vitro. Res Vet Sci 121: 124-129.
  14. Jasiecka-Mikołajczyk A, Jaroszewski JJ, Maślanka T (2021) Oclacitinib, a Janus Kinase Inhibitor, Reduces the Frequency of IL-4- and IL-10-, but Not IFN-γ-, Producing Murine CD4+ and CD8+ T Cells and Counteracts the Induction of Type 1 Regulatory T Cells. Molecules 26: 5655.
  15. Lee S, Shah T, Yin C, Hochberg J, Ayello J, Morris E, van de Ven C, Cairo MS (2018) Ruxolitinib significantly enhances in vitro apoptosis in Hodgkin lymphoma and primary mediastinal B-cell lymphoma and survival in a lymphoma xenograft murine model. Oncotarget 9: 9776-9788.
  16. Majewska A, Dembele K, Dziendzikowska K, Prostek A, Gajewska M (2022) Cytokine and Lymphocyte Profiles in Dogs with Atopic Dermatitis after Allergen-Specific Immunotherapy. Vaccines (Basel) 10: 1037.
  17. Majewska A, Gajewska M, Dembele K, Maciejewski H, Prostek A, Jank M (2016) Lymphocytic, cytokine and transcriptomic profiles in peripheral blood of dogs with atopic dermatitis. BMC Vet Res 12: 174.
  18. Maślanka T, Otrocka-Domagała I, Zuśka-Prot M, Mikiewicz M, Przybysz J, Jasiecka A, Jaroszewski JJ (2016) IκB kinase β inhibitor, IMD-0354, prevents allergic asthma in a mouse model through inhibition of CD4(+) effector T cell responses in the lung-draining mediastinal lymph nodes. Eur J Pharmacol 775: 78-85.
  19. Nuttall TJ, Knight PA, McAleese SM, Lamb JR, Hill PB (2002) Expression of Th1, Th2 and immunosuppressive cytokine gene transcripts in canine atopic dermatitis. Clin Exp Allergy 32: 789-795.
  20. Olivry T, Dean GA, Tompkins MB, Dow JL, Moore PF (1999) Toward a canine model of atopic dermatitis: amplification of cytokine-gene transcripts in the skin of atopic dogs. Exp Dermatol 8: 204-211.
  21. Olivry T, Naydan DK, Moore PF (1997) Characterization of the cutaneous inflammatory infiltrate in canine atopic dermatitis. Am J Dermatopathol 19: 477-486.
  22. Ramirez GA, Yacoub MR, Ripa M, Mannina D, Cariddi A, Saporiti N, Ciceri F, Castagna A, Colombo G, Dagna L (2018) Eosinophils from Physiology to Disease: A Comprehensive Review. Biomed Res Int 2018: 9095275.
  23. Reagan-Shaw S, Nihal M, Ahmad N (2007) Dose translation from animal to human studies revisited. FASEB J 22: 659-661.
  24. Schlotter YM, Rutten VP, Riemers FM, Knol EF, Willemse T (2011) Lesional skin in atopic dogs shows a mixed Type-1 and Type-2 immune responsiveness. Vet Immunol Immunopathol 143: 20-26.
  25. Sinke JD, Thepen T, Bihari IC, Rutten VP, Willemse T (1997) Immunophenotyping of skin-infiltrating T-cell subsets in dogs with atopic dermatitis. Vet Immunol Immunopathol 57: 13-23.
  26. Stosović R, Bogić M (1998) The role of eosinophilic leukocytes in allergic inflammation. Srp Arh Celok Lek 126: 130-137.
  27. Wada T, Ishiwata K, Koseki H, Ishikura T, Ugajin T, Ohnuma N, Obata K, Ishikawa R, Yoshikawa S, Mukai K, Kawano Y, Minegishi Y, Yokozeki H, Watanabe N, Karasuyama H (2010) Selective ablation of basophils in mice reveals their nonredundant role in acquired immunity against ticks. J Clin Invest 120: 2867-2875.
  28. Wang S, Li H, Lian Z, Deng S (2023) The Role of m6A Modifications in B-Cell Development and B-Cell-Related Diseases. Int J Mol Sci 24: 4721.
  29. Weller PF, Spencer LA (2017) Functions of tissue-resident eosinophils. Nat Rev Immunol 17: 746-760.
Go to article

Authors and Affiliations

A. Jasiecka-Mikołajczyk
1
T. Maślanka
1

  1. Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-718 Olsztyn, Poland
Download PDF Download RIS Download Bibtex

Abstract

The purpose of this study was to verify the possibility of pharmacological induction of Foxp3 +CD25 +CD8 + and Foxp3 -CD103 +CD8 + T regulatory cells ‘armed’ with immunosuppressive molecules, i.e. CD39 and IL-10. To achieve this purpose, stimulated and unstimulated murine lymphocytes were exposed to IL-27, teriflunomide (TER) and all trans retinoic acid (ATRA). The study found that: (a) IL-27 induced CD39 expression on Foxp3 +CD25 +CD8 + T cells and the ability of CD103+Foxp3-CD8+ T cells to produce IL-10 as well as increasing the absolute number of IL-10 +CD103 +Foxp3 -CD8 + T cells; (b) TER induced Foxp3 expression in CD25+CD8+ T cells and CD103 expression on Foxp3 -CD8 + T cells as well as increasing the absolute number of Foxp3 +CD25 +CD8 + T cells; (c) ATRA induced the capacity of Foxp3 +CD25 +CD8 + T cells to produce IL-10. The following desired interactions were demonstrated between IL-27 and ATRA: (a) a strong synergistic effect with respect to increasing CD39 expression and the ability to produce IL-10 by Foxp3 +CD25 +CD8 + T cells; (b) a synergistic effect with respect to increasing the absolute count of CD39 +Foxp3 +CD25 +CD8 + T cells. The study revealed that TER abolished all these effects. Therefore, a combination of the tested agents did not induce the generation of Foxp3 +CD25 +CD8 + and Foxp3 -CD103+CD8+ T cells characterized by extensive CD39 expression and IL-10 production. Thus, in the context of the pharmacological induction of IL-10 +CD39 +Foxp3 +CD25 +CD8 + and IL-10 +CD103 +Foxp3 -CD8 + T cells, these findings strongly suggest that a combination of TER with IL-27 and/or ATRA does not provide any benefits over TER alone; moreover, such a combination may result in abolishing the desired effects exerted by IL-27 and/or ATRA.
Go to article

Bibliography

1. Aubagio®, assessment report. International non-proprietary name: Teriflunomide. Procedure No. EMEA/H/C/002514/ 0000. 27 June 2013 EMA/529295/2013. Available from here: https://www.ema.europa.eu/en/documents/assessmentreport/aubagio-epar-public-assessment-report_en.pdf
2. Bastid J, Cottalorda-Regairaz A, Alberici G, Bonnefoy N, Eliaou JF, Bensussan A (2013) ENTPD1/CD39 is a promising therapeutic target in oncology. Oncogene 32: 1743-1751.
3. Bastid J, Regairaz A, Bonnefoy N, Déjou C, Giustiniani J, Laheurte C, Cochaud S, Laprevotte E, Funck-Brentano E, Hemon P, Gros L, Bec N, Larroque C, Alberici G, Bensussan A, Eliaou JF (2015) Inhibition of CD39 enzymatic function at the surface of tumor cells alle-viates their immunosuppressive activity. Cancer Immunol Res 3: 254-265.
4. Batten M, Kljavin NM, Li J, Walter MJ, de Sauvage FJ, Ghilardi N (2008) Cutting edge: IL-27 is a potent inducer of IL-10 but not FoxP3 in murine T cells. J Immunol 180: 2752-2756.
5. Benson MJ, Pino-Lagos K, Rosemblatt M, Noelle RJ (2007) All-trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation. J Exp Med 204: 1765-1774.
6. Canale FP, Ramello MC, Núñez N, Araujo Furlan CL, Bossio SN, Gorosito Serrán M, Tosello Boari J, Del Castillo A, Ledesma M, Sedlik C, Piaggio E, Gruppi A, Acosta Rodríguez EA, Montes CL (2018) CD39 Expression Defines Cell Exhaustion in Tu-mor-Infiltrating CD8+ T Cells. Cancer Res 78: 115-128.
7. Chaput N, Louafi S, Bardier A, Charlotte F, Vaillant JC, Ménégaux F, Rosenzwajg M, Lemoine F, Klatzmann D, Taieb J (2009) Identi-fication of CD8+CD25+Foxp3+ suppressive T cells in colorectal cancer tissue. Gut 58: 520-529.
8. Churlaud G, Pitoiset, F, Jebbawi, F, Lorenzon R, Bellier B, Rosenzwajg M, Klatzmann D (2015) Human and Mouse CD8+CD25+FOXP3+ Regulatory T Cells at Steady State and during Interleukin-2 Therapy. Front Immunol 6: 171.
9. Correale J, Villa A (2010) Role of CD8+ CD25+ Foxp3+ regulatory T cells in multiple sclerosis. Ann Neurol 67: 625-638.
10. Eusebio M, Kraszula L, Kupczyk M, Kuna P, Pietruczuk M (2012) Low frequency of CD8+CD25+FOXP3(BRIGHT) T cells and FOXP3 mRNA expression in the peripheral blood of allergic asthma patients. J Biol Regul Homeost Agents 26: 211-220.
11. Friedman DJ, Künzli BM, A-Rahim YI, Sevigny J, Berberat PO, Enjyoji K, Csizmadia E, Friess H, Robson SC (2009) From the Cover: CD39 deletion exacerbates experimental murine colitis and human polymorphisms increase susceptibility to inflammatory bowel disease. Proc Natl Acad Sci USA 106: 16788-16793.
12. Jasiecka-Mikołajczyk A, Maślanka T (2023) Depletion of T and B cells in lymphoid tissues of mice induced by oclacitinib, a Janus ki-nase inhibitor. Pol J Vet Sci 26: 431-440.
13. Jasiecka-Mikołajczyk A, Socha P (2020) Teriflunomide inhibits activation-induced CD25 expression on T cells and may affect Foxp3-expressing regulatory T cells. Res Vet Sci 132: 17-27.
14. Jing J, Nelson C, Paik J, Shirasaka Y, Amory JK, Isoherranen N (2017) Physiologically Based Pharmacokinetic Model of All- trans-Retinoic Acid with Application to Cancer Populations and Drug Interactions. J Pharmacol Exp Ther 361: 246-258.
15. Kim G, Shinnakasu R, Saris CJ, Cheroutre H, Kronenberg M (2013) A novel role for IL-27 in mediating the survival of activated mouse CD4 T lymphocytes. J Immunol 190: 1510-1518.
16. Kiniwa Y, Miyahara Y, Wang HY, Peng W, Peng G, Wheeler TM, Thompson TC, Old LJ, Wang RF (2007) CD8+ Foxp3+ regulatory T cells mediate immunosuppression in prostate cancer. Clin Cancer Res 13: 6947-6958.
17. Lin L, Dai F, Wei J, Chen Z (2021) CD8+ Tregs ameliorate inflammatory reactions in a murine model of allergic rhinitis. Allergy Asthma Clin Immunol 17: 74.
18. Liston A, Aloulou M (2022) A fresh look at a neglected regulatory lineage: CD8+Foxp3+ Regulatory T cells. Immunol Lett 247: 22-26.
19. Liu Y, Lan Q, Lu L, Chen M, Xia Z, Ma J, Wang J, Fan H, Shen Y, Ryffel B, Brand D, Quismorio F, Liu Z, Horwitz DA, Xu A, Zheng SG (2014) Phenotypic and functional characteristic of a newly identified CD8+ Foxp3- CD103+ regulatory T cells. J Mol Cell Biol 6: 81-92.
20. Liu Z, Liu JQ, Talebian F, Wu LC, Li S, Bai XF (2013) IL-27 enhances the survival of tumor antigen-specific CD8+ T cells and pro-grams them into IL-10-producing, memory precursor-like effector cells. Eur J Immunol 43: 468-479.
21. Loza MJ, Anderson AS, O’Rourke KS, Wood J, Khan IU (2011) T-cell specific defect in expression of the NTPDase CD39 as a bi-omarker for lupus. Cell Immunol 271: 110-117.
22. Mahic M, Henjum K, Yaqub S, Bjørnbeth BA, Torgersen KM, Taskén K, Aandahl EM (2008) Generation of highly suppressive adap-tive CD8+CD25+FOXP3+ regulatory T cells by continuous antigen stimulation. Eur J Immunol 38: 640-646.
23. Maślanka T (2022) Effect of IL-27, teriflunomide and retinoic acid and their combinations on CD4+ T regulatory T cells – an in vitro study. Molecules 27: 8471.
24. Matsui M, Kishida T, Nakano H, Yoshimoto K, Shin-Ya M, Shimada T, Nakai S, Imanishi J, Yoshimoto T, Hisa Y, Mazda O (2009) Interleukin-27 activates natural killer cells and suppresses NK-resistant head and neck squamous cell carcinoma through inducing anti-body-dependent cellular cytotoxicity. Cancer Res 69: 2523-2530.
25. Ménoret S, Tesson L, Remy S, Gourain V, Sérazin C, Usal C, Guiffes A, Chenouard V, Ouisse LH, Gantier M, Heslan JM, Fourgeux C, Poschmann J, Guillonneau C, Anegon I (2023) CD4+ and CD8+ regulatory T cell characterization in the rat using a unique transgenic Foxp3-EGFP model. BMC Biol 21: 8.
26. Molodtsov A, Turk MJ (2018) Tissue Resident CD8 Memory T Cell Responses in Cancer and Autoimmunity. Front Immunol 9: 2810.
27. Murugaiyan G, Mittal A, Weiner HL (2010) Identification of an IL-27/osteopontin axis in dendritic cells and its modulation by IFN-gamma limits IL-17-mediated autoimmune inflammation. Proc Natl Acad Sci USA 107: 11495-11500.
28. Nwankwo E, Allington DR, Rivey MP (2012) Emerging oral immunomodulating agents – focus on teriflunomide for the treatment of multiple sclerosis. Degener Neurol Neuromuscul Dis 2: 15-28.
29. Ponthan F, Kogner P, Bjellerup P, Klevenvall L, Hassan M (2001) Bioavailability and dose-dependent anti-tumour effects of 9-cis retin-oic acid on human neuroblastoma xenografts in rat. Br J Cancer 85: 2004-2009.
30. Ringshausen I, Oelsner M, Bogner C, Peschel C, Decker T (2008) The immunomodulatory drug Leflunomide inhibits cell cycle progres-sion of B-CLL cells. Leukemia 22: 635-638.
31. Suzuki M, Konya C, Goronzy JJ, Weyand CM (2008) Inhibitory CD8+ T cells in autoimmune disease. Hum Immunol 69: 781-789.
32. Szondy Z, Reichert U, Fésüs L (1998) Retinoic acids regulate apoptosis of T lymphocytes through an interplay between RAR and RXR receptors. Cell Death Differ 5: 4-10.
33. Tang Q, Bluestone JA (2008) The Foxp3+ regulatory T cell: a jack of all trades, master of regulation. Nat Immunol 9: 239-244.
34. Wing JB, Tanaka A, Sakaguchi S (2019) Human FOXP3+ regulatory T cell heterogeneity and function in autoimmunity and cancer. Im-munity 50: 302-316.
35. Wen Z, Shimojima Y, Shirai T, Li Y, Ju J, Yang Z, Tian L, Goronzy JJ, Weyand CM (2016) NADPH oxidase deficiency underlies dysfunction of aged CD8+ Tregs. J Clin Invest 126: 1953-1967.
36. Xiao S, Jin H, Korn T, Liu SM, Oukka M, Lim B, Kuchroo VK (2008) Retinoic acid increases Foxp3+ regulatory T cells and inhibits development of Th17 cells by enhancing TGF-beta-driven Smad3 signaling and inhibiting IL-6 and IL-23 receptor expression. J Immu-nol 181: 2277-2284.
37. Zhong H, Liu Y, Xu Z, Liang P, Yang H, Zhang X, Zhao J, Chen J, Fu S, Tang Y, Lv J, Wang J, Olsen N, Xu A, Zheng SG (2018) TGF-β-Induced CD8+CD103+ regulatory T cells show potent therapeutic effect on chronic graft-versus-host disease lupus by suppress-ing B cells. Front Immunol 9: 35.
Go to article

Authors and Affiliations

T. Maślanka
1
A. Jasiecka-Mikołajczyk
1

  1. Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-718 Olsztyn, Poland
Download PDF Download RIS Download Bibtex

Abstract

Due to the unrecognized effect of tigecycline (TIG) on CD4+ and CD8+ T cells, the present study has been undertaken in order to determine whether the drug can affect these cells in respect of their counts, and the production of IFN-γ, IL-17 (pro-inflammatory and immune-protective cytokines), IL-4 (anti-inflammatory and immune-protective cytokine), IL-10 and TGF-β (anti-inflammatory and immune-suppressive cytokines). Murine lymphocytes were treated with TIG for 48 and 96 h at concentrations reflecting its plasma levels obtained in vivo at therapeutic doses, and at 10-fold lower concentrations. It was found that TIG neither affected substantially the percentage and absolute counts of entire CD4+ and CD8+ T cell populations nor influenced the Foxp3+CD25+CD4+ regulatory/suppressive T cell subset. Furthermore, the percentages of IL-4-, IL-10-, IL-17- and TGF-β-producing CD4+ T cells were not altered following the exposure to TIG. Similarly, TIG did not influence IFN-γ production by CD8+ T cells. Thus, with respect to the parameters evaluated, TIG does not seem to exert immune-suppressive and anti-inflammatory effects.

Go to article

Authors and Affiliations

A. Jasiecka-Mikołajczyk
J.J. Jaroszewski
T. Maślanka
Download PDF Download RIS Download Bibtex

Abstract

The aim of this study was to determine the influence of feed on the pharmacokinetics of flumequine (FLU) administered to broiler chickens as follows: directly into the crop (10 mg/kg of BW) of fasted (group I/control) and non-fasted chickens (group II), or administered continu- ously with drinking water (1 g/L for 72 h) and with unlimited access to feed (group III). Plasma concentration of FLU was determined by high-performance liquid chromatography with fluo- rescence detection. In group II, a significant decrease in the maximum concentration (Cmax = 2.13±0.7 μg/mL) and the area under the concentration curve from zero to infinity (AUC0→∞ = 7.47±2.41 μg·h/mL) was noted as compared to the control group (Cmax = 4.11±1.68 μg/mL and AUC0→∞ = 18.17±6.85 μg·h/mL, respectively). In group III, the decrease in AUC was signifi- cant only in the first 3 hours (AUC0→3 = 5.02±1.34 μg·h/mL) as compared to the control group (AUC0→3 = 7.79±3.29 μg·h/mL). The results indicate that feed reduced the bioavailability of FLU from the gastrointestinal tract by at least 50% after the administration of a single oral dose. However, continuous administration of FLU with drinking water could compensate for the feed-induced decrease in absorption after single oral dose.

Go to article

Authors and Affiliations

H. Madej-Śmiechowska
A. Orzoł
A. Jasiecka-Mikołajczyk
H. Ziółkowski
J.J. Jaroszewski

This page uses 'cookies'. Learn more