A systematic review and meta-analysis on the prevalence of Dupuytren disease in the general population of Western countries

Rosanne Lanting, Dieuwke C Broekstra, Paul M N Werker, Edwin R van den Heuvel, Rosanne Lanting, Dieuwke C Broekstra, Paul M N Werker, Edwin R van den Heuvel

Abstract

Background: Dupuytren disease is a fibroproliferative disease of palmar fascia of the hand. Its prevalence has been the subject of several reviews; however, an accurate description of the prevalence range in the general population--and of the relation between age and disease--is lacking.

Methods: Embase and PubMed were searched using database-specific Medical Subject Headings; titles and abstracts were searched for the words "Dupuytren," "incidence," and "prevalence." Two reviewers independently assessed the articles using inclusion and exclusion criteria, and rated the included studies with a quality assessment instrument. In a meta-analysis, the median prevalence, as a function of age by sex, was estimated, accompanied by 95 percent prediction intervals. The observed heterogeneity in prevalence was investigated with respect to study quality and geographic location.

Results: Twenty-three of 199 unique identified articles were included. The number of participants ranged from 37 to 97,537, and age ranged from 18 to 100 years. Prevalence varied from 0.6 to 31.6 percent. The quality of studies differed but could not explain the heterogeneity among studies. Mean prevalence was estimated as 12, 21, and 29 percent at ages 55, 65, and 75 years, respectively, based on the relation between age and prevalence determined from 10 studies.

Conclusions: The authors describe a prevalence range of Dupuytren disease in the general population of Western countries. The relation between age and prevalence of Dupuytren disease is given according to sex, including 95 percent prediction intervals. It is possible to determine disease prevalence at a certain age for the total population, and for men and women separately.

Figures

Fig. 72.1
Fig. 72.1
Types of cells derived as a result of chemical (polyethylene glycol/dimethyl sulfoxide - PEG/DMSO) ex vivo fusion of two different cell lineages. Fusion of cells derived from the same lineages creates syncytium with multiple nuclei N ≥ 2 (1) or with single nucleus – homotypic synkaryon (2). Fusion of cells derived from different lineages creates heterotypic synkaryon (3) or heterokaryon (4). If the fusion of cells derived from different lineages is not complete, hemi-fused cells are created (5). Toxicity of fusion can cause cell death (6). Cells can also not undergo fusion due to lack of other cells in proximity or inappropriate fusion conditions (7)
Fig. 72.2
Fig. 72.2
The mechanism of polyethylene glycol/dimethyl sulfoxide (PEG/DMSO) induced donor-recipient chimeric cells creation via ex vivo cell fusion (CF). PEG mediated CF is a three-step process requiring the following: (1) aggregation or “close” (the intercellular distance may vary for different cells and fusion models) approach of membrane lipid bilayers due to hydrophobic properties of PEG that causes membrane dehydration; (2) removal of the water between adjacent cells; (3) the intermediate membrane destabilization (facilitated by PEG) is followed by creation of pores (facilitated by DMSO) in the membranes of cells undergoing fusion; (4) positive osmotic pressure created by PEG improves stabilization of fusion intermediates and leads to expansion of the pores, cell swelling and cell-to-cell fusion. The products of PEG/DMSO solution induced cell fusion may include (5) heterokaryon and synkaryon cells as well as cells that did not undergo fusion process. More detailed descriptions of cell fusion mechanism can be found in articles by Lentz [62, 63]
Fig. 72.3
Fig. 72.3
Experimental model of ex vivo creation of the donor-recipient chimeric cells (DRCC). DRCC will be created ex vivo by the chemical polyethylene glycol (PEG) induced cell fusion of the bone marrow cells harvested from the ACI (RT1a) and Lewis (RT1l) rat donors. Isolated bone marrow cells will be separately stained with two different (red/orange and green) fluorescent dyes. Next, the ex vivo fusion will be performed using PEG. Supportive therapy using the fused DRCC will be given based on the double fluorescent staining and will be injected into the bone of Lewis (RT1l) rat recipients along with the donor matching (ACI) VCA (skin allograft) transplant. * – Seven day protocol of combined αβ-TCR mAb (250 μg/day) and CsA (16 mg/kg/day) therapy
Fig. 72.4
Fig. 72.4
Future applications of the ex vivo created donor-recipient chimeric cells used as supportive therapy in the clinical scenario. Human donor-recipient chimeric cells can be utilized as a supportive therapy for solid organ (living donor- kidney, liver transplantation) and in the future for vascularized composite allotransplantation (VCA). Progenitor cells derived from sources such as bone marrow or cord blood will be isolated, fluorescently labeled using two different cell membrane dyes (PKH26 and PKH67), and will be fused ex vivo using PEG technique creating the donor-recipient chimeric cells. Based on the double fluorescent staining, the ex vivo fused chimeric cells will be sorted out and delivered via either the intraosseous or intravenous route to the recipient at the day of solid organ or VCA transplants. Panel (a) – Patients receiving transplant from the living donor will be supported with the bone marrow derived donor-recipient chimeric cells collected from both the donor and the transplant recipient. Panel (b) – If access to the donor and/or recipients bone marrow cells is not possible (i.e. recipient is suffering from severe bone marrow deficiencies due to gamma irradiation or organ donor deceased), the donor and recipient HLA-matched cord blood cells can be used to create an ex vivo donor-recipient chimeric cells and to apply them as a supportive therapy

References

    1. Schwann T. Mikroskopische Untersuchungen uber die Ubereinstimmung in der Struktur und dem Wachsten der Thiere und Pflanzen. Berlin: Saunderschen Buchhandlung; 1839.
    1. Dittmar T, Zanker KS. Cell fusion in health and disease. Volume II: cell fusion in disease. Introduction. Adv Exp Med Biol. 2011;714:1–3.
    1. Chen EH, Olson EN. Unveiling the mechanisms of cell-cell fusion. Science. 2005;308(5720):369–73. doi: 10.1126/science.1104799.
    1. Zhou X, Platt JL. Molecular and cellular mechanisms of mammalian cell fusion. Adv Exp Med Biol. 2011;713:33–64. doi: 10.1007/978-94-007-0763-4_4.
    1. Ramakrishnan M, Mathur SR, Mukhopadhyay A. Fusion-derived epithelial cancer cells express hematopoietic markers and contribute to stem cell and migratory phenotype in ovarian carcinoma. Cancer Res. 2013;73(17):5360–70. doi: 10.1158/0008-5472.CAN-13-0896.
    1. Silk AD, et al. Fusion between hematopoietic and epithelial cells in adult human intestine. PLoS ONE. 2013;8(1):e55572. doi: 10.1371/journal.pone.0055572.
    1. Barski G, Sorieul S, Cornefert F. “Hybrid” type cells in combined cultures of two different mammalian cell strains. J Natl Cancer Inst. 1961;26:1269–91.
    1. Terada N, et al. Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion. Nature. 2002;416(6880):542–5. doi: 10.1038/nature730.
    1. Ying QL, et al. Changing potency by spontaneous fusion. Nature. 2002;416(6880):545–8. doi: 10.1038/nature729.
    1. Dittmar T, et al. Characterization of hybrid cells derived from spontaneous fusion events between breast epithelial cells exhibiting stem-like characteristics and breast cancer cells. Clin Exp Metastasis. 2011;28(1):75–90. doi: 10.1007/s10585-010-9359-3.
    1. Lu X, Kang Y. Efficient acquisition of dual metastasis organotropism to bone and lung through stable spontaneous fusion between MDA-MB-231 variants. Proc Natl Acad Sci U S A. 2009;106(23):9385–90. doi: 10.1073/pnas.0900108106.
    1. Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256(5517):495–7. doi: 10.1038/256495a0.
    1. Miller FR, Mohamed AN, McEachern D. Production of a more aggressive tumor cell variant by spontaneous fusion of two mouse tumor subpopulations. Cancer Res. 1989;49(15):4316–21.
    1. Nagler C, et al. Co-cultivation of murine BMDCs with 67NR mouse mammary carcinoma cells give rise to highly drug resistant cells. Cancer Cell Int. 2011;11(1):21. doi: 10.1186/1475-2867-11-21.
    1. LaBarge MA, Blau HM. Biological progression from adult bone marrow to mononucleate muscle stem cell to multinucleate muscle fiber in response to injury. Cell. 2002;111(4):589–601. doi: 10.1016/S0092-8674(02)01078-4.
    1. Alvarez-Dolado M, et al. Fusion of bone-marrow-derived cells with Purkinje neurons, cardiomyocytes and hepatocytes. Nature. 2003;425(6961):968–73. doi: 10.1038/nature02069.
    1. Vassilopoulos G, Wang PR, Russell DW. Transplanted bone marrow regenerates liver by cell fusion. Nature. 2003;422(6934):901–4. doi: 10.1038/nature01539.
    1. Wang X, et al. Cell fusion is the principal source of bone-marrow-derived hepatocytes. Nature. 2003;422(6934):897–901. doi: 10.1038/nature01531.
    1. Weimann JM, et al. Contribution of transplanted bone marrow cells to Purkinje neurons in human adult brains. Proc Natl Acad Sci U S A. 2003;100(4):2088–93. doi: 10.1073/pnas.0337659100.
    1. Nygren JM, et al. Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat Med. 2004;10(5):494–501. doi: 10.1038/nm1040.
    1. Rizvi AZ, et al. Bone marrow-derived cells fuse with normal and transformed intestinal stem cells. Proc Natl Acad Sci U S A. 2006;103(16):6321–5. doi: 10.1073/pnas.0508593103.
    1. Johansson CB, et al. Extensive fusion of haematopoietic cells with Purkinje neurons in response to chronic inflammation. Nat Cell Biol. 2008;10(5):575–83. doi: 10.1038/ncb1720.
    1. Camargo FD, Finegold M, Goodell MA. Hematopoietic myelomonocytic cells are the major source of hepatocyte fusion partners. J Clin Invest. 2004;113(9):1266–70. doi: 10.1172/JCI21301.
    1. Willenbring H, et al. Myelomonocytic cells are sufficient for therapeutic cell fusion in liver. Nat Med. 2004;10(7):744–8. doi: 10.1038/nm1062.
    1. Powell AE, et al. Fusion between Intestinal epithelial cells and macrophages in a cancer context results in nuclear reprogramming. Cancer Res. 2011;71(4):1497–505. doi: 10.1158/0008-5472.CAN-10-3223.
    1. Dales S, Siminovitch L. The development of vaccinia virus in Earle’s L strain cells as examined by electron microscopy. J Biophys Biochem Cytol. 1961;10:475–503. doi: 10.1083/jcb.10.4.475.
    1. Okada Y, Hosokawa Y. Isolation of a new variant of HVJ showing low cell fusion activity. Biken J. 1961;4:217–20.
    1. Okada Y. Analysis of giant polynuclear cell formation caused by HVJ virus from Ehrlich’s ascites tumor cells. III. Relationship between cell condition and fusion reaction or cell degeneration reaction. Exp Cell Res. 1962;26:119–28. doi: 10.1016/0014-4827(62)90207-0.
    1. Yeheskely-Hayon D, et al. Optically induced cell fusion using bispecific nanoparticles. Small. 2013;9(22):3771–7. doi: 10.1002/smll.201300696.
    1. Gottesman A, Milazzo J, Lazebnik Y. V-fusion: a convenient, nontoxic method for cell fusion. Biotechniques. 2010;49(4):747–50. doi: 10.2144/000113515.
    1. Golestani R, Pourfathollah AA, Moazzeni SM. Cephalin as an efficient fusogen in hybridoma technology: can it replace poly ethylene glycol? Hybridoma (Larchmt) 2007;26(5):296–301. doi: 10.1089/hyb.2007.0510.
    1. Cheong SC, et al. Generation of cell hybrids via a fusogenic cell line. J Gene Med. 2006;8(7):919–28. doi: 10.1002/jgm.906.
    1. Pedrazzoli F, et al. Cell fusion in tumor progression: the isolation of cell fusion products by physical methods. Cancer Cell Int. 2011;11:32. doi: 10.1186/1475-2867-11-32.
    1. Norwood TH, Zeigler CJ, Martin GM. Dimethyl sulfoxide enhances polyethylene glycol-mediated somatic cell fusion. Somatic Cell Genet. 1976;2(3):263–70. doi: 10.1007/BF01538964.
    1. Gurtovenko AA, Anwar J. Modulating the structure and properties of cell membranes: the molecular mechanism of action of dimethyl sulfoxide. J Phys Chem B. 2007;111(35):10453–60. doi: 10.1021/jp073113e.
    1. Guo-Parke H, et al. Configuration of electrofusion-derived human insulin-secreting cell line as pseudoislets enhances functionality and therapeutic utility. J Endocrinol. 2012;214(3):257–65. doi: 10.1530/JOE-12-0188.
    1. Schwaber J, Cohen EP. Human x mouse somatic cell hybrid clone secreting immunoglobulins of both parental types. Nature. 1973;244(5416):444–7. doi: 10.1038/244444a0.
    1. Tesarik J, et al. Chemically and mechanically induced membrane fusion: non-activating methods for nuclear transfer in mature human oocytes. Hum Reprod. 2000;15(5):1149–54. doi: 10.1093/humrep/15.5.1149.
    1. Bhutani N, et al. Reprogramming towards pluripotency requires AID-dependent DNA demethylation. Nature. 2010;463(7284):1042–7. doi: 10.1038/nature08752.
    1. Duelli DM, et al. A virus causes cancer by inducing massive chromosomal instability through cell fusion. Curr Biol. 2007;17(5):431–7. doi: 10.1016/j.cub.2007.01.049.
    1. Lu X, Kang Y. Cell fusion hypothesis of the cancer stem cell. Adv Exp Med Biol. 2011;714:129–40. doi: 10.1007/978-94-007-0782-5_6.
    1. Blau HM, Blakely BT. Plasticity of cell fate: insights from heterokaryons. Semin Cell Dev Biol. 1999;10(3):267–72. doi: 10.1006/scdb.1999.0311.
    1. Duelli DM, Lazebnik YA. Primary cells suppress oncogene-dependent apoptosis. Nat Cell Biol. 2000;2(11):859–62. doi: 10.1038/35041112.
    1. Malka F, et al. The mitochondria of cultured mammalian cells: I. Analysis by immunofluorescence microscopy, histochemistry, subcellular fractionation, and cell fusion. Methods Mol Biol. 2007;372:3–16. doi: 10.1007/978-1-59745-365-3_1.
    1. Rao PN, Johnson RT. Mammalian cell fusion. IV. Regulation of chromosome formation from interphase nuclei by various chemical compounds. J Cell Physiol. 1971;78(2):217–23. doi: 10.1002/jcp.1040780208.
    1. Park SU, et al. Effects of chromosomal polyploidy on survival of colon cancer cells. Korean J Gastroenterol. 2011;57(3):150–7. doi: 10.4166/kjg.2011.57.3.150.
    1. Walter MA, Goodfellow PN. Irradiation and fusion gene transfer. Mol Biotechnol. 1995;3(2):117–28. doi: 10.1007/BF02789107.
    1. Upender MB, et al. Chromosome transfer induced aneuploidy results in complex dysregulation of the cellular transcriptome in immortalized and cancer cells. Cancer Res. 2004;64(19):6941–9. doi: 10.1158/0008-5472.CAN-04-0474.
    1. Waid TH, et al. Treatment of acute cellular rejection with T10B9.1A-31 or OKT3 in renal allograft recipients. Transplantation. 1992;53(1):80–6. doi: 10.1097/00007890-199201000-00015.
    1. Kovarik J, et al. Disposition of basiliximab, an interleukin-2 receptor monoclonal antibody, in recipients of mismatched cadaver renal allografts. Transplantation. 1997;64(12):1701–5. doi: 10.1097/00007890-199712270-00012.
    1. Koido S, et al. Regulation of tumor immunity by tumor/dendritic cell fusions. Clin Dev Immunol. 2010;2010:516768. doi: 10.1155/2010/516768.
    1. Weise JB, et al. A dendritic cell based hybrid cell vaccine generated by electrofusion for immunotherapy strategies in HNSCC. Auris Nasus Larynx. 2004;31(2):149–53. doi: 10.1016/j.anl.2004.01.007.
    1. Kanamoto A, Maki T. Chimeric donor cells play an active role in both induction and maintenance phases of transplantation tolerance induced by mixed chimerism. J Immunol. 2004;172(3):1444–8. doi: 10.4049/jimmunol.172.3.1444.
    1. Siemionow M, et al. Development and maintenance of donor-specific chimerism in semi-allogenic and fully major histocompatibility complex mismatched facial allograft transplants. Transplantation. 2005;79(5):558–67. doi: 10.1097/01.TP.0000152799.16035.B7.
    1. Ayala R, et al. Long-term follow-up of donor chimerism and tolerance after human liver transplantation. Liver Transpl. 2009;15(6):581–91. doi: 10.1002/lt.21736.
    1. Muramatsu K, Kuriyama R, Taguchi T. Intragraft chimerism following composite tissue allograft. J Surg Res. 2009;157(1):129–35. doi: 10.1016/j.jss.2008.06.026.
    1. Rahhal DN, et al. Dissociation between peripheral blood chimerism and tolerance to hindlimb composite tissue transplants: preferential localization of chimerism in donor bone. Transplantation. 2009;88(6):773–81. doi: 10.1097/TP.0b013e3181b47cfa.
    1. Bonde S, et al. Cell fusion of bone marrow cells and somatic cell reprogramming by embryonic stem cells. FASEB J. 2010;24(2):364–73. doi: 10.1096/fj.09-137141.
    1. Cwykiel J, Klimczak A. Therapeutic Potential of Ex-vivo Fused Chimeric Cells in Prolonging Vascularized Skin Allograft Survival. PRS. 2011; 127:26.
    1. Li LH, et al. Electrofusion between heterogeneous-sized mammalian cells in a pellet: potential applications in drug delivery and hybridoma formation. Biophys J. 1996;71(1):479–86. doi: 10.1016/S0006-3495(96)79249-4.
    1. Chow T, et al. The transfer of host MHC class I protein protects donor cells from NK cell and macrophage-mediated rejection during hematopoietic stem cell transplantation and engraftment in mice. Stem Cells. 2013;31(10):2242–52. doi: 10.1002/stem.1458.
    1. Lentz BR. PEG as a tool to gain insight into membrane fusion. Eur Biophys J. 2007;36(4–5):315–26. doi: 10.1007/s00249-006-0097-z.
    1. Lentz BR. Polymer-induced membrane fusion: potential mechanism and relation to cell fusion events. Chem Phys Lipids. 1994;73(1–2):91–106. doi: 10.1016/0009-3084(94)90176-7.

Source: PubMed

3
Subskrybuj