Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 Despite extensive research over the past several decades, the etiological agents of sarcoidosis remain unknown. Numerous potential etiological agents have been identified and the most recent hypothesis suggests that host-microbe interaction and genetic fa + 2161 word(s) 2161 2020-04-15 09:47:32 |
2 Despite extensive research over the past several decades, the etiological agents of sarcoidosis remain unknown. Numerous potential etiological agents have been identified and the most recent hypothesis suggests that host-microbe interaction and genetic fa Meta information modification 2161 2020-04-20 07:23:36 | |
3 Despite extensive research over the past several decades, the etiological agents of sarcoidosis remain unknown. Numerous potential etiological agents have been identified and the most recent hypothesis suggests that host-microbe interaction and genetic fa Meta information modification 2161 2020-04-20 07:24:25 | |
4 format change + 24 word(s) 2185 2020-04-24 09:43:56 | |
5 Added Immunopathogenesis of sarcoidosis + 575 word(s) 2760 2020-05-11 02:05:43 | |
6 format change -1483 word(s) 1277 2020-10-28 04:49:14 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Jain, R.; Yadav, D.; Puranik, N.; Guleria, R.; Jin, J. Sarcoidosis. Encyclopedia. Available online: https://encyclopedia.pub/entry/609 (accessed on 19 April 2024).
Jain R, Yadav D, Puranik N, Guleria R, Jin J. Sarcoidosis. Encyclopedia. Available at: https://encyclopedia.pub/entry/609. Accessed April 19, 2024.
Jain, Rashi, Dhananjay Yadav, Nidhi Puranik, Randeep Guleria, Jun-O Jin. "Sarcoidosis" Encyclopedia, https://encyclopedia.pub/entry/609 (accessed April 19, 2024).
Jain, R., Yadav, D., Puranik, N., Guleria, R., & Jin, J. (2020, April 20). Sarcoidosis. In Encyclopedia. https://encyclopedia.pub/entry/609
Jain, Rashi, et al. "Sarcoidosis." Encyclopedia. Web. 20 April, 2020.
Sarcoidosis
Edit

Sarcoidosis is a multisystem granulomatous disease with nonspecific clinical manifestations that commonly a ects the pulmonary system and other organs including the eyes, skin, liver, spleen, and lymph nodes. Sarcoidosis usually presents with persistent dry cough, eye and skin manifestations, weight loss, fatigue, night sweats, and erythema nodosum.

sarcoidosis biomarkers diagnosis cause management

1. Introduction

Sarcoidosis is not influenced by sex or age, although it is more common in adults (< 50 years) of African-American or Scandinavians decent. Diagnosis can be dicult because of nonspecific symptoms and can only be verified following histopathological examination. Various factors, including infection, genetic predisposition, and environmental factors, are involved in the pathology of sarcoidosis. Exposures to insecticides, herbicides, bioaerosols, and agricultural employment are also associated with an increased risk for sarcoidosis. Due to its unknown etiology, early diagnosis and detection are dicult; however, the advent of advanced technologies, such as endobronchial ultrasound-guided biopsy, high-resolution computed tomography, magnetic resonance imaging, and 18F-fluorodeoxyglucose positron emission tomography has improved our ability to reliably diagnose this condition and accurately forecast its prognosis. In a recent review published in the Journal of Clinical Medicine (https://doi.org/10.3390/jcm9041081) discusses the causes and clinical features of sarcoidosis, and the improvements made in its prognosis, therapeutic management, and the recent discovery of potential biomarkers associated with the diagnostic assay used for sarcoidosis confirmation.

2. The cause of Sarcoidosis 

The exact cause of sarcoidosis is not known. Many researchers have hypothesized the role of genetic susceptibility, environmental factors, putative antigens, and autoimmunity in the development of this disease, but no single cause has been identified to date.

2.1. Genetic Factors

Various studies suggest that genetic factors could play a crucial role in establishing the risk and clinical development of sarcoidosis[1]. Eleven sarcoidosis risk loci (BTNL2, HLA-B, HLA-DPB1, ANXA11, IL23R, SH2B3/ATXN2, IL12B, NFKB1/MANBA, FAM177B, chromosome 11q13.1, and RAB23) have been identified to date[2]. A previous study reported that familial sarcoidosis occurred in 17% of African-Americans[3], while only 1.4% of Spanish people exhibited this same risk[4]. According to A Case-Control Etiologic Sarcoidosis Study (ACCESS) the chance of developing sarcoidosis is five-fold among sib­lings[5]. Monozygotic siblings with sarcoidosis had an 80-fold higher risk of developing the condition, although the estimated risk of developing sarcoidosis in dizygotic twins was only seven-fold[6].

Genome wide association studies have demonstrated that several HLA and non-HLA alleles are associated with the development of this disease[7]. HLA-DRB1*0301/ DQB1*0201[8], transforming growth factor β (TGF-β)[9], tumor necrosis factor α (TNF-α)[10], and Toll-like receptor 4 (TLR-4)[11] are all considered significant indicators for susceptibility to sarcoidosis[12][13].

2.2. Environmental Risk Factors

Various environmental factors, including exposure to wood stoves, soil, tree pollen, inorganic particulates, insecticides, and nanoparticles, have been associated with an increased risk for developing sarcoidosis. In addition to these factors, some workers, such as those involved in hardware, gardening materials, building supplies, and metal work as well as ship servicemen in the navy, fire workers, and educators, are prone to sarcoidosis[14][15][16]. It has been suggested that silica exposure also triggers the risk of sarcoidosis[17]. The underlying hypothesis for this association is that the environment is an important risk factor for the development of sarcoidosis, which has been further strengthened by reports that US World Trade Center workers exposed to the crash debris, in particular firefighters; all experienced an increased risk for developing sarcoidosis or “sarcoid-like” disease[18].

2.3. Infection

In addition to all of the factors mentioned above, infectious agents such as mycobacteria, have been suggested to be associated with the development of sarcoidosis, because the production of granulomas is a key factor in the immune defense response against these agents. Studies have identified numerous microbial agents as a potential eliciting agents of the immune response in sarcoidosis including Leptospira species, Mycoplasma species, herpes virus, retrovirus, Chlamydia pneumoniae, Borrelia burgdorferi, [19] Pneumocystis jirovecii[20], Mycobacterium (M.tb)[21], and Propionibacterium species[22]. Isolation of M.tb. DNA, from tissue specimens collected from sarcoidosis patients, with sequences specific to mycobacterial proteins, such as ESAT-6, Kat G, and SoD A, illustrate that Mycobacterium is the strongest candidate for infection-mediated sarcoidosis[23][24][25]. It has been reported that patients treated with interferon α therapy for hepatitis C infection developed sarcoidosis[26][27]. A few studies have suggested that hepatitis C infection on its own could increase the risk of developing sarcoidosis. However, it seems more likely that therapy with interferon α increases interferon-γ and interleukin-2 expression, stimulating granuloma formation and thus sarcoidosis[28][29].

2.4. Autoimmunity

Autoimmunity has not been studied as extensively but given the underlying pathological mechanism of sarcoidosis there is certainly potential for these conditions to play a contributing role in disease development. Although no disease-specific auto-antibodies have been observed, it has been shown that the major histocompatibility complex (MHC) class II molecules on antigen-presenting cells possess an autoantigen that is recognized by the T-cell receptor (TCR) of the responding T-cells in sarcoidosis patients[30][31]. Vimentin-derived peptides are the most plausible candidate for the activation of both T-cells and B-cells in the lung[32]. Autoimmunity presents a as a novel spectrum for sarcoidosis immunopathogenesis and may help elucidate sarcoid etiology[33][34][35].

Another important aspect of autoimmunity is the imbalanced gut microbiome. Gianchecchi et al. reported the associations between the presence of microbiome dysbiosis and the development of autoimmune conditions[36]. Sarcoidosis overlaps with other autoimmune diseases, including rheumatoid arthritis, autoimmune thyroid disease, Sjogren’s syndrome, and ankylosing spondylitis[37]. The role of the microbiota in these autoimmune diseases has been evaluated in previous studies and been shown to lay a significant role in their pathogenesis[38]; thus, study of the microbiome of sarcoidosis patients and its correlation with other diseases could open new avenues for investigating the underlying causes of this disease[39][40].

3. Immunopathogenesis of sarcoidosis

Many etiological agents, including infectious microbes, as well as organic and inorganic compounds, contribute to the development of sarcoidosis. These antigens are first cleared by the immune system, but this is not infallible and some undegraded antigens may remain in the cells, which can initiate an immune feedback loop. In response to this feedback loop, the antigen-presenting cells (APCs), such as dendritic cells (DCs), alveolar macrophages (AMs), and alveolar epithelial cells, produce high levels of TNF-α, and secrete interleukins-12, -15, and -18, macrophage inflammatory protein-1 (MIP-1), monocyte chemoattractant protein-1 (MCP-1), and granulocyte macrophage colony-stimulating factor (GM-CSF)[41]. These APCs also present antigens to CD4+ T-cells initiating granuloma construction, a critical feature of sarcoidosis. The growth of these granulomas establishes the primary abnormality in most cases of sarcoidosis. Sarcoid granulomas are ordered, structured masses comprised of macrophages and their derivatives, epithelioid cells, giant cells, and T-cells.

Activated CD4+ T-cells can differentiate into two distinct subsets, namely, T helper 1 (Th1) and T helper 2 (Th2) cells, based on their cytokines profile. Th1 cells predominantly secrete interleukin-2 (IL-2) and interferon-gamma (IFN-γ), while IL-4 and IL-13 are the major secretions of Th2 cells. Resolution or maintenance of granuloma is determined by the proportion of Th1 and Th2 cells, respectively. Alveolar macrophages are activated in the Th2 milieu and stimulate fibroblast and collagen proliferation culminating in progressive fibrosis[42].

Incapacitation of Tregs is also a key feature of granuloma maintenance. It is presumed that infiltrating Tregs fail to reduce the exaggerated inflammatory response, thereby contributing to granuloma persistence and integrity. Tregs also release transforming growth factor β (TGF-β) that may contribute to fibrosis and granuloma organization[43].

Th17 and Th17.1 cells have only recently been linked to the pathogenesis of sarcoidosis[44]. These cells are recruited to the disease site and are involved in the construction of the granuloma. The balance between Th17 and Treg cells is thought to be disrupted in sarcoidosis[45] and is an important factor in its prognosis[46]. The regulation of antigen processing, antigen presentation to the APCs, and cytokine release are all controlled through genetic elements and may link the various causal factors of sarcoidosis together[47][48][49].

References

  1. Johan Grunewald; Paolo Spagnolo; Jan Wahlström; Anders Eklund; Immunogenetics of Disease-Causing Inflammation in Sarcoidosis. Clinical Reviews in Allergy & Immunology 2015, 49, 19-35, 10.1007/s12016-015-8477-8.
  2. Annegret Fischer; David Ellinghaus; Marcel Nutsua; Sylvia Hofmann; Courtney G. Montgomery; Michael C. Iannuzzi; Benjamin A. Rybicki; Martin Petrek; Frantisek Mrazek; Stefan Pabst; et al.Christian GrohéJohan GrunewaldMarcus RonningerAnders EklundLeonid PadyukovVioleta Mihailovic-VucinicDragana JovanovicMartina SterclovaJiri HomolkaMarkus M. NöthenStefan HermsChristian GiegerKonstantin StrauchJuliane WinkelmannBernhard O. BoehmStephan BrandCarsten BüningManfred SchürmannEva EllinghausHansjörg BaurechtWolfgang LiebAlmut NebelJoachim Müller-QuernheimAndre FrankeStefan Schreiber Identification of Immune-Relevant Factors Conferring Sarcoidosis Genetic Risk. American Journal of Respiratory and Critical Care Medicine 2015, 192, 727-736, 10.1164/rccm.201503-0418oc.
  3. Benjamin A. Rybicki; Michael C. Iannuzzi; Epidemiology of Sarcoidosis: Recent Advances and Future Prospects. Seminars in Respiratory and Critical Care Medicine 2007, 28, 22-35, 10.1055/s-2007-970331.
  4. Estrella Fernandez Fabrellas; Epidemiología de la sarcoidosis. Archivos de Bronconeumología 2009, 43, 92-100, 10.1157/13098420.
  5. Benjamin A. Rybicki; Michael C. Iannuzzi; Margaret M. Frederick; Bruce W. Thompson; Milton D. Rossman; Eddy A. Bresnitz; Michael L. Terrin; David R. Moller; Juliana Barnard; Robert P. Baughman; et al.Louis DePaloGary HunninghakeCarol JohnsMarc A. JudsonGenell L. KnatterudGeoffrey McLennanLee S. NewmanDavid L. RabinCecile RoseAlvin S. TeirsteinSteven E. WeinbergerHenry YeagerReuben Cherniackthe ACCESS Research Group Familial Aggregation of Sarcoidosis. American Journal of Respiratory and Critical Care Medicine 2001, 164, 2085-2091, 10.1164/ajrccm.164.11.2106001.
  6. Asger Sverrild; Vibeke Backer; Kirsten Ohm Kyvik; Jaakko Kaprio; Nils Milman; Claus Bo Svendsen; Simon Francis Thomsen; Heredity in sarcoidosis: a registry-based twin study. Thorax 2008, 63, 894-896, 10.1136/thx.2007.094060.
  7. Manfred Schürmann; Philipp Reichel; Bertram Müller-Myhsok; Max Schlaak; Joachim Müller-Quernheim; Eberhard Schwinger; Results from a Genome-wide Search for Predisposing Genes in Sarcoidosis. American Journal of Respiratory and Critical Care Medicine 2001, 164, 840-846, 10.1164/ajrccm.164.5.2007056.
  8. Mami Ishihara; Shigeaki Ohno; Takako Ishida; Hitoshi Ando; Taeko Naruse; Yoshisuke Nose; Hidetoshi Inoko; Molecular genetic studies of HLA class II alleles in sarcoidosis. Tissue Antigens 1994, 43, 238-241, 10.1111/j.1399-0039.1994.tb02331.x.
  9. Stefan Pabst; T. Franken; J. Schönau; S. Stier; G. Nickenig; R. Meyer; D. Skowasch; C. Grohé; Transforming growth factor- gene polymorphisms in different phenotypes of sarcoidosis. European Respiratory Journal 2010, 38, 169-175, 10.1183/09031936.00120410.
  10. S. Sharma; B. Ghosh; Association of TNF polymorphisms with sarcoidosis, its prognosis and tumour necrosis factor (TNF)-α levels in Asian Indians. Clinical & Experimental Immunology 2007, 151, 251-259, 10.1111/j.1365-2249.2007.03564.x.
  11. S. Pabst; G. Baumgarten; A. Stremmel; M. Lennarz; P. Knüfermann; A. Gillissen; H. Vetter; C Grohé; Toll-like receptor (TLR) 4 polymorphisms are associated with a chronic course of sarcoidosis. Clinical & Experimental Immunology 2006, 143, 420-426, 10.1111/j.1365-2249.2006.03008.x.
  12. Grunewald, J. Role of genetics in susceptibility and outcome of sarcoidosis. Semin. Respir. Crit. Care Med. 2010, 31, 380–389.
  13. Iannuzzi, M.C. Genetics of sarcoidosis. Semin. Respir. Crit. Care Med. 2007, 28, 15–21.
  14. Kucera, G.P.; Rybicki, B.A.; Kirkey, K.L.; Coon, S.W.; Major, M.L.; Maliarik, M.J.; Iannuzzi, M.C. Occupational risk factors for sarcoidosis in african-american siblings. Chest 2003, 123, 1527–1535.
  15. Lee S. Newman; Cecile S. Rose; Eddy A. Bresnitz; Milton Rossman; Juliana Barnard; Margaret Frederick; Michael L. Terrin; Steven E. Weinberger; David R. Moller; Geoffrey McLennan; et al.Gary HunninghakeLouis DePaloRobert P. BaughmanMichael C. IannuzziMarc A. JudsonGenell L. KnatterudBruce W. ThompsonAlvin S TeirsteinHenry YeagerCarol J. JohnsDavid L. RabinBenjamin A. RybickiReuben Cherniack A Case Control Etiologic Study of Sarcoidosis. American Journal of Respiratory and Critical Care Medicine 2004, 170, 1324-1330, 10.1164/rccm.200402-249oc.
  16. Kira L. Newman; Lee S. Newman; Occupational causes of sarcoidosis. Current Opinion in Allergy and Clinical Immunology 2012, 12, 145-150, 10.1097/aci.0b013e3283515173.
  17. Per Vihlborg; Ing-Liss Bryngelsson; Lena Andersson; Pål Graff; Risk of sarcoidosis and seropositive rheumatoid arthritis from occupational silica exposure in Swedish iron foundries: a retrospective cohort study. BMJ Open 2017, 7, e016839, 10.1136/bmjopen-2017-016839.
  18. Gabriel Izbicki; Robert Chavko; Gisela I. Banauch; Michael D. Weiden; Kenneth I. Berger; Thomas K. Aldrich; Charles B. Hall; Kerry J. Kelly; David J. Prezant; World Trade Center “Sarcoid-Like” Granulomatous Pulmonary Disease in New York City Fire Department Rescue Workers. Chest 2007, 131, 1414-1423, 10.1378/chest.06-2114.
  19. L.S. Newman; Aetiologies of sarcoidosis. Sarcoidosis 2005, 32, 23-48, 10.1183/1025448x.00032003.
  20. S. Vidal; C. De La Horra; J. Martin; M. A. Montes‐Cano; E. Rodríguez; N. Respaldiza; J.M. Varela; Francisco J. Medrano; Enrique Calderón; Pneumocystis jirovecii colonisation in patients with interstitial lung disease. Clinical Microbiology and Infection 2006, 12, 231-235, 10.1111/j.1469-0691.2005.01337.x.
  21. Wonder P. Drake; Lee S Newman; Mycobacterial antigens may be important in sarcoidosis pathogenesis. Current Opinion in Pulmonary Medicine 2006, 12, 359-363, 10.1097/01.mcp.0000239554.01068.94.
  22. Ikuo Ishige; Yoshinobu Eishi; Tamiko Takemura; Intetsu Kobayashi; Koh Nakata; Isao Tanaka; Sakae Nagaoka; Kazuro Iwai; Kunitomo Watanabe; Touichiro Takizawa; et al.Morio Koike Propionibacterium acnes is the most common bacterium commensal in peripheral lung tissue and mediastinal lymph nodes from subjects without sarcoidosis.. Sarcoidosis, vasculitis, and diffuse lung diseases : official journal of WASOG 2005, 22, 33-42.
  23. Shannon S Allen; Whitney Evans; James Carlisle; Rana Hajizadeh; Michele Nadaf; Bryan E Shepherd; David T Pride; Joyce E Johnson; Wonder P. Drake; Superoxide dismutase A antigens derived from molecular analysis of sarcoidosis granulomas elicit systemic Th-1 immune responses. Respiratory Research 2008, 9, 36-36, 10.1186/1465-9921-9-36.
  24. Song, Z.; Marzilli, L.; Greenlee, B.M.; Chen, E.S.; Silver, R.F.; Askin, F.B.; Teirstein, A.S.; Zhang, Y.; Cotter, R.J.; Moller, D.R. Mycobacterial catalase-peroxidase is a tissue antigen and target of the adaptive immune response in systemic sarcoidosis. J. Exp. Med. 2005, 201, 755–767.
  25. Drake,W.P.; Dhason, M.S.; Nadaf, M.; Shepherd, B.E.; Vadivelu, S.; Hajizadeh, R.; Newman, L.S.; Kalams, S.A. Cellular recognition of mycobacterium tuberculosis esat-6 and katg peptides in systemic sarcoidosis. Infect. Immun. 2007, 75, 527–530.
  26. Atsushi Hirano; Mikio Kataoka; Yasunari Nakata; Katsuyuki Takeda; Takayuki Kamao; Junichi Hiramatsu; Goro Kimura; Yasushi Tanimoto; Arihiko Kanehiro; Mitsune Tanimoto; et al. Sarcoidosis occurring after interferon-alpha therapy for chronic hepatitis C: Report of two cases. Respirology 2005, 10, 529-534, 10.1111/j.1440-1843.2005.00745.x.
  27. Hasan Salameh; Remi Trien; Chad J. Cooper; David Paez; Edgardo Colon; Shajeea Ajmal; Interferon-alpha-induced sarcoidosis in a patient being treated for hepatitis C. American Journal of Case Reports 2014, 15, 235-238, 10.12659/ajcr.890180.
  28. Vadim Brjalin; Riina Salupere; Valentina Tefanova; Kaiu Prikk; Natalia Lapidus; Enn Jõeste.; Sarcoidosis and chronic hepatitis C: A case report. World Journal of Gastroenterology 2012, 18, 5816-5820, 10.3748/wjg.v18.i40.5816.
  29. Manuel Ramos-Casals; Juan Mañá; Norma Nardi; Pilar Brito-Zerón; Antoni Xaubet; José Maria Sánchez-Tapias; Ricard Cervera; Josep Font; Sarcoidosis in Patients With Chronic Hepatitis C Virus Infection. Medicine 2005, 84, 69-80, 10.1097/01.md.0000157577.69729.e6.
  30. Johan Grunewald; Ylva Kaiser; Mahyar Ostadkarampour; Natalia V. Rivera; Francesco Vezzi; Britta Lötstedt; Remi-André Olsen; Lina Sylwan; Sverker Lundin; Max Käller; et al.Tatiana SandalovaKerstin M. AhlgrenJan WahlströmAdnane AchourMarcus RonningerAnders Eklund T-cell receptor–HLA-DRB1 associations suggest specific antigens in pulmonary sarcoidosis. European Respiratory Journal 2015, 47, 898-909, 10.1183/13993003.01209-2015.
  31. Jan Wahlström; Jörn Dengjel; Ola Winqvist; Ira Targoff; Bengt Persson; Hüseyin Duyar; Hans-Georg Rammensee; Anders Eklund; Robert Weissert; Johan Grunewald; et al. Autoimmune T cell responses to antigenic peptides presented by bronchoalveolar lavage cell HLA-DR molecules in sarcoidosis. Clinical Immunology 2009, 133, 353-363, 10.1016/j.clim.2009.08.008.
  32. Gernot Zissel; Joachim Müller-Quernheim; Specific antigen(s) in sarcoidosis: a link to autoimmunity?. European Respiratory Journal 2016, 47, 707-709, 10.1183/13993003.01791-2015.
  33. Ylva Kaiser; Anders Eklund; Johan Grunewald; Moving target: shifting the focus to pulmonary sarcoidosis as an autoimmune spectrum disorder. European Respiratory Journal 2019, 54, 1802153, 10.1183/13993003.021532018.
  34. Anna Starshinova; Anna M. Malkova; Natalia Y. Basantsova; Yulia S. Zinchenko; Igor V. Kudryavtsev; Gennadiy A. Ershov; Lidia A. Soprun; Vera A. Mayevskaya; Leonid P. Churilov; Piotr K. Yablonskiy; et al. Sarcoidosis as an Autoimmune Disease. Frontiers in Immunology 2020, 10, 2933, 10.3389/fimmu.2019.02933.
  35. Anna Häggmark; Carl Hamsten; Emil Wiklundh; Cecilia Lindskog; Cecilia Mattsson; Eni Andersson; Ingrid E. Lundberg; Hans Grönlund; Jochen M. Schwenk; Anders Eklund; et al.Johan GrunewaldPeter Nilsson Proteomic Profiling Reveals Autoimmune Targets in Sarcoidosis. American Journal of Respiratory and Critical Care Medicine 2015, 191, 574-583, 10.1164/rccm.201407-1341oc.
  36. Elena Gianchecchi; Alessandra Fierabracci; Recent Advances on Microbiota Involvement in the Pathogenesis of Autoimmunity. International Journal of Molecular Sciences 2019, 20, 283, 10.3390/ijms20020283.
  37. Peter Korsten; Björn Tampe; Maximilian F. Konig; Elena Nikiphorou; Sarcoidosis and autoimmune diseases. Current Opinion in Pulmonary Medicine 2018, 24, 504-512, 10.1097/mcp.0000000000000500.
  38. Fengna Chu; Mingchao Shi; Yue Lang; Donghui Shen; Tao Jin; Jie Zhu; Li Cui; Gut Microbiota in Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis: Current Applications and Future Perspectives. Mediators of Inflammation 2018, 2018, 1-17, 10.1155/2018/8168717.
  39. André Becker; Giovanna Vella; Valentina Galata; Katharina Rentz; Christoph Beisswenger; Christian Herr; Jörn Walter; Sascha Tierling; Hortense Slevogt; Andreas Keller; et al.Robert Bals The composition of the pulmonary microbiota in sarcoidosis – an observational study. Respiratory Research 2019, 20, 46, 10.1186/s12931-019-1013-2.
  40. Pleiades T. Inaoka; Masato Shono; Mishio Kamada; J. Luis Espinoza; Host-microbe interactions in the pathogenesis and clinical course of sarcoidosis. Journal of Biomedical Science 2019, 26, 45, 10.1186/s12929-019-0537-6.
  41. Carlo Agostini; Fausto Adami; Gianpietro Semenzato; New pathogenetic insights into the sarcoid granuloma. Current Opinion in Rheumatology 2000, 12, 71-76, 10.1097/00002281-200001000-00012.
  42. Thomas A. Wynn; Luke Barron; Macrophages: Master Regulators of Inflammation and Fibrosis. Seminars in Liver Disease 2010, 30, 245-257, 10.1055/s-0030-1255354.
  43. Branton, M.H.; Kopp, J.B.; Tgf-beta and fibrosis. Infect 1999, 1, 1349–1365.
  44. Jelle R. Miedema; Ylva Kaiser; Caroline E. Broos; Marlies S. Wijsenbeek; Johan Grunewald; M. Kool; Th17-lineage cells in pulmonary sarcoidosis and Löfgren's syndrome: Friend or foe?. Journal of Autoimmunity 2018, 87, 82-96, 10.1016/j.jaut.2017.12.012.
  45. Hui Huang; Zhiwei Lu; Chunguo Jiang; Jia Liu; Yan Xun Wang; Zuojun Xu; Imbalance between Th17 and Regulatory T-Cells in Sarcoidosis. International Journal of Molecular Sciences 2013, 14, 21463-21473, 10.3390/ijms141121463.
  46. Yongzhe Liu; Lan Qiu; Yanxun Wang; Halimulati Aimurola; Yuyue Zhao; Shan Li; Zuojun Xu; The Circulating Treg/Th17 Cell Ratio Is Correlated with Relapse and Treatment Response in Pulmonary Sarcoidosis Patients after Corticosteroid Withdrawal. PLoS ONE 2016, 11, e0148207, 10.1371/journal.pone.0148207.
  47. Adrian Kelly; John Trowsdale; Genetics of antigen processing and presentation. Immunogenetics 2018, 71, 161-170, 10.1007/s00251-018-1082-2.
  48. Jamie A Lee; Robert S. Sinkovits; Dennis Mock; Eva L Rab; Jennifer Cai; Peng Yang; Brian Saunders; Robert C Hsueh; Sangdun Choi; Shankar Subramaniam; et al.Richard H. Scheuermann Components of the antigen processing and presentation pathway revealed by gene expression microarray analysis following B cell antigen receptor (BCR) stimulation. BMC Bioinformatics 2006, 7, 237-237, 10.1186/1471-2105-7-237.
  49. David Bennett; Elena Bargagli; Rosa Metella Refini; Paola Rottoli; New concepts in the pathogenesis of sarcoidosis.. Expert Review of Respiratory Medicine 2019, 13, 981-991, 10.1080/17476348.2019.1655401.
More
Information
Subjects: Pathology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 929
Revisions: 6 times (View History)
Update Date: 28 Oct 2020
1000/1000