Dersleri yüzünden oldukça stresli bir ruh haline sikiş hikayeleri bürünüp özel matematik dersinden önce rahatlayabilmek için amatör pornolar kendisini yatak odasına kapatan genç adam telefonundan porno resimleri açtığı porno filmini keyifle seyir ederek yatağını mobil porno okşar ruh dinlendirici olduğunu iddia ettikleri özel sex resim bir masaj salonunda çalışan genç masör hem sağlık hem de huzur sikiş için gelip masaj yaptıracak olan kadını gördüğünde porn nutku tutulur tüm gün boyu seksi lezbiyenleri sikiş dikizleyerek onları en savunmasız anlarında fotoğraflayan azılı erkek lavaboya geçerek fotoğraflara bakıp koca yarağını keyifle okşamaya başlar

GET THE APP

Journal of Oncology Research and Treatment - Tissue-Specific Differences in Mesenchymal Stromal Cells Influence Homing and Function

Journal of Oncology Research and Treatment
Open Access

Like us on:

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)
  • Commentary   
  • J Oncol Res Treat
  • DOI: 10.4172/aot.s1.1000001

Tissue-Specific Differences in Mesenchymal Stromal Cells Influence Homing and Function

Christen L. Ebens1*, Julia A. Riedl2 and Jakub Tolar2,3
1Department of Pediatrics, Division of Blood & Marrow Transplant and Cellular Therapy, University of Minnesota, Minneapolis, MN, USA
2Medical Scientist Training Program, University of Minnesota, Minneapolis, MN, USA
3Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
*Corresponding Author: Christen L. Ebens, Division of Blood & Marrow Transplant and Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA, Tel: 612-626-8094, Fax: 612-626-2815, Email: ebens012@umn.edu

Received: 25-Mar-2021 / Accepted Date: 08-Apr-2021 / Published Date: 15-Apr-2021 DOI: 10.4172/aot.s1.1000001

Abstract

Mesenchymal Stromal Cell(MSC) therapies to modulate inflammation and promote tissue regeneration in Recessive Dystrophic Epidermolysis Bullosa(RDEB) are under active investigation in clinical trials. However, optimal tissue source and fundamental mechanisms of clinical benefit of MSCs require additional investigation. Riedl et al.recently published “ABCB5+ dermal mesenchymal stromal cells with favorable skin homing and localimmunomodulation for Recessive Dystrophic Epidermolysis Bullosa treatment,” in Stem Cells [1] highlightingdifferences in homing of MSCs, impact on local macrophages, and distinct transcriptional profiles related to tissue oforigin (skin dermis versus bone marrow). Elucidation of these critical features and underlying mechanisms will aid intranslation of MSC cellular therapies to promote wound healing in RDEB.

Keywords: Mesenchymal Stromal Cell; Recessive DystrophicEpidermolysis Bullosa; Mesenchymal Stromal Cells; Bone Marrow

Commentary

Recessive Dystrophic Epidermolysis Bullosa(RDEB) is a genodermatosis characterized by exquisite mucocutaneous fragility. Affected individuals have biallelic mutations in COL7A1 resulting in absent or abnormal type VII collagen (C7). C7 is most abundantly produced by epithelial cells but can additionally be secreted by fibroblasts and mesenchymal stromal cells. C7 is critical to the mechanical integrity of the mucocutaneous barrier. In RDEB, minor trauma results in separation, or blistering, of skin at the dermalepidermal junction. Infants are born with blisters and erosions. Over a lifetime, individuals with RDEB experience repeated cycles of wounding and healing, inducing dermal fibrosis, contractures, and pseudosyndactyly of digits, esophageal strictures [1,2], and increased risk of lethal squamous cell carcinoma as early as the second decade of life [3]. Chronic pain and itch, high metabolic demand and malnutrition, micronutrient deficiencies, corneal abrasions, and anemia of chronic disease are common features in severe, generalized RDEB. RDEB is a disease of systemic inflammation without a cure. RDEB care consists exclusively of symptomatic management including avoidance of trauma; wound care; protective bandages; treatment of infections, pain and itch; and nutritional, micronutrient, and iron supplementation [4-7]. Translational research efforts in RDEB aim to provide more targeted therapies.

Regenerative medicine approaches to RDEB have focused on two main endpoints (1) increased functional C7 and (2) immunomodulation. For the former, active clinical trials include use of gene-corrected ex vivo skin grafts, topical and systemic agents for premature termination codon read through, and delivery of C7 topically. Intradermal injection of gene-corrected autologous fibroblasts to provide C7 demonstrated wound healing benefits and persistence of skin C7 for up to 12 months [8]. Mesenchymal stromal cells, initially pursued for C7 production capabilities, but have more recently gained interest for their immunomodulatory effects.

Mesenchymal stromal cells (MSCs) were first described as fibroblastic cells from the bone marrow capable of clonal production of bone [9]. With subsequent characterization, MSCs are now defined as cells of stromal origin, capable of self-renewal and trilineage chondrogenic, osteogenic and adipogenic differentiation [10]. Following successful restoration of C7 with allogeneic Bone Marrow Transplant(BMT) in a murine model of RDEB[11], BMT was instituted as a systemic therapy for human patients [12]. While patients demonstrated improvement in their blistering phenotype, C7 was not similarly increased as in the mice. Researchers hypothesized a portion of the clinical benefit could be attributable to Bone Marrow(BM)-derived MSCs in the donor inoculum during BMT. Indeed non-hematopoietic donor cells could be identified in the dermis following BMT [13]. To leverage this benefit, subsequent clinical protocols incorporated BM-MSCs, initially 3rd party and more recently donor-derived to avoid immune rejection of the cells [14]. At the same time, both intradermal injection[15,16] and intravenous [17,18] administration of allogeneic BM- MSCs for RDEB as monotherapy have demonstrated benefit with regard to improved wound healing, presumably by dampening local inflammation. Yet, are BM-MSCs the ideal MSC population for a disease process anchored in the skin?

With identification of MSCs in tissues beyond the BM, nomenclature evolved to reflect the unique characteristics of MSCs from difference niches [19]. At the same time, researchers began studying a unique subpopulation of MSCs resident in the skin dermis- ABCB5+DSCs (dermal MSCs). ABCB5, or ATP-binding cassette subfamily member 5, is a surface glycoprotein that identifies (and allows for purification of) a population of DSCs with immunosuppressive qualities, including expression of Programmed cell Death(PD)-1 to inhibit T-cell activation, evasion of immune rejection, and induction of tolerogenic regulatory T cells[20]. ABCB5+DSCs were shown upon neonatal systemic administration in a murine model of RDEB to prolong life and decrease infiltration of macrophages in the skin[21]. Further investigation in another murine model of nonhealing wounds (iron-overload) demonstrated ABCB5+ DSCs to influence immunosuppressive M2 over proinflammatory M1 skewing of macrophage differentiation through interleukin-1 receptor antagonist (IL-1RA) secretion [22]. These cells were subsequently characterized and manufactured for clinical trial use [23] in a variety of inflammatory conditions, including RDEB (Phase I/II trial results pending).

To better understand niche differences in MSC populations relevant to clinical use in RDEB, Riedl et al. [1] directly compared ABCB5+DSC and BM-MSC influence on macrophage cytokine production in co-cultures, skin homing in mice, and transcriptional profiles of the two MSC populations. Increased macrophage antiinflammatory IL-RA secretion had previously been demonstrated in co-culture with ABCB5+DSCs [22], but we showed greater IL-1RA secretion following direct co-culture compared to a Tran’s well system, suggesting a direct interaction between cell types as opposed to a paracrine effect. As anticipated, we found superior homing of ABCB5+DSCs to skin and wound engraftment 14 days after injection compared to BM-MSCs (2.7 increase, p<0.001). Transcriptional analysis revealed ABCB5+DSCs to have increased expression of 17 homeobox(Hox) genes, as well as Vascular Cell Adhesion Molecule(VCAM-1) and major Histocompatibility Complex(MHC) Class II protein, HLA-DPB1, compared to BM-MSCs.

Conclusion

The significance of differences between ABCB5+DSCs and BM-MSCs reported by Riedl et al.[1] requires furtherinvestigation but are intriguing considering the importance of Hoxgenes (particularly HOXA3) in coordination of wound healing,VCAM-1 in homing to the perivascular skin niche, and MHC Class IIin immune evasion. In aggregate, our investigations confirm superiorcutaneous anti-inflammatory benefits of ABCB5+DSCs compared toBM-MSCs in vitro and in pre-clinical murine studies, suggestmechanism for such benefits, and support further investigations ofABCB5+DSCs in skin diseases including RDEB.

Conflict of Interest

The authors have no potential conflicts of interest to disclose.

References

  1. Riedl J, Leonard MP, Eide C, Kluth MA, Ganss C, et al. (2021) ABCB5+dermal mesenchymal stromal cells with favorable skin homing and local immunomodulation for recessive dystrophic Epidermolysis Bullosa treatment. Stem Cells.
  2. Feinstein JA, Jambal P, Peoples K, Lucky AW, Khuu P, et al. (2019) Assessment of the timing of milestone clinical events in patients with epidermolysis bullosa from north america. JAMA Dermatol 155: 196-203.
  3. Fine JD, Johnson LB, Weiner M, Li KP, Suchindran C (2009) Epidermolysis bullosa and the risk of life-threatening cancers: The National EB Registry experience, 1986-2006. J Am Acad Dermatol 60: 203-211.
  4. Angelis A, Kanavos P, Lopez-Bastida J, Linertova R, Oliva-Moreno J, et al. (2016) Social/economic costs and health-related quality of life in patients with epidermolysis bullosa in Europe. Eur J Health Econ 1: 31-42.
  5. Wu YH, Sun FK, Lee PY (2020) Family caregivers' lived experiences of caring for epidermolysis bullosa patients: A phenomenological study. J Clin Nurs 29: 1552-1560.
  6. Bruckner AL, Losow M, Wisk J, Patel N, Reha A, et al. (2020) The challenges of living with and managing epidermolysis bullosa: Insights from patients and caregivers. Orphanet J Rare Dis. 15: 1.
  7. Shayegan LH, Levin LE, Galligan ER, Lucky AW, Bruckner AL, et al. (2020) Skin cleansing and topical product use in patients with epidermolysis bullosa: Results from a multicenter database. Pediatr Dermatol 37: 326-332.
  8. Lwin SM, Syed F, Di WL, Kadiyirire T, Liu L, et al. (2019) Safety and early efficacy outcomes for lentiviral fibroblast gene therapy in recessive dystrophic epidermolysis bullosa. JCI Insight 4.
  9. Friedenstein AJ, Chailakhjan RK, Lalykina KS (1970) The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet 3: 393-403.
  10. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, et al. (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8: 315-317.
  11. Tolar J, Ishida-Yamamoto A, Riddle M, McElmurry RT, Osborn M, et al. (2009) Amelioration of epidermolysis bullosa by transfer of wild-type bone marrow cells. Blood 113: 1167-1174.
  12. Wagner JE, Ishida-Yamamoto A, McGrath JA, Hordinsky M, Keene DR, et al. (2010) Bone marrow transplantation for recessive dystrophic epidermolysis bullosa. N Engl J Med 363: 629-639.
  13. Tolar J, Wagner JE (2013) Allogeneic blood and bone marrow cells for the treatment of severe epidermolysis bullosa: Repair of the extracellular matrix. Lancet 382: 1214-1223.
  14. Ebens CL, McGrath JA, Tamai K, Hovnanian A, Wagner JE, et al. (2019) Bone marrow transplant with post-transplant cyclophosphamide for recessive dystrophic epidermolysis bullosa expands the related donor pool and permits tolerance of nonhaematopoietic cellular grafts. Br J Dermatol 181: 1238-1246.
  15. Conget P, Rodriguez F, Kramer S, Allers C, Simon V, et al. (2010) Replenishment of type VII collagen and re-epithelialization of chronically ulcerated skin after intradermal administration of allogeneic mesenchymal stromal cells in two patients with recessive dystrophic epidermolysis bullosa. Cytotherapy 12: 429-431.
  16. El-Darouti M, Fawzy M, Amin I, Abdel Hay R, Hegazy R, et al. (2016) Treatment of dystrophic epidermolysis bullosa with bone marrow non-hematopoeitic stem cells: A randomized controlled trial. Dermatol Ther 29: 96-100.
  17. Petrof G, Lwin SM, Martinez-Queipo M, Abdul-Wahab A, Tso S, Mellerio JE, et al. (2015) Potential of systemic allogeneic mesenchymal stromal cell therapy for children with recessive dystrophic epidermolysis bullosa. J Invest Dermatol 135: 2319-2321.
  18. Rashidghamat E, Kadiyirire T, Ayis S, Petrof G, Liu L, et al. (2019) Phase I/II open-label trial of intravenous allogeneic mesenchymal stromal cell therapy in adults with recessive dystrophic epidermolysis bullosa. J Am Acad Dermatol 83: 447-454.
  19. Viswanathan S, Shi Y, Galipeau J, Krampera M, Leblanc K, et al. (2019) Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy (ISCT(R)) Mesenchymal Stromal Cell committee position statement on nomenclature. Cytotherapy 21: 1019-1024.
  20. Schatton T, Yang J, Kleffel S, Uehara M, Barthel SR, et al. (2015) ABCB5 Identifies Immunoregulatory Dermal Cells. Cell Rep 12: 1564-1574.
  21. Webber BR, O'Connor KT, McElmurry RT, Durgin EN, Eide CR, et al. (2017) Rapid generation of Col7a1(-/-) mouse model of recessive dystrophic epidermolysis bullosa and partial rescue via immunosuppressive dermal mesenchymal stem cells. Lab Invest 97: 1218-1224.
  22. Beken SV, de Vries JC, Meier-Schiesser B, Meyer P, Jiang D, et al. (2019) Newly Defined ATP-binding cassette subfamily b member 5 positive dermal mesenchymal stem cells promote healing of chronic iron-overload wounds via secretion of interleukin-1 receptor antagonist. Stem Cells 37: 1057-1074.
  23. Tappenbeck N, Schroder HM, Niebergall-Roth E, Hassinger F, Dehio U, et al. (2019) In vivo safety profile and biodistribution of GMP-manufactured human skin-derived ABCB5-positive mesenchymal stromal cells for use in clinical trials. Cytotherapy 21: 546-560.

Citation: Ebens CL, Riedl JA, Tolar J (2021) Tissue-Specific Differences in Mesenchymal Stromal Cells Influence Homing and Function. J Oncol Res Treat S1:001. DOI: 10.4172/aot.s1.1000001

Copyright: © 2021 Ebens CL, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution Licenpsee,r mwithsich unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Top