1
|
Sarkar T, Sarkar S, Gangopadhyay DN. Gene Therapy and its Application in Dermatology. Indian J Dermatol 2020; 65:341-350. [PMID: 33165431 PMCID: PMC7640808 DOI: 10.4103/ijd.ijd_323_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Gene therapy is an experimental technique to treat genetic diseases. It is based on the introduction of nucleic acid with the help of a vector, into a diseased cell or tissue, to correct the gene expression and thus prevent, halt, or reverse a pathological process. It is a promising treatment approach for genetic diseases, inherited diseases, vaccination, cancer, immunomodulation, as well as healing of some refractory ulcers. Both viral and nonviral vectors can be used to deliver the correct gene. An ideal vector should have the ability for sustained gene expression, acceptable coding capacity, high transduction efficiency, and devoid of mutagenicity. There are different techniques of vector delivery, but these techniques are still under research for assessment of their safety and effectiveness. The major challenges of gene therapy are immunogenicity, mutagenicity, and lack of sustainable therapeutic benefit. Despite these constraints, therapeutic success was obtained in a few genetic and inherited skin diseases. Skin being the largest, superficial, easily accessible and assessable organ of the body, may be a promising target for gene therapy research in the recent future.
Collapse
Affiliation(s)
- Tanusree Sarkar
- From Department of Dermatology, Burdwan Medical College, West Bengal, India
| | - Somenath Sarkar
- Department of Dermatology, B. S Medical College, West Bengal, India
| | | |
Collapse
|
2
|
Downregulation of TNIP1 Expression Leads to Increased Proliferation of Human Keratinocytes and Severer Psoriasis-Like Conditions in an Imiquimod-Induced Mouse Model of Dermatitis. PLoS One 2015; 10:e0127957. [PMID: 26046540 PMCID: PMC4457880 DOI: 10.1371/journal.pone.0127957] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 04/22/2015] [Indexed: 02/04/2023] Open
Abstract
Psoriasis is a chronic, inflammatory skin disease involving both environmental and genetic factors. According to genome-wide association studies (GWAS), the TNIP1 gene, which encodes the TNF-α–induced protein 3-interacting protein 1 (TNIP1), is strongly linked to the susceptibility of psoriasis. TNIP1 is a widely expressed ubiquitin sensor that binds to the ubiquitin-editing protein A20 and restricts TNF- and TLR-induced signals. In our study, TNIP1 expression decreased in specimens of epidermis affected by psoriasis. Based on previous studies suggesting a role for TNIP1 in modulating cancer cell growth, we investigated its role in keratinocyte proliferation, which is clearly abnormal in psoriasis. To mimic the downregulation or upregulation of TNIP1 in HaCaT cells and primary human keratinocytes (PHKs), we used a TNIP1 specific small interfering hairpin RNA (TNIP1 shRNA) lentiviral vector or a recombinant TNIP1 (rTNIP1) lentiviral vector, respectively. Blocking TNIP1 expression increased keratinocyte proliferation, while overexpression of TNIP1 decreased keratinocyte proliferation. Furthermore, we showed that TNIP1 signaling might involve extracellular signal-regulated kinase1/2 (Erk1/2) and CCAAT/enhancer-binding protein β (C/EBPβ) activity. Intradermal injection of TNIP1 shRNA in BALB/c mice led to exaggerated psoriatic conditions in imiquimod (IMQ)-induced psoriasis-like dermatitis. These findings indicate that TNIP1 has a protective role in psoriasis and therefore could be a promising therapeutic target.
Collapse
|
3
|
A novel gene therapy strategy using secreted multifunctional anti-HIV proteins to confer protection to gene-modified and unmodified target cells. Gene Ther 2013; 21:175-87. [PMID: 24305417 DOI: 10.1038/gt.2013.70] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 08/26/2013] [Accepted: 09/24/2013] [Indexed: 11/08/2022]
Abstract
Current human immunodeficiency virus type I (HIV) gene therapy strategies focus on rendering HIV target cells non-permissive to viral replication. However, gene-modified cells fail to accumulate in patients and the virus continues to replicate in the unmodified target cell population. We have designed lentiviral vectors encoding secreted anti-HIV proteins to protect both gene-modified and unmodified cells from infection. Soluble CD4 (sCD4), a secreted single chain variable fragment (sscFv(17b)) and a secreted fusion inhibitor (sFI(T45)) were used to target receptor binding, co-receptor binding and membrane fusion, respectively. Additionally, we designed bi- and tri-functional fusion proteins to exploit the multistep nature of HIV entry. Of the seven antiviral proteins tested, sCD4, sCD4-scFv(17b), sCD4-FI(T45) and sCD4-scFv(17b)-FI(T45) efficiently inhibited HIV entry. The neutralization potency of the bi-functional fusion proteins sCD4-scFv(17b) and sCD4-FI(T45) was superior to that of sCD4 and the Food and Drug Administration-approved fusion inhibitor T-20. In co-culture experiments, sCD4, sCD4-scFv(17b) and sCD4-FI(T45) secreted from gene-modified producer cells conferred substantial protection to unmodified peripheral blood mononuclear cells. In conclusion, continuous delivery of secreted anti-HIV proteins via gene therapy may be a promising strategy to overcome the limitations of the current treatment.
Collapse
|
4
|
Staunstrup NH, Bak RO, Cai Y, Svensson L, Petersen TK, Rosada C, Stenderup K, Bolund L, Mikkelsen JG. A lentiviral vector-based genetic sensor system for comparative analysis of permeability and activity of vitamin D3 analogues in xenotransplanted human skin. Exp Dermatol 2013; 22:178-83. [DOI: 10.1111/exd.12091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2013] [Indexed: 01/18/2023]
Affiliation(s)
| | - Rasmus O. Bak
- Department of Biomedicine; Aarhus University; Aarhus C; Denmark
| | - Yujia Cai
- Department of Biomedicine; Aarhus University; Aarhus C; Denmark
| | - Lars Svensson
- Department of Disease Pharmacology; LEO Pharma; Ballerup; Denmark
| | | | - Cecilia Rosada
- Department of Clinical Medicine; Aarhus University; Aarhus C; Denmark
| | - Karin Stenderup
- Department of Clinical Medicine; Aarhus University; Aarhus C; Denmark
| | | | | |
Collapse
|
5
|
Abstract
Post-transcriptional managing of RNA plays a key role in the intricate network of cellular pathways that regulate our genes. Numerous small RNA species have emerged as crucial regulators of RNA processing and translation. Among these, microRNAs (miRNAs) regulate protein synthesis through specific interactions with target RNAs and are believed to play a role in almost any cellular process and tissue. Skin is no exception, and miRNAs are intensively studied for their role in skin homoeostasis and as potential triggers of disease. For use in skin and many other tissues, therapeutic RNA managing by small RNA technologies is now widely explored. Despite the easy accessibility of skin, the natural barrier properties of skin have challenged genetic intervention studies, and unique tools for studying gene expression and the regulatory role of small RNAs, including miRNAs, in human skin are urgently needed. Human immunodeficiency virus (HIV)-derived lentiviral vectors (LVs) have been established as prominent carriers of foreign genetic cargo. In this review, we describe the use of HIV-derived LVs for efficient gene transfer to skin and establishment of long-term transgene expression in xenotransplanted skin. We outline the status of engineered LVs for delivery of small RNAs and their in vivo applicability for expression of genes and small RNA effectors including small hairpin RNAs, miRNAs and miRNA inhibitors. Current findings suggest that LVs may become key tools in experimental dermatology with particular significance for cutaneous RNA managing and in vivo genetic intervention.
Collapse
|
6
|
Geusens B, Strobbe T, Bracke S, Dynoodt P, Sanders N, Gele MV, Lambert J. Lipid-mediated gene delivery to the skin. Eur J Pharm Sci 2011; 43:199-211. [DOI: 10.1016/j.ejps.2011.04.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2010] [Revised: 11/16/2010] [Accepted: 04/09/2011] [Indexed: 11/29/2022]
|
7
|
Kunicher N, Tzur T, Amar D, Chaouat M, Yaacov B, Panet A. Characterization of factors that determine lentiviral vector tropism in skin tissue using an ex vivo model. J Gene Med 2011; 13:209-20. [DOI: 10.1002/jgm.1554] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
8
|
Bak RO, Mikkelsen JG. Regulation of cytokines by small RNAs during skin inflammation. J Biomed Sci 2010; 17:53. [PMID: 20594301 PMCID: PMC2905360 DOI: 10.1186/1423-0127-17-53] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 07/01/2010] [Indexed: 02/08/2023] Open
Abstract
Intercellular signaling by cytokines is a vital feature of the innate immune system. In skin, an inflammatory response is mediated by cytokines and an entwined network of cellular communication between T-cells and epidermal keratinocytes. Dysregulated cytokine production, orchestrated by activated T-cells homing to the skin, is believed to be the main cause of psoriasis, a common inflammatory skin disorder. Cytokines are heavily regulated at the transcriptional level, but emerging evidence suggests that regulatory mechanisms that operate after transcription play a key role in balancing the production of cytokines. Herein, we review the nature of cytokine signaling in psoriasis with particular emphasis on regulation by mRNA destabilizing elements and the potential targeting of cytokine-encoding mRNAs by miRNAs. The proposed linkage between mRNA decay mediated by AU-rich elements and miRNA association is described and discussed as a possible general feature of cytokine regulation in skin. Moreover, we describe the latest attempts to therapeutically target cytokines at the RNA level in psoriasis by exploiting the cellular RNA interference machinery. The applicability of cytokine-encoding mRNAs as future clinical drug targets is evaluated, and advances and obstacles related to topical administration of RNA-based drugs targeting the cytokine circuit in psoriasis are described.
Collapse
Affiliation(s)
- Rasmus O Bak
- Department of Human Genetics, University of Aarhus, DK-8000 Aarhus C, Denmark
| | | |
Collapse
|
9
|
Pfützner W. Vectors for gene therapy of skin diseases. J Dtsch Dermatol Ges 2010; 8:582-91. [DOI: 10.1111/j.1610-0387.2010.07435.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
10
|
Scheidemann F, Therrien JP, Vogel J, Pfützner W. In vivosynthesis and secretion of erythropoietin by genetically modified primary human keratinocytes grafted onto immunocompromised mice. Exp Dermatol 2010; 19:289-97. [DOI: 10.1111/j.1600-0625.2009.00984.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
11
|
Epo delivery by genetically engineered C2C12 myoblasts immobilized in microcapsules. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 670:54-67. [PMID: 20384218 DOI: 10.1007/978-1-4419-5786-3_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ver the last half century, the use of erythropoietin (Epo) in the management of malignancies has been extensively studied. Originally viewed as the renal hormone responsible for red blood cell production, many recent in vivo and clinical approaches demonstrate that various tissues locally produce Epo in response to physical or metabolic stress. Thus, not only its circulating erythrocyte mass regulator activity but also the recently discovered nonhematological actions are being thoroughly investigated in order to fulfill the specific Epo delivery requirements for each therapeutic approach.
Collapse
|
12
|
Jakobsen M, Stenderup K, Rosada C, Moldt B, Kamp S, Dam TN, Jensen TG, Mikkelsen JG. Amelioration of psoriasis by anti-TNF-alpha RNAi in the xenograft transplantation model. Mol Ther 2009; 17:1743-53. [PMID: 19568223 DOI: 10.1038/mt.2009.141] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-alpha) is upregulated in psoriatic skin and represents a prominent target in psoriasis treatment. The level of TNF-alpha-encoding mRNA, however, is not increased in psoriatic skin, and it remains unclear whether intervention strategies based on RNA interference (RNAi) are therapeutically relevant. To test this hypothesis the present study describes first the in vitro functional screening of a panel of short hairpin RNAs (shRNAs) targeting human TNF-alpha mRNA and, next, the transfer of the most potent TNF-alpha shRNA variant, as assessed in vitro, to human skin in the psoriasis xenograft transplantation model by the use of lentiviral vectors. TNF-alpha shRNA treatment leads to amelioration of the psoriasis phentotype in the model, as documented by reduced epidermal thickness, normalization of the skin morphology, and reduced levels of TNF-alpha mRNA as detected in skin biopsies 3 weeks after a single vector injection of lentiviral vectors encoding TNF-alpha shRNA. Our data show efficient lentiviral gene delivery to psoriatic skin and therapeutic applicability of anti-TNF-alpha shRNAs in human skin. These findings validate TNF-alpha mRNA as a target molecule for a potential persistent RNA-based treatment of psoriasis and establish the use of small RNA effectors as a novel platform for target validation in psoriasis and other skin disorders.
Collapse
Affiliation(s)
- Maria Jakobsen
- Department of Human Genetics, University of Aarhus, Aarhus DK-8000, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Krishnamachary B, Glunde K, Wildes F, Mori N, Takagi T, Raman V, Bhujwalla ZM. Noninvasive detection of lentiviral-mediated choline kinase targeting in a human breast cancer xenograft. Cancer Res 2009; 69:3464-71. [PMID: 19336572 DOI: 10.1158/0008-5472.can-08-4120] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Elevated phosphocholine (PC) and total choline (tCho) metabolites are widely established characteristics of most cancer cells, including breast cancer. Effective silencing of choline kinase (chk), the enzyme that converts choline to PC, is associated with reduced tumor growth. The functional importance and down-regulation of chk using RNA interference has been previously established. Here, we report on the preclinical evaluation of lentiviral vector-mediated down-regulation of chk using short hairpin RNA (shRNA) in established tumors derived from human breast cancer cells. Concentrated lentivirus expressing shRNA against chk was injected i.v. in the tail vein of MDA-MB-231 tumor-bearing female severe combined immunodeficient mice. Transduction efficiency in cells and tumors in vivo was assessed optically by enhanced green fluorescent protein expression and additionally from chk mRNA and protein levels. An 80% reduction in chk mRNA and protein was achieved following approximately 90% transduction efficiency in cells. After transduction with chk-shRNA, (1)H magnetic resonance spectroscopy (MRS) of cell and tumor extracts showed decreases in PC and tCho levels (P < 0.01 and 0.05, respectively) in comparison with controls. PC levels were monitored noninvasively by (31)P MRS in tumors and by (1)H MRS in cell and tumor tissue extracts. Noninvasive (31)P MR spectra of chk-shRNA-transduced tumors in vivo showed lower PC and phosphomonoester levels that were associated with reduced tumor growth and proliferation. This study shows the use of lentiviral vectors to target chk in a human breast cancer xenograft and noninvasive MRS detection of this targeting.
Collapse
Affiliation(s)
- Balaji Krishnamachary
- Johns Hopkins University In Vivo Cellular Molecular Imaging Center Program, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Scheidemann F, Löser M, Niedermeier A, Kromminga A, Therrien JP, Vogel J, Pfützner W. The skin as a biofactory for systemic secretion of erythropoietin: potential of genetically modified keratinocytes and fibroblasts. Exp Dermatol 2008; 17:481-8. [DOI: 10.1111/j.1600-0625.2007.00680.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
15
|
Kunicher N, Falk H, Yaacov B, Tzur T, Panet A. Tropism of Lentiviral Vectors in Skin Tissue. Hum Gene Ther 2008; 19:255-66. [DOI: 10.1089/hum.2007.121] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Nikolai Kunicher
- Department of Virology, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Haya Falk
- Department of Virology, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Barak Yaacov
- Department of Virology, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Tomer Tzur
- Department of Plastic and Reconstructive Surgery, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Amos Panet
- Department of Virology, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| |
Collapse
|
16
|
Wang Q, Ilves H, Chu P, Contag CH, Leake D, Johnston BH, Kaspar RL. Delivery and Inhibition of Reporter Genes by Small Interfering RNAs in a Mouse Skin Model. J Invest Dermatol 2007; 127:2577-84. [PMID: 17522708 DOI: 10.1038/sj.jid.5700891] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
RNA interference offers the potential of a novel therapeutic approach for treating skin disorders. To this end, we investigated delivery of nucleic acids, including a plasmid expressing the reporter gene luciferase, to mouse skin by intradermal injection into footpads using in vivo bioluminescence imaging over multiple time points. In order to evaluate the ability of RNA interference to inhibit skin gene expression, reporter gene constructs were co-injected with specific or non-specific siRNAs and the in vivo effects measured. Our results revealed that specific unmodified and modified siRNAs (but not nonspecific matched controls) strongly inhibit reporter gene expression in mice. These results indicate that small interfering RNA, delivered locally as RNA directly or expressed from viral or non-viral vectors, may be effective agents for treating skin disorders.
Collapse
Affiliation(s)
- Qian Wang
- Molecular Imaging Program at Stanford, and Department of Radiology, Stanford University School of Medicine, Stanford, California, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Kikuchi Y, Tamai K, Kaneda Y. Cutaneous gene delivery. J Dermatol Sci 2007; 50:87-98. [PMID: 17765482 DOI: 10.1016/j.jdermsci.2007.07.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Revised: 07/10/2007] [Accepted: 07/20/2007] [Indexed: 12/25/2022]
Abstract
Over the past decade, many approaches to transferring genes into the skin have been investigated. However, most such approaches have been specifically aimed against genodermatosis, and have not produced sufficient results. The goal of such research is to develop a method in which genes are transferred easily, efficiently and stably into keratinocytes, especially into keratinocyte stem cells, and in which the transgene expression persists without a reaction from the host immune response. Although accidental development of cancer has occurred in trials of gene therapy for X-linked severe combined immunodeficiency (X-SCID), resulting in slowing of the progress of this research, the lessons of these setbacks have been applied to further research. Moreover, combined with the techniques acquired from tissue engineering, recent developments in our knowledge about stem cells will lead to new treatments for genodermatoses. The present review summarizes the methods by which therapeutic genes can be transferred into keratinocytes, with discussion of how gene transfer efficiency can be improved, with particular emphasis on disruption of the skin barrier function. It concludes with discussion of the challenges and prospects of keratinocyte gene therapy, in terms of achieving efficient and long-lasting therapeutic effects.
Collapse
Affiliation(s)
- Yasushi Kikuchi
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | |
Collapse
|
18
|
Hachiya A, Sriwiriyanont P, Patel A, Saito N, Ohuchi A, Kitahara T, Takema Y, Tsuboi R, Boissy RE, Visscher MO, Wilson JM, James WM, Kobinger GP. Gene transfer in human skin with different pseudotyped HIV-based vectors. Gene Ther 2007; 14:648-56. [PMID: 17268532 DOI: 10.1038/sj.gt.3302915] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pseudotyping lentiviral vector with other viral surface proteins could be applied for treating genetic anomalies in human skin. In this study, the modification of HIV vector tropism by pseudotyping with the envelope glycoprotein from vesicular stomatitis virus (VSV), the Zaire Ebola (EboZ) virus, murine leukemia virus (MuLV), lymphocytic choriomeningitis virus (LCMV), Rabies or the rabies-related Mokola virus encoding LacZ as a reporter gene was evaluated qualitatively and quantitatively in human skin xenografts. High transgene expression was detected in dermal fibroblasts transduced with VSV-G-, EboZ- or MuLV-pseudotyped HIV vector with tissue irregularities in the dermal compartments following repeated injections of EboZ- or LCMV-pseudotyped vectors. Four weeks after transduction, double-labeling immunofluorescence of beta-galactosidase and involucrin or integrin beta1 demonstrated that VSV-G-, EboZ- or MuLV-pseudotyped HIV vector effectively targeted quiescent epidermal stem cells which underwent terminal differentiation resulting in transgene expression in their progenies. Among the six different pseudotyped HIV-based vectors evaluated, VSV-G-pseudotyped vector was found to be the most efficient viral glycoprotein for cutaneous transduction as demonstrated by the highest level of beta-galactosidase expression and genome copy number evaluated by TaqMan PCR.
Collapse
Affiliation(s)
- A Hachiya
- Kao Biological Science Laboratories, Haga, Tochigi, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Possibilities of using the skin for somatic gene therapy have been investigated for more than 20 years. Strategies have included both direct gene transfer into the skin and indirect gene transfer utilizing cultured cells as an intermediate step for gene manipulation. Viral as well as nonviral vectors have been used, and both gene addition and gene editing have been performed. Although cutaneous gene therapy has now begun translating into clinical medicine (as seen by the first clinical gene therapy project of an inherited skin disorder) further developments are still required.
Collapse
|
20
|
Abstract
The capacity to induce neoplasia in human tissue in the laboratory has recently provided a new platform for cancer research. Malignant conversion can be achieved in vivo by expressing genes of interest in human tissue that has been regenerated on immune-deficient mice. Induction of cancer in regenerated human skin recapitulates the three-dimensional architecture, tissue polarity, basement membrane structure, extracellular matrix, oncogene signalling and therapeutic target proteins found in intact human skin in vivo. Human-tissue cancer models therefore provide an opportunity to elucidate fundamental cancer mechanisms, to assess the oncogenic potency of mutations associated with specific human cancers and to develop new cancer therapies.
Collapse
Affiliation(s)
- Paul A Khavari
- Veterans Affairs, Palo Alto Healthcare System, Palo Alto, California 94304, USA.
| |
Collapse
|
21
|
Lewin AS, Glazer PM, Milstone LM. Gene therapy for autosomal dominant disorders of keratin. J Investig Dermatol Symp Proc 2005; 10:47-61. [PMID: 16250209 DOI: 10.1111/j.1087-0024.2005.10207.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Dominant mutations that interfere with the assembly of keratin filaments cause painful and disfiguring epidermal diseases like pachyonychia congenita and epidermolysis bullosa simplex. Genetic therapies for such diseases must either suppress the production of the toxic proteins or correct the genetic defect in the chromosome. Because epidermal skin cells may be genetically modified in tissue culture or in situ, gene correction is a legitimate goal for keratin diseases. In addition, recent innovations, such as RNA interference in animals, make an RNA knockdown approach plausible in the near future. Although agents of RNA reduction (small interfering RNA, ribozymes, triplex oligonucleotides, or antisense DNA) can be delivered as nucleotides, the impermeability of the skin to large charged molecules presents a serious impediment. Using viral vectors to deliver genes for selective inhibitors of gene expression presents an attractive alternative for long-term treatment of genetic disease in the skin.
Collapse
MESH Headings
- Animals
- Darier Disease/genetics
- Darier Disease/therapy
- Dependovirus/genetics
- Ectodermal Dysplasia/genetics
- Ectodermal Dysplasia/therapy
- Epidermolysis Bullosa Simplex/genetics
- Epidermolysis Bullosa Simplex/therapy
- Gene Silencing
- Gene Targeting
- Genes, Dominant
- Genetic Therapy
- Genetic Vectors
- Humans
- Keratins/genetics
- Keratoderma, Palmoplantar/genetics
- Keratoderma, Palmoplantar/therapy
- Mice
- Mutation
- Nails, Malformed/genetics
- Nails, Malformed/therapy
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/therapeutic use
- RNA Interference
- RNA, Catalytic/genetics
- RNA, Catalytic/therapeutic use
- RNA, Small Interfering/genetics
- RNA, Small Interfering/therapeutic use
Collapse
Affiliation(s)
- Alfred S Lewin
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida 32610-0266, USA.
| | | | | |
Collapse
|
22
|
Brill-Almon E, Stern B, Afik D, Kaye J, Langer N, Bellomo S, Shavit M, Pearlman A, Lippin Y, Panet A, Shani N. Ex vivo transduction of human dermal tissue structures for autologous implantation production and delivery of therapeutic proteins. Mol Ther 2005; 12:274-82. [PMID: 16043098 DOI: 10.1016/j.ymthe.2005.03.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2005] [Revised: 03/17/2005] [Accepted: 03/24/2005] [Indexed: 11/21/2022] Open
Abstract
Systemic delivery of therapeutic proteins through gene transfer approaches has been carried out mostly by ex vivo transduction of single cells or by direct in vivo injection of an expression vector. In this work an intact miniature biopsy of human dermis (microdermis) is harvested and transduced ex vivo by a viral vector encoding a gene for the therapeutic protein. The microdermis preserves its three-dimensional structure and viability during the ex vivo manipulations. Furthermore, upon transduction with adenoviral and adeno-associated viral vectors the microdermis secretes recombinant human erythropoietin (hEPO). Biochemical analysis of the secreted hEPO showed similarity to the clinically approved recombinant hEPO. Subcutaneous implantation of microdermal hEPO into SCID mice exhibited hEPO secretion in the blood circulation and preserved elevated hematocrit for several months, demonstrating the technology's potential for sustained delivery of protein therapeutics.
Collapse
Affiliation(s)
- Einat Brill-Almon
- Medgenics, Inc., Biogenics Ltd., Teradion Business Park, Misgav, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lu Z, Ghazizadeh S. Host immune responses in ex vivo approaches to cutaneous gene therapy targeted to keratinocytes. Exp Dermatol 2005; 14:727-35. [PMID: 16176280 PMCID: PMC1343507 DOI: 10.1111/j.1600-0625.2005.00351.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Epidermal gene therapy may benefit a variety of inherited skin disorders and certain systemic diseases. Both in vivo and ex vivo approaches of gene transfer have been used to target human epidermal stem cells and achieve long-term transgene expression in immunodeficient mouse/human chimera models. Immunological responses however, especially in situations where a neoantigen is expressed, are likely to curtail expression and thereby limit the therapy. In vivo gene transfer to skin has been shown to induce transgene-specific immune responses. Ex vivo gene transfer approaches, where keratinocytes are transduced in culture and transplanted back to patient, however, may avoid signals provided to the immune system by in vivo administration of vectors. In the current study, we have developed a stable epidermal graft platform in immunocompetent mice to analyze host responses in ex vivo epidermal gene therapy. Using green fluorescent protein (GFP) as a neoantigen and an ex vivo retrovirus-mediated gene transfer to mouse primary epidermal cultures depleted of antigen-presenting cells (APCs), we show induction of GFP-specific immune responses leading to the clearance of transduced cells. Similar approach in immunocompetent mice tolerant to GFP resulted in permanent engraftment of transduced cells and continued GFP expression. Activation of transgene-specific immune responses in ex vivo gene transfer targeted to keratinocytes require cross-presentation of transgene product to APCs, a process that is most amenable to immune modulation. This model may be used to explore strategies to divert transgene-specific immune responses to less destructive or tolerogenic ones.
Collapse
Affiliation(s)
| | - Soosan Ghazizadeh
- *Corresponding author: Soosan Ghazizadeh, Ph.D., Department of Dermatology, College of Physicians & Surgeons, Columbia University, 630 West 168 Street VC-15-208, New York, NY 10032, Tel: (212) 305-9025, Fax: (212) 305-7391,
| |
Collapse
|
24
|
Hengge UR, Bardenheuer W. Gene therapy and the skin. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2005; 131C:93-100. [PMID: 15468151 DOI: 10.1002/ajmg.c.30038] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Significant progress has been made during the past decade in corrective gene therapy of the skin. This includes advances in vector technology, targeted gene expression, gene replacement, gene correction, and the availability of appropriate animal models for a variety of candidate diseases. While non-viral integration of large genes such as essential basement membrane proteins has been mastered, new challenges such as the control of immune responses lie ahead of the research community. Among the first skin diseases, patients with junctional epidermolysis bullosa (JEB) and xeroderma pigmentosum (XP) will enter clinical trials.
Collapse
Affiliation(s)
- U R Hengge
- Department of Dermatology, University of Düsseldorf, Germany.
| | | |
Collapse
|
25
|
Jensen TG. Strategies for long-term gene expression in the skin to treat metabolic disorders. Expert Opin Biol Ther 2005; 4:677-82. [PMID: 15155159 DOI: 10.1517/14712598.4.5.677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Due to its accessibility, size and contact with the blood circulation, the skin is an attractive target for somatic gene therapy. Permanent cutaneous expression can be achieved by genetic manipulation of epidermal keratinocytes ex vivo followed by transplantation or by local injection of viral vectors. Furthermore, progress is being made to develop topical gene transfer methods leading to permanent gene expression. There is experimental evidence showing that genetically engineered skin can produce and secrete medically relevant proteins to the circulation and also produce enzymes that can clear metabolites accumulating in various diseases. Thus, cutaneous gene transfer approaches may be relevant not only for local skin diseases, but also for certain systemic disorders.
Collapse
|
26
|
Song J, Izumi K, Lanigan T, Feinberg SE. Development and characterization of a canine oral mucosa equivalent in a serum-free environment. J Biomed Mater Res A 2005; 71:143-53. [PMID: 15368264 DOI: 10.1002/jbm.a.30144] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The objectives of this study were to develop a serum-free system for culturing canine oral keratinocytes, the construction and characterization of a canine ex vivo produced oral mucosa equivalent (EVPOME), and transduction green fluorescent protein (GFP) into keratinocytes as a post-grafting tracking marker. Dissociated canine buccal mucosa keratinocytes were cultured in a chemically defined serum-free medium, Epilife trade mark. First-passage keratinocytes were transfected with the GFP gene using a lentiviral vector, sorted by flow cytometer and seeded onto a dermal equivalent, AlloDerm(R) to form EVPOMEs. The EVPOME was characterized by histology and immunohistochemistry, for p63, Ki-67, and involucrin. Laser confocal microscopy was used to locate GFP-transfected keratinocytes within the EVPOME. Cultured canine oral keratinocytes grew rapidly over the first three passages and then the proliferative rate decreased. The canine EVPOME formed a well-stratified epithelial layer. The majority of p63 and Ki-67 immunopositive cells were located in the basal layer whereas cytoplasmic involucrin expression was seen in the suprabasal layers, similar to native canine buccal mucosa. Under laser confocal microscopy, significant green fluorescence was observed throughout the EVPOME. In conclusion, canine EVPOMEs were successfully fabricated in a defined serum-free system with similar characteristics to native buccal mucosa. GFP-transfected canine oral keratinocytes could be identified within the EVPOME.
Collapse
Affiliation(s)
- Junhui Song
- Department of Oral and Maxillofacial Surgery, University of Michigan Medical Center, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109-0018, USA
| | | | | | | |
Collapse
|
27
|
Woodley DT, Keene DR, Atha T, Huang Y, Ram R, Kasahara N, Chen M. Intradermal injection of lentiviral vectors corrects regenerated human dystrophic epidermolysis bullosa skin tissue in vivo. Mol Ther 2005; 10:318-26. [PMID: 15294178 DOI: 10.1016/j.ymthe.2004.05.016] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Accepted: 05/10/2004] [Indexed: 11/21/2022] Open
Abstract
Dystrophic epidermolysis bullosa (DEB) is a family of inherited mechanobullous disorders caused by mutations in the gene, COL7A1, that codes for type VII, (anchoring fibril), collagen, which is critical for epidermal-dermal adherence. Most gene therapy approaches have been ex vivo, involving cell culture and culture graft transplantation, which is logistically difficult. To develop a more simplified approach, we engineered a self-inactivating lentiviral vector expressing human type VII collagen and injected this vector intradermally into hairless, immunodeficient mice and into a human DEB composite skin equivalent grafted onto immunodeficient mice. In both situations, the vector transduced dermal cells, which in turn synthesized and exported type VII collagen into the extracellular space. Remarkably, the type VII collagen selectively adhered to and incorporated into the basement membrane zone (BMZ) between the dermis and the epidermis, where it formed anchoring fibril structures. In the case of the DEB skin equivalent, the newly expressed type VII collagen reversed the DEB phenotype characterized by poor epidermal-dermal adherence and anchoring fibril defects. A single lentiviral vector injection provided stable type VII collagen at the BMZ for at least 3 months. These data demonstrate efficient and long-term type VII collagen gene transfer in vivo using direct intradermal injection of an engineered lentiviral vector.
Collapse
Affiliation(s)
- David T Woodley
- Division of Dermatology, Department of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Hengge UR, Bardenheuer W, Doroudi R, Mirmohammadsadegh A. Thérapie génique et peau. Ann Dermatol Venereol 2005; 132:154-63. [PMID: 15798569 DOI: 10.1016/s0151-9638(05)79230-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- U-R Hengge
- Département de Dermatologie, Université Heinrich-Heine, Duesseldorf, Allemagne.
| | | | | | | |
Collapse
|
29
|
Abstract
Significant progress has been made in corrective gene therapy of the skin in the last decade. This includes advances in vector technology, targeted gene expression, gene replacement, gene correction, and the availability of appropriate animal models for a variety of candidate diseases. While non-viral integration of large genes such as essential basement membrane proteins has been mastered, new challenges such as the control of immune responses lie ahead of the research community until skin gene therapy will become clinical reality. Among the first skin diseases patients with junctional epidermolysis bullosa and xeroderma pigmentosum have entered clinical trials.
Collapse
Affiliation(s)
- Ulrich R Hengge
- Department of Dermatology, Heinrich-Heine-University, D-40225 Duesseldorf, Germany.
| |
Collapse
|
30
|
Del Rio M, Gache Y, Jorcano JL, Meneguzzi G, Larcher F. Current approaches and perspectives in human keratinocyte-based gene therapies. Gene Ther 2004; 11 Suppl 1:S57-63. [PMID: 15454958 DOI: 10.1038/sj.gt.3302370] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Inherited and acquired disorders are liable to treatment with somatic gene therapy. The skin, and in particular epidermal cells, are particularly suited to genetic manipulation and follow-up of therapeutic effects. Cutaneous gene therapy may be effective for skin defects and systemic abnormalities. The robust basic and preclinical data available today would support the application of keratinocyte-based gene therapy to patients.
Collapse
Affiliation(s)
- M Del Rio
- Epithelial Damage, Repair and Tissue Engineering Project. CIEMAT. Avenida Complutense 22, Madrid, Spain
| | | | | | | | | |
Collapse
|
31
|
Abstract
Systemic administration of therapeutic proteins has value in treating a wide variety of disorders, including erythropoietin (Epo)-responsive anemias. Recombinant proteins, however, are costly and require repeated injections, while gene delivery approaches have suffered from inefficiency and difficulties with regulation. The skin effectively delivers polypeptides to the circulation, and improved approaches would support sustainable, topically regulated protein expression after a single vector injection. Toward this goal, we generated lentivectors in which both gene delivery and persistence in skin are regulated by administration of distinct steroid ligands. Following a single injection of regulated lentivector into human skin regenerated on immunodeficient mice, topical glucocorticoid ligands regulated Epo levels and hematocrit over time. Abrogation of gene delivery was achieved by both glucocorticoid cessation and proviral excision via a 4-hydroxytamoxifen-inducible Cre recombinase. These findings establish an approach to durable, topically controlled systemic delivery of therapeutic proteins from human skin tissue.
Collapse
|
32
|
Ghazizadeh S, Katz AB, Harrington R, Taichman LB. Lentivirus-mediated gene transfer to human epidermis. J Investig Dermatol Symp Proc 2004; 9:269-75. [PMID: 15369223 DOI: 10.1111/j.1087-0024.2004.09302.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
For long-term cutaneous gene therapy, the therapeutic gene must be targeted to stem cells and be stably transmitted to and expressed in descendant cells. Retroviral vectors are highly efficient in gene transfer to human keratinocyte stem cells in culture; however, they cannot transduce quiescent stem cells in vivo. As lentiviral vectors (LVV) transduce non-proliferating cells, their ability to target human epidermal stem cells was evaluated. LVV were highly efficient in gene transfer to clonogenic keratinocytes in vitro. Despite higher transgene DNA content and comparable levels of transgene mRNA, levels of transgene product directed by lentivectors were 3-folds lower than that of retrovectors. When transduced keratinocytes were grafted onto mice, transgene expression persisted for at least 20 wk; however, transgene product was detected primarily in the uppermost layers of epidermis. Inclusion of an element that is known to facilitate nuclear export of intron-less transcripts, resulted in enhanced transgene expression in keratinocytes. In vivo transduction of xenografted human skin with these vectors resulted in efficient gene transfer to epidermal progenitor cells. These results demonstrate stem cell transduction by LVV and point out the utility of using these vectors for direct gene transfer to and sustained expression in human epidermis.
Collapse
Affiliation(s)
- Soosan Ghazizadeh
- Department of Oral Biology and Pathology, Stony Brook University, Stony Brook, New York, USA.
| | | | | | | |
Collapse
|
33
|
Dunlap S, Yu X, Cheng L, Civin CI, Alani RM. High-efficiency stable gene transduction in primary human melanocytes using a lentiviral expression system. J Invest Dermatol 2004; 122:549-51. [PMID: 15009744 DOI: 10.1046/j.0022-202x.2004.22214.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
34
|
Abstract
Human immunodeficiency virus type I (HIV) is the etiologic agent of acquired immunodeficiency syndrome or AIDS. Vectors based upon HIV have been in use for over a decade. Beginning in 1996, with the demonstration of improved pseudotyping using vesicular stomatitis virus (VSV) G protein along with transduction of resting mammalian cells, a series of improvements have been made in these vectors, making them both safer and more efficacious. Taking a cue from vector development of murine leukemia virus (MLV), split coding and self-inactivating HIV vectors now appear quite suitable for phase I clinical trials. In parallel, a number of pre-clinical efficacy studies in animals have demonstrated the utility of these vectors for various diseases processes, especially neurodegenerative and hematopoietic illnesses. These vectors are also appropriate for the study of other viruses (specifically of viral entry) and investigation of the HIV replicative cycle, along with straightforward transgene delivery to target cells of interest. Vectors based upon other lentiviruses have shown similar abilities and promise. Although concerns remain, particularly with regards to detection and propagation of replication-competent lentivirus, it is almost certain that these vectors will be introduced into the clinic within the next 3-5 years.
Collapse
Affiliation(s)
- Ricardo Quinonez
- Department of Molecular Virology and Microbiology, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030, USA
| | | |
Collapse
|
35
|
Sawamura D, Akiyama M, Shimizu H. Direct injection of naked DNA and cytokine transgene expression: implications for keratinocyte gene therapy. Clin Exp Dermatol 2002; 27:480-4. [PMID: 12372091 DOI: 10.1046/j.1365-2230.2002.01101.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Intradermally injected DNA diffuses into the epidermis and can then enter keratinocytes and become expressed by these cells. Using this method, plasmids containing cytokine genes that have been introduced into keratinocytes can induce a level of cytokine expression sufficient to provide biological effects in the treated skin. Furthermore, transgenic cytokines released from the transduced keratinocytes can also enter the circulation and have downstream effects on other target organs. Thus far, naked DNA injection appears to be a safe, simple, and relatively efficient method that enables genes to be expressed in transplanted human skin on immunosuppressed animals. In humans, keratinocyte gene therapy using the cytokine gene DNA injection method has the potential to become a powerful therapeutic tool for dermatologists in the management of certain inflammatory and other dermatoses.
Collapse
Affiliation(s)
- D Sawamura
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan.
| | | | | |
Collapse
|
36
|
Abstract
Recent progress in molecular genetics has illuminated the basis for a wide variety of inherited and acquired diseases. Gene therapy offers an attractive therapeutic approach capitalizing upon these new mechanistic insights. The skin is a uniquely attractive tissue site for development of new genetic therapeutic approaches both for its accessibility as well as for the large number of diseases that are amenable in principle to cutaneous gene transfer. Amongst these opportunities are primary monogenic skin diseases, chronic wounds and systemic disorders characterized by low or absent levels of circulating polypeptides. For cutaneous gene therapy to be effective, however, significant progress is required in a number of domains. Recent advances in vector design, administration, immune modulation, and regulation of gene expression have brought the field much nearer to clinical utility.
Collapse
Affiliation(s)
- P A Khavari
- VA Palo Alto Healthcare System and the Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.
| | | | | |
Collapse
|
37
|
Cao T, Tsai SY, O'Malley BW, Wang XJ, Roop DR. The epidermis as a bioreactor: topically regulated cutaneous delivery into the circulation. Hum Gene Ther 2002; 13:1075-80. [PMID: 12067440 DOI: 10.1089/104303402753812476] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Previous studies have documented that the skin can be used as a bioreactor to produce proteins for systemic release to treat diseases. A gene-switch system has been developed that allows regulated expression of therapeutic genes. To determine whether this system could be used in the skin, we developed a transgenic mouse model in which expression of a therapeutic gene could be topically induced in epidermal keratinocytes. After a single induction, high levels of the therapeutic protein, human growth hormone (hGH), were released from keratinocytes into the circulation. The serum levels of hGH were dependent on the amount of inducer applied, and repeated induction resulted in increased weight gain by transgenic versus control mice. Furthermore, physiological levels of hGH were detected in the serum of nude mice after topical induction of small transgenic skin grafts. These results clearly demonstrate the feasibility of using the gene-switch system to regulate the delivery of therapeutic proteins into the circulation via genetically modified keratinocytes.
Collapse
Affiliation(s)
- Tongyu Cao
- Pacific Biomedical Research Center, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | | | | | | | | |
Collapse
|
38
|
Abstract
The last two years have seen new tissue-engineered skin substitutes come onto the market and begin to resolve the various roles to which each is best suited. It is becoming evident that some of the very expensive cell-based products have cost-benefit advantage despite their high price and are valuable within the restricted applications for which they are intended. The use of skin substitutes for testing purposes has extended from epidermal keratinocytes to other integumentary epithelia and into preparations containing multiple cell types in which reactions resulting from paracrine interactions can be examined. Challenges remain in the application of gene therapy techniques to skin substitutes, both the control of transgene expression and in the selection of suitable genes to transfect. A coming challenge is the production of tissue-engineered products without the use of animal products other than human cells. A challenge that may be diminishing is the importance of acute rejection of allogeneic tissue-engineered skin substitutes.
Collapse
Affiliation(s)
- Jonathan Mansbridge
- Advanced Tissue Sciences, 10933 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
39
|
Suzuki I, Im S, Tada A, Scott C, Akcali C, Davis MB, Barsh G, Hearing V, Abdel-Malek Z. Participation of the melanocortin-1 receptor in the UV control of pigmentation. J Investig Dermatol Symp Proc 1999; 4:29-34. [PMID: 10537004 DOI: 10.1038/sj.jidsp.5640177] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The cloning of the melanocortin-1 receptor (MC1R) gene from human melanocytes and the demonstration that these cells respond to the melanocortins alpha-melanocyte stimulating hormone (alpha-MSH) and adrenocorticotropic hormone (ACTH) with increased proliferation and melanogenesis have renewed the interest in investigation the physiological role of these hormones in regulating human pigmentation. Alpha-melanocyte stimulating hormone and ACTH are both synthesized in the human epidermis, and their synthesis is upregulated by exposure to ultraviolet radiation (UVR). Activation of the MC1R by ligand binding results in stimulation of cAMP formation, which is a principal mechanism for inducing melanogenesis. The increase in cAMP is required for the pigmentary response of human melanocytes to UVR, and for allowing them to overcome the UVR-induced G1 arrest. Treatment of human melanocytes with alpha-MSH increases eumelanin synthesis, an effect that is expected to enhance photoprotection of the skin. Population studies have revealed more than 20 allelic variants of the MC1R gene. Some of these variants are overexpressed in individuals with skin type I or II, red hair, and poor tanning ability. Future studies will aim at further exploration of the role of these variants in MC1R function, and in determining constitutive human pigmentation, the response to sun exposure, and possibly the susceptibility to skin cancer.
Collapse
Affiliation(s)
- I Suzuki
- Department of Dermatology, University of Cincinnati, Ohio 45267-0592, USA
| | | | | | | | | | | | | | | | | |
Collapse
|