Journal of Mucosal Immunology Research
Open Access

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)
  • Editorial   
  • J Mucosal Immunol Res; 2022, Vol 6(1): 135
  • DOI: 10.4172/jmir.1000135

Method of Murine dendritic cell Cultures for Proteomics Experiments

Christian Villiers*
Department of Mucosal immunology and Biology Cente, University of Paris-Saclay, France
*Corresponding Author: Christian Villiers, Department of Mucosal immunology and Biology Cente, University of Paris-Saclay, France

Received: 06-Jan-2022 / Manuscript No. jmir-22-52531 / Editor assigned: 01-Jan-1970 / PreQC No. jmir-22-52531 / Reviewed: 01-Jan-1970 / QC No. jmir-22-52531 / Revised: 01-Jan-1970 / Manuscript No. jmir-22-52531 / Accepted Date: 21-Jan-2022 / Published Date: 28-Jan-2022 DOI: 10.4172/jmir.1000135

Editorial

The Dendritic cells are known to be initiated by a wide reach of microbial items, prompting cytokine creation and expanded degrees of film makers like major histocompatibility complex class II particles. Such actuated dendritic cells have the ability to actuate credulous T cells. In the current review we exhibited that juvenile dendritic cells emit both the YM1 lectin and lipocalin-2 [1]. By testing the ligands of these two proteins, chitosan and side rophores, separately, we likewise illustrated that chitosan, a debasement result of different parasitic and protozoa cell dividers, incites an actuation of dendritic cells at the layer level, as shown by the up-guideline of layer proteins like class II atoms, CD80 and CD86 through a TLR4-subordinate component, be that as it may, can’t actuate cytokine creation. This prompted the development of enacted dendritic cells unfit to invigorate T cells. In any case, stimulation with other microbial items conquered this incomplete enactment and re-established the limit of these actuated dendritic cells to animate T cells.

Moreover, progressive excitement with chitosan and afterward by lipopolysaccharide instigated a dose dependent change in the cytokinic IL-12/IL-10 equilibrium created by the dendritic cells. Atomic and Cellular Proteomics 8:1252-1264, 2009. Suitable detecting of microorganisms is a basic advance in sending off an efficient resistant reaction [2]. The resistant framework has developed to expand a perplexing arrangement of icrobe reconnaissance in which particular particles (called microbe acknowledgment receptors) and particular cell types (for example dendritic cells) assume significant parts. Dendritic cells (DCs) are very powerful antigen-introducing cells.

Upon feeling by risk signals, including signals created by outside microbes yet additionally flags got from unusual cells, DCs go through actuation/development to a functioning antigen-introducing aggregate. This aggregate incorporates move to the cell surface of antigen-loaded MHC class II particles (likewise called signal 1), up- uideline of costimulatory layer proteins, for example, CD80 and CD86 (additionally called signal 2), and emission of pro inflammatory cytokines, for example, TNF, IL-6, or IL-12 (too called signal 3)[3].

Ongoing proof has shown that each of the three signals are expected to incite T lymphocyte enactment. Most peril signals are detected through receptors having a place to the Toll-like receptor (TLR) family. These receptors can tie a wide scope of ligands, and the vast majority of the realized ligands are bacterial items (for example glycolipids and lipopolysaccharides) or viral items (for example twofold abandoned RNA). In any case, a couple endogenous TLR ligands, for example, anionic polysaccharides , are additionally known and may clarify part of the conduct of dendritic cells toward endogenous initiation signals. In any case, little is known about the risk signals coming from eukaryotic microorganisms (for example growths, protozoa, and so on) that are detected by dendritic cells. TLRs have been involved in the detecting of contagious polysaccharides by monocytes and dendritic cells . Be that as it may, there are not very many models in which the TLR associated with parasitic polysaccharide acknowledgment has been described.

Besides there is by all accounts a few pathways for polysaccharide detecting, prompting different DC aggregates. Moreover detecting of peril flags now and then includes atoms other than the ligand and the TLR as shown by the complex detecting of LPS in which protein arbiters, for example, CD14 and MD2 are additionally embroiled. To acquire bits of knowledge into potential harm detecting by DC, we chose to perform proteomics investigates proteins emitted by juvenile dendritic cells to search for discharged proteins restricting putative peril signals. We found proteins ready to tie bacterial siderophores and chitosans and further portrayed the impacts of these ligands on dendritic cells. Murine dendritic cells were delivered from bone marrow ancestors C57BL/6 mice were bought from Charles River. DCs were created from bone narrow as depicted already (16, 17). Momentarily bone marrow cells were detached by flushing from the femurs [4]. Erythrocytes and GR1-positive cells were eliminated by attractive cell arranging.

The leftover contrarily arranged cells were re suspended at 5 105 cells/ml in complete Iscove’s altered Dulbecco’s medium enhanced with 1% granulocyte/monocyte state animating component transfected J558 cell line supernatant (this cell line was a liberal gift, 40 mg/ml mouse recombinant FLT-3L, and 5 mg/ml mouse recombinant IL-6. At day 3, the cell supernatant was taken out, and the cells were re suspended in similar conditions [5]. From day 6 to day 11, IL-6 was eliminated, and FLT3-L was decreased to 20 mg/ml. At day 11, the bone marrow cells are separated into DCs and prepared for the different tests.

Dendritic cells (DC) are the most efficient group of antigen-presenting cells. As such, DC is highly specialized in the detection and phagocytosis of pathogens, the processing of antigens as well as stimulation and inflammatory signals in order to induce adequate T cell responses. The potential of DC to define the quality and extent of an adaptive immune response has attracted major interest in vaccine science, DC being key targets to fight infectious as well as cancer diseases [6].

Dendritic cells detect microbial ligands via Pattern Recognition Receptors (PRR) such as Toll-like Receptors. Upon encounter with microbes, DCs are strongly activated, characterized by the upregulation of co-stimulatory molecules (CD40, CD80, and CD86) and the production of cytokines and chemokines. In order to monitor and react efficiently to a pathogenic challenge, DC forms a complex and heterogeneous network in the organism. Several DC subsets have been described in both humans and mice, the latter being the animal model preferentially used and most accessible in the field. Migratory DC capture pathogens at the site of infection and rapidly reach the nearest draining lymph node for antigen presentation. Conventional tissue-resident DC (cDC) act as sentinels in secondary lymphoid organs and other tissues for antigen capture and presentation in situ. Other inflammatory DC may differentiate from blood-derived monocytes and infiltrate secondary organs and tissue during infection or inflammatory response. DC types may be further subdivided into different subsets and are identified according to the expression of surface markers. For instance in the mouse spleen, DC subtypes include plasmacytoid DC (B220+ CD11cint GR1−), monocyte-derived DC (MoDC; B220− CD11cint GR1±), and cDC (B220− CD11chigh GR1−). The latter are commonly divided into CD8α+ (CD11chigh, B220−, DEC205+, CD24high, CD11b−) and CD8α− (CD11chigh, B220−, DEC205−, CD24low, CD11b+, CD172+, CD4±) cDC subsets [7]. Interestingly, several lines of evidence support the notion of division of labor and cross-talk within the DC network; altogether, DC subsets display differences in the capacity to monitor tissue or circulate, the expression of PRRs, the production of cytokines, as well as antigen uptake and presentation mechanisms.

Further to the inherent complexity and heterogeneity of the DC system, a number of technical challenges have set a bottleneck to advances in DC research. First is the natural scarcity of DC in vivo, which not only reflects their functional potency, but also is a major limitation on the cellular material available for experimentation. Secondly, isolated splenic cDC show dramatic activation and apoptosis in culture, clearly detectable after a few hours of incubation. This greatly hampers experimental settings whenever relatively large quantities of cells or long incubation times are required [8].

In contrast to the B-cell and T cell fields, there is not as yet a DC line thoroughly characterized and widely accepted for in vitro research. In the mouse model, the DC culture system that has been widely used is Bone Marrow-derived DCs (BMDC), based on the differentiation of DC by treatment of BM progenitors with GM-CSF. More recently, BMDC are also generated using Flt3L, obtaining a mixture of equivalents to both CD8α+ and CD8α− cDC subsets and pDC. In human DC research, DC is similarly derived from the culture of peripheral blood mononuclear cells or CD14+ monocytes with GM-CSF/IL-4 (the MoDC system; Inaba et al., 2009). In both the mouse BMDC and the human MoDC systems, DC differentiation is driven in vitro, during 6–10 days, and is often followed by LPS treatment overnight to “mature” DC. These methods provide large quantities of DC, but require repetitive sacrifice of mice or human blood sampling, and are relatively tedious and time-consuming, as compared to the use of immortalized cell lines.

A limited number of DC lines have been described. These include the D1 cells, a growth factor-dependent immature DC line derived from mouse spleen DC, which can be “matured” with LPS [9]. The generation of murine DC lines based on oncogene-driven immortalization has also been reported, including the SRDC line, the SVDC line, and the DC 2.4 cell line. In humans, DC lines can be generated from the culture of leukemic DC found in the blood of acute myeloid leukemia patients. Other related human and murine model cell lines used in the DC field are Raw264.7 and J774 in mice, and THP-1, HL-60 and MUTZ-3 lines in humans. Some of the issues generally encountered with these DC lines or model cell lines are the requirement of particular growth factors or conditions to maintain cultures, as well as concerns over their equivalence to natural DC counterparts in vivo. In the light of the technical difficulties encountered in the study of DC biology, DC lines that retain the major functions of DC (further reflecting different subsets) and that are easily maintained in culture are still long sought.

In recent years, we developed a transgenic mouse expressing the SV40LgT oncogene (with an eGFP reporter) under the CD11c promoter, as a model system for histolytic disorders such as severe forms of multisystem Langerhans cells histiocytosis (Steiner et al., 2008). These mice indeed display DC tumor genesis, mainly in the spleen and liver, which affects in particular the CD8α cDC subset. In addition to the relevance to histiocytosis, using this model, it has been possible to derive several murine DC lines, originating from CD8α DC tumors primarily in spleen (therefore termed Muted for “murine tumor”). Importantly, DC tumor cells are not indefinitely viable directly ex vivo but can undergo immortalization in vitro. We now present the derivation procedure used to generate these immortalized MutuDC lines, followed by their thorough characterization by direct comparison to WT splenic cDC. We validate that MutuDC lines have retained the major features characteristic of their natural counterpart, the normal CD8α cDC subset. These include the response to particular TLR-Ls such as CpG (TLR9-L) and PolyIC (TLR3-L) but not R-848 (TLR7-L), IL-12 secretion and antigen cross-presentation capacity [10]. We furthermore show that the MutuDC lines may be modified by lentiviral transduction or by crossing the CD11c:SV40LgT-transgenic mice to the genetic background of interest to obtain genetically modified MutuDC lines. Finally, we discuss that the ease of culture and manipulation of the MutuDC lines render them a potent auxiliary tool

Acknowledgment:

None

Conflict of Interest:  

None

References

  1. Caminschi I, Proietto AI, Ahmet F, Kitsoulis S, Shin The J, et al. (2008) The dendritic cell subtype-restricted C-type lectin Clec9A is a target for vaccine enhancement. Blood 112:3264-3273.
  2. Indexed at, Google Scholar, Crossref

  3. Cox J, Mann M (2008) Max Quant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol 26:1367-1372.
  4. Indexed at, Google Scholar, Crossref

  5. Hochrein H, Shortman K, Vremec D, Scott B, Hertzog P, et al. (2001) Differential production of IL-12, IFN-alpha, and IFN-gamma by mouse dendritic cell subsets. J Immunol 166:5448-5455.
  6. Indexed at, Google Scholar, Crossref

  7. Inaba K, Swiggard WJ, Steinman RM, Romani N, Schuler G, et al. (2009) Isolation of dendritic cells. Curr Protoc Immunol 3: 3-7.
  8. Indexed at, Google Scholar, Crossref

  9. Merad M, Manz MG (2009) Dendritic cell homeostasis. Blood 113:3418-3427.
  10. Indexed at, Goggle Scholar, Crossref

  11. Naik SH, Metcalf D, Van Nieuwenhuijze A, Wicks I, Wu L, et al. (2006) Intrasplenic steady-state dendritic cell precursors that are distinct from monocytes. Nat Immunol 7:663-671.
  12. Indexed at, Google Scholar, Crossref

  13. Napolitani G, Rinaldi A, Bertoni F, Sallusto F, Lanzavecchia A (2005) Selected Toll-like receptor agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells. Nat Immunol 6:769-776.
  14. Indexed at, Google Scholar, Crossref

  15. Reis e Sousa C (2004) Toll-like receptors and dendritic cells: for whom the bug tolls. Semin Immunol 16: 27-34.
  16. Indexed at, Google Scholar, Crossref

  17. Shortman K, Liu YJ (2002) Mouse and human dendritic cell subtypes. Nat Rev Immunol 2:151-161.
  18. Indexed at, Google Scholar, Crossref

  19. RM (2012) Steinman Decisions About Dendritic Cells: Past, Present, and Future Ann. Rev Immunol 30:1-22.
  20. Indexed at, Google Scholar, Crossref

  21. Shortman K, Naik SH (2007) Steady-state and inflammatory dendritic-cell development. Nat Rev Immunol 7:3010-3019.
  22. Indexed at, Google Scholar, Crossref

Citation: Villiers C (2022) Specialist Based Demonstrating of Host-Microorganism Frameworks: The Triumphs. J Mucosal Immunol Res 6: 135. DOI: 10.4172/jmir.1000135

Copyright: © 2022 Villiers C. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Top