1
|
Pan Y, Liu Z, Zhang H. Research progress of lens zonules. ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2023; 3:80-85. [PMID: 37846380 PMCID: PMC10577871 DOI: 10.1016/j.aopr.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/23/2023] [Accepted: 02/23/2023] [Indexed: 10/18/2023]
Abstract
Background The lens zonule, a circumferential system of fibres connecting the ciliary body to the lens, is responsible for centration of the lens. The structural, functional, and positional abnormalities of the zonular apparatus can lead to the abnormality of the intraocular structure, presenting a significant challenge to cataract surgery. Main text The lens zonule is the elaborate system of extracellular fibers, which not only centers the lens in the eye but also plays an important role in accommodation and lens immunity, maintains the shape of the lens, and corrects spherical aberration. The zonules may directly participate in the formation of cataract via the immune mechanism. Abnormal zonular fibers that affect the position and shape of the lens may play an important role in the pathogenesis of angle closure disease and increase the complexity of the surgery. Capsular tension rings and related endocapsular devices are used to provide sufficient capsular bag stabilization and ensure the safety of cataract surgery procedures. Better preoperative and intraoperative evaluation methods for zonules are needed for clinicians. Conclusions The microstructure, biomechanical properties, and physiological functions of the lens zonules help us to better understand the pathogenesis of cataract and glaucoma, facilitating the development of safer surgical procedures for cataract. Further studies are needed to carefully analyze the structure-function relationship of the zonular apparatus to explore new treatment strategies for cataract and glaucoma.
Collapse
Affiliation(s)
- Yingying Pan
- Department of Ophthalmology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Zhaoqiang Liu
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Han Zhang
- Department of Ophthalmology, Shandong Provincial Hospital, Shandong University, Jinan, China
| |
Collapse
|
2
|
Johnson J, Kim SY, Sam PK, Asokan R, Cari EL, Bales ES, Luu TH, Perez L, Kallen AN, Nel-Themaat L, Polotsky AJ, Post MD, Orlicky DJ, Jordan KR, Bitler BG. Expression and T cell regulatory action of the PD-1 immune checkpoint in the ovary and fallopian tube. Am J Reprod Immunol 2023; 89:e13649. [PMID: 36394352 PMCID: PMC10559227 DOI: 10.1111/aji.13649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 10/06/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022] Open
Abstract
PROBLEM Immune cell trafficking and surveillance within the ovary and fallopian tube are thought to impact fertility and also tumorigenesis in those organs. However, little is known of how native cells of the ovary and fallopian tube interact with resident immune cells. Interaction of the Programmed Cell Death Protein-1 (PD-1/PDCD-1/CD279) checkpoint with PD-L1 is associated with downregulated immune response. We have begun to address the question of whether PD-1 ligand or its receptors (PD-L1/-L2) can regulate immune cell function in these tissues of the female reproductive tract. METHOD OF STUDY PD-1 and ligand protein expression was evaluated in human ovary and fallopian tube specimens, the latter of which included stages of tubal cell transformation and early tumorigenesis. Ovarian expression analysis included the determination of the proteins in human follicular fluid (HFF) specimens collected during in vitro fertilization procedures. Finally, checkpoint bioactivity of HFF was determined by treatment of separately-isolated human T cells and the measurement of interferon gamma (IFNγ). RESULTS We show that membrane bound and soluble variants of PD-1 and ligands are expressed by permanent constituent cell types of the human ovary and fallopian tube, including granulosa cells and oocytes. PD-1 and soluble ligands were present in HFF at bioactive levels that control T cell PD-1 activation and IFNγ production; full-length checkpoint proteins were found to be highly enriched in HFF exosome fractions. CONCLUSION The detection of PD-1 checkpoint proteins in the human ovary and fallopian tube suggests that the pathway is involved in immunomodulation during folliculogenesis, the window of ovulation, and subsequent egg and embryo immune-privilege. Immunomodulatory action of receptor and ligands in HFF exosomes is suggestive of an acute checkpoint role during ovulation. This is the first study in the role of PD-1 checkpoint proteins in human tubo-ovarian specimens and the first examination of its potential regulatory action in the contexts of normal and assisted reproduction.
Collapse
Affiliation(s)
- Joshua Johnson
- University of Colorado Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Building RC2, Room P15 3103, Mail Stop 8613, Aurora, Colorado 80045
- University of Colorado Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, 12631 East 17th Avenue, Room 4409, B198-3 Aurora, Colorado 80045
| | - So-Youn Kim
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, 985860 Nebraska Medical Center, Omaha, Nebraska 68198
| | | | - Rengasamy Asokan
- University of Colorado Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Building RC2, Room P15 3103, Mail Stop 8613, Aurora, Colorado 80045
| | - Evelyn Llerena Cari
- University of Colorado Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Building RC2, Room P15 3103, Mail Stop 8613, Aurora, Colorado 80045
- University of Colorado Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, 12631 East 17th Avenue, Room 4409, B198-3 Aurora, Colorado 80045
| | - Elise S. Bales
- University of Colorado Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Building RC2, Room P15 3103, Mail Stop 8613, Aurora, Colorado 80045
| | - Thanh-Ha Luu
- University of Colorado Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Building RC2, Room P15 3103, Mail Stop 8613, Aurora, Colorado 80045
- University of Colorado Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, 12631 East 17th Avenue, Room 4409, B198-3 Aurora, Colorado 80045
| | | | | | - Liesl Nel-Themaat
- University of Colorado Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, 12631 East 17th Avenue, Room 4409, B198-3 Aurora, Colorado 80045
- Shady Grove Fertility – Colorado, Denver, CO
| | - Alex J. Polotsky
- University of Colorado Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Building RC2, Room P15 3103, Mail Stop 8613, Aurora, Colorado 80045
- University of Colorado Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, 12631 East 17th Avenue, Room 4409, B198-3 Aurora, Colorado 80045
- Shady Grove Fertility – Colorado, Denver, CO
| | - Miriam D. Post
- University of Colorado Anschutz Medical Campus, Department of Pathology, Mailstop F768, 12605 East 16th Avenue, Aurora, Colorado 80045
| | - David J. Orlicky
- University of Colorado Anschutz Medical Campus, Department of Pathology, Mailstop F768, 12605 East 16th Avenue, Aurora, Colorado 80045
| | - Kimberly R. Jordan
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Human Immunology and Immunotherapy Initiative, Human Immune Monitoring Shared Resource, RC1-North, 8113, Aurora, Colorado 80045
| | - Benjamin G. Bitler
- University of Colorado Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Building RC2, Room P15 3103, Mail Stop 8613, Aurora, Colorado 80045
| |
Collapse
|
3
|
Hill MA, Kwon JH, Gerry B, Hardy WA, Walkowiak OA, Kavarana MN, Nadig SN, Rajab TK. Immune Privilege of Heart Valves. Front Immunol 2021; 12:731361. [PMID: 34447390 PMCID: PMC8383064 DOI: 10.3389/fimmu.2021.731361] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 07/22/2021] [Indexed: 01/22/2023] Open
Abstract
Immune privilege is an evolutionary adaptation that protects vital tissues with limited regenerative capacity from collateral damage by the immune response. Classical examples include the anterior chamber of the eye and the brain. More recently, the placenta, testes and articular cartilage were found to have similar immune privilege. What all of these tissues have in common is their vital function for evolutionary fitness and a limited regenerative capacity. Immune privilege is clinically relevant, because corneal transplantation and meniscal transplantation do not require immunosuppression. The heart valves also serve a vital function and have limited regenerative capacity after damage. Moreover, experimental and clinical evidence from heart valve transplantation suggests that the heart valves are spared from alloimmune injury. Here we review this evidence and propose the concept of heart valves as immune privileged sites. This concept has important clinical implications for heart valve transplantation.
Collapse
Affiliation(s)
- Morgan Ashley Hill
- College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jennie H Kwon
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Brielle Gerry
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - William A Hardy
- College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Olivia Agata Walkowiak
- College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Minoo N Kavarana
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Satish N Nadig
- Division of Transplant Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - T Konrad Rajab
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
4
|
Liu C, Wang X, Huang W, Meng W, Su Z, Xing Q, Shi H, Zhang D, Zhou M, Zhao Y, Wang H, Pan G, Zhong X, Pei D, Guo Y. Hypoproliferative human neural progenitor cell xenografts survived extendedly in the brain of immunocompetent rats. Stem Cell Res Ther 2021; 12:376. [PMID: 34215315 PMCID: PMC8254296 DOI: 10.1186/s13287-021-02427-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 05/31/2021] [Indexed: 11/25/2022] Open
Abstract
Background There is a huge controversy about whether xenograft or allograft in the “immune-privileged” brain needs immunosuppression. In animal studies, the prevailing sophisticated use of immunosuppression or immunodeficient animal is detrimental for the recipients, which results in a short lifespan of animals, confounds functional behavioral readout of the graft benefits, and discourages long-term follow-up. Methods Neuron-restricted neural progenitor cells (NPCs) were derived from human embryonic stem cells (ESCs, including H1, its gene-modified cell lines for better visualization, and HN4), propagated for different passages, and then transplanted into the brain of immunocompetent rats without immunosuppressants. The graft survivals, their cell fates, and HLA expression levels were examined over time (up to 4 months after transplantation). We compared the survival capability of NPCs from different passages and in different transplantation sites (intra-parenchyma vs. para- and intra-cerebroventricle). The host responses to the grafts were also investigated. Results Our results show that human ESC-derived neuron-restricted NPCs survive extendedly in adult rat brain parenchyma with no need of immunosuppression whereas a late-onset graft rejection seems inevitable. Both donor HLA antigens and host MHC-II expression level remain relatively low with little change over time and cannot predict the late-onset rejection. The intra-/para-cerebroventricular human grafts are more vulnerable to the immune attack than the intrastriatal counterparts. Prevention of graft hyperplasia by using hypoproliferative late passaged human NPCs further significantly extends the graft survival time. Our new data also shows that a subpopulation of host microglia upregulate MHC-II expression in response to the human graft, but fail to present the human antigen to the host immune system, suggestive of the immune-isolation role of the blood–brain barrier (BBB). Conclusions The present study confirms the “immune privilege” of the brain parenchyma and, more importantly, unveils that choosing hypoproliferative NPCs for transplantation can benefit graft outcome in terms of both lower tumor-genic risk and the prolonged survival time without immunosuppression. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02427-1.
Collapse
Affiliation(s)
- Chunhua Liu
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, 510005, China
| | - Xiaoyun Wang
- Guangdong Work Injury Rehabilitation Center, Guangzhou, 510440, China
| | - Wenhao Huang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China
| | - Wei Meng
- Guangdong Work Injury Rehabilitation Center, Guangzhou, 510440, China
| | - Zhenghui Su
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China
| | - Qi Xing
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China
| | - Heng Shi
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| | - Di Zhang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China
| | - Min Zhou
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China
| | - Yifan Zhao
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, 510005, China
| | - Haitao Wang
- CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China
| | - Guangjin Pan
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China
| | - Xiaofen Zhong
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China.
| | - Duanqing Pei
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China. .,Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, 510005, China.
| | - Yiping Guo
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China. .,Drug Discovery Pipeline, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, China.
| |
Collapse
|
5
|
Bergman JE, Davies C, Denton AJ, Ashman PE, Mittal R, Eshraghi AA. Advancements in Stem Cell Technology and Organoids for the Restoration of Sensorineural Hearing Loss. J Am Acad Audiol 2021; 32:636-645. [PMID: 34034344 DOI: 10.1055/s-0041-1728677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Sensorineural hearing loss (SNHL) is a significant cause of morbidity worldwide and currently has no curative treatment. Technological advancements in stem cell therapy have led to numerous studies that examine the generation of otic sensory cells from progenitors to restore inner ear function. Recently, organoids have emerged as a promising technique to further advance the process of creating functional replacement cells after irreversible hearing loss. Organoids are the three-dimensional generation of stem cells in culture to model the tissue organization and cellular components of the inner ear. Organoids have emerged as a promising technique to create functioning cochlear structures in vitro and may provide crucial information for the utilization of stem cells to restore SNHL. PURPOSE The purpose of this review is to discuss the recent advancements in stem cell-based regenerative therapy for SNHL. RESULTS Recent studies have improved our understanding about the developmental pathways involved in the generation of hair cells and spiral ganglion neurons. However, significant challenges remain in elucidating the molecular interactions and interplay required for stem cells to differentiate and function as otic sensory cells. A few of the challenges encountered with traditional stem cell therapy may be addressed with organoids. CONCLUSION Stem cell-based regenerative therapy holds a great potential for developing novel treatment modalities for SNHL. Further advancements are needed in addressing the challenges associated with stem cell-based regenerative therapy and promote their translation from bench to bedside.
Collapse
Affiliation(s)
- Jenna E Bergman
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| | - Camron Davies
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| | - Alexa J Denton
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| | - Peter E Ashman
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| | - Rahul Mittal
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| | - Adrien A Eshraghi
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida.,Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Department of Biomedical Engineering, University of Miami, Coral Gables, Miami, Florida.,Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
6
|
Chiaramonte M, Arizza V, La Rosa S, Queiroz V, Mauro M, Vazzana M, Inguglia L. Allograft Inflammatory Factor AIF-1: early immune response in the Mediterranean sea urchin Paracentrotus lividus. ZOOLOGY 2020; 142:125815. [DOI: 10.1016/j.zool.2020.125815] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/24/2022]
|
7
|
Barboza LCM, Lezirovitz K, Zanatta DB, Strauss BE, Mingroni-Netto RC, Oiticica J, Haddad LA, Bento RF. Transplantation and survival of mouse inner ear progenitor/stem cells in the organ of Corti after cochleostomy of hearing-impaired guinea pigs: preliminary results. Braz J Med Biol Res 2016; 49:e5064. [PMID: 27007652 PMCID: PMC4819408 DOI: 10.1590/1414-431x20155064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/16/2015] [Indexed: 11/30/2022] Open
Abstract
In mammals, damage to sensory receptor cells (hair cells) of the inner ear results in permanent sensorineural hearing loss. Here, we investigated whether postnatal mouse inner ear progenitor/stem cells (mIESCs) are viable after transplantation into the basal turns of neomycin-injured guinea pig cochleas. We also examined the effects of mIESC transplantation on auditory functions. Eight adult female Cavia porcellus guinea pigs (250-350g) were deafened by intratympanic neomycin delivery. After 7 days, the animals were randomly divided in two groups. The study group (n=4) received transplantation of LacZ-positive mIESCs in culture medium into the scala tympani. The control group (n=4) received culture medium only. At 2 weeks after transplantation, functional analyses were performed by auditory brainstem response measurement, and the animals were sacrificed. The presence of mIESCs was evaluated by immunohistochemistry of sections of the cochlea from the study group. Non-parametric tests were used for statistical analysis of the data. Intratympanic neomycin delivery damaged hair cells and increased auditory thresholds prior to cell transplantation. There were no significant differences between auditory brainstem thresholds before and after transplantation in individual guinea pigs. Some mIESCs were observed in all scalae of the basal turns of the injured cochleas, and a proportion of these cells expressed the hair cell marker myosin VIIa. Some transplanted mIESCs engrafted in the cochlear basilar membrane. Our study demonstrates that transplanted cells survived and engrafted in the organ of Corti after cochleostomy.
Collapse
Affiliation(s)
- L C M Barboza
- Departamento de Otorrinolaringologia (LIM32), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - K Lezirovitz
- Departamento de Otorrinolaringologia (LIM32), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - D B Zanatta
- Setor de Vetores Virais, Laboratório de Genética e Cardiologia Molecular, Instituto do Coração, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - B E Strauss
- Setor de Vetores Virais, Laboratório de Genética e Cardiologia Molecular, Instituto do Coração, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - R C Mingroni-Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - J Oiticica
- Departamento de Otorrinolaringologia (LIM32), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - L A Haddad
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - R F Bento
- Departamento de Otorrinolaringologia (LIM32), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
8
|
|
9
|
Thompson CD, Zurko JC, Hanna BF, Hellenbrand DJ, Hanna A. The therapeutic role of interleukin-10 after spinal cord injury. J Neurotrauma 2013; 30:1311-24. [PMID: 23731227 DOI: 10.1089/neu.2012.2651] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating condition affecting 270,000 people in the United States. A potential treatment for decreasing the secondary inflammation, excitotoxic damage, and neuronal apoptosis associated with SCI, is the anti-inflammatory cytokine interleukin-10. The best characterized effects of IL-10 are anti-inflammatory-it downregulates pro-inflammatory species interleukin-1β (IL-1β), interleukin-2 (IL-2), interleukin-6 (IL-6), tumor necrosis factor-α, interferon-γ, matrix metalloproteinase-9, nitric oxide synthase, myeloperoxidase, and reactive oxygen species. Pro-apoptotic factors cytochrome c, caspase 3, and Bax are downregulated by IL-10, whereas anti-apoptotic factors B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X, B-cell lymphoma-extra large (Bcl-xl) are upregulated by IL-10. IL-10 also provides trophic support to neurons through the IL-10 receptor. Increased tissue sparing, functional recovery, and neuroprotection are seen with an immediate post-SCI systemic administration of IL-10. Treatment of SCI with IL-10 has been used successfully in combination with Schwann cell and olfactory glial cell grafts, as well as methylprednisolone. Minocycline, tetramethylpyrazine, and hyperbaric oxygen treatment all increase IL-10 levels in a SCI models and result in increased tissue sparing and functional recovery. A chronic systemic administration of IL-10 does not appear to be beneficial to SCI recovery and causes increased susceptibility to septicemia, pneumonia, and peripheral neuropathy. However, a localized upregulation of IL-10 has been shown to be beneficial and can be achieved by herpes simplex virus gene therapy, injection of poliovirus replicons, or surgical placement of a slow-release compound. IL-10 shows promise as a treatment for SCI, although research on local IL-10 delivery timeline and dosage needs to be expanded.
Collapse
Affiliation(s)
- Colton D Thompson
- Department of Neurological Surgery, University of Wisconsin , Madison, Wisconsin, USA
| | | | | | | | | |
Collapse
|
10
|
Tuning Cross-Presentation of Apoptotic T Cells in Immunopathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 785:27-35. [DOI: 10.1007/978-1-4614-6217-0_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
|
12
|
Zimring JC, Stowell SR, Johnsen JM, Hendrickson JE. Effects of genetic, epigenetic, and environmental factors on alloimmunization to transfused antigens: Current paradigms and future considerations. Transfus Clin Biol 2012; 19:125-31. [PMID: 22682308 DOI: 10.1016/j.tracli.2012.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 03/24/2012] [Indexed: 01/03/2023]
Abstract
Transfused red blood cells, platelets, or coagulation factors have the capacity to induce alloantibodies, which once formed, can be a clinical barrier to future transfusion therapy and/or transplantation. Large observational studies over the last 50 years have characterized some of the general properties of transfusion induced alloimmunization, which appear to vary to a considerable extent from what is generally observed for human responses to other immunogens, such as microbial pathogens and vaccines. Transfused cells and factor only induce immune responses in the minority of recipients. There are data to suggest that differences in the unit may play a role. However, there are clearly differences in recipient biology, as once a recipient makes one antibody they are much more likely to make additional antibodies; indeed, recipients have been categorized as "responder" and "non-responder" by the field. Recent mechanistic studies have begun to define potential causes for such differences in alloimmunization from patient to patient, but much progress needs to be made to understand how, why, and in whom alloimmunization occurs. This review gives a general background on immunology in the context of transfusion, summarizes recent progress in the field, and discusses future directions for exploration. Particular attention is paid to the general concept that the human immune system is melded by the wide range of antigens encountered in our environment, and that the effects of such on the immune system may have a profound effect upon response to transfused cells.
Collapse
Affiliation(s)
- J C Zimring
- Puget Sound Blood Center Research Institute, 1551, Eastlake Avenue E, Seattle, WA 98102, USA.
| | | | | | | |
Collapse
|
13
|
Barnaba V, Paroli M, Piconese S. The ambiguity in immunology. Front Immunol 2012; 3:18. [PMID: 22566903 PMCID: PMC3341998 DOI: 10.3389/fimmu.2012.00018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Accepted: 02/02/2012] [Indexed: 01/07/2023] Open
Abstract
In the present article, we discuss the various ambiguous aspects of the immune system that render this complex biological network so highly flexible and able to defend the host from different external invaders. This ambiguity stems mainly from the property of the immune system to be both protective and harmful. Immunity cannot be fully protective without producing a certain degree of damage (immunopathology) to the host. The balance between protection and tissue damage is, therefore, critical for the establishment of immune homeostasis and protection. In this review, we will consider as ambiguous, various immunological tactics including: (a) the opposing functions driving immune responses, immune-regulation, and contra-regulation, as well as (b) the phenomenon of chronic immune activation as a result of a continuous cross-presentation of apoptotic T cells by dendritic cells. All these plans participate principally to maintain a state of chronic low-level inflammation during persisting infections, and ultimately to favor the species survival.
Collapse
Affiliation(s)
- Vincenzo Barnaba
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma Rome, Italy
| | | | | |
Collapse
|
14
|
Abstract
PURPOSE Systemic metastatic retinal lymphoma (SMRL) is exceptionally rare, as systemic lymphomas most often metastasize to the uvea. We have evaluated a series of SMRL cases to elucidate the clinical and pathological features of SMRL. METHODS The pathological specimens of intraocular lymphomas (IOLs) at the National Eye Institute from 1991 to 2009 were retrospectively reviewed. These cases were diagnosed by cytology, cytokine measurement (ELISA for interleukin (IL)-10 and IL-6 levels) and Immunoglobulin-Heavy (IgH) and T-cell-receptor (TCR) gene analyses. RESULTS There were nine B-cell SMRLs (B-SMRL) among 96 B-cell retinal lymphomas (9.4%) and three T-cell SMRLs (T-SMRL) among five T-cell retinal lymphomas (60%) from a total of 116 IOLs, in which 101 were retinal lymphoma. The original sites were nasopharynx (3), testis (2), skin (2), breast (1), blood (1), retroperitoneum (1), ileo-caecum (1) and stomach (1). Cytology of vitreous samples illustrated atypical lymphoma cells with either B- or T-monoclonality. More B-SMRLs had a high ratio of vitreal IL-10 to IL-6 than T-SMRLs. Molecular pathology demonstrated lymphoma cells with gene rearrangements of IgH in all B-SMRLs and TCR in all T-SMRLs. CONCLUSIONS SMRL and primary retinal lymphoma present with similar clinical manifestations. Systemic T-cell lymphoma invades the retina and vitreous more aggressively than systemic B-cell lymphoma. A diagnosis of SMRL is made when there is a clinical history of systemic lymphoma (particularly from nasopharynx, testis and skin), and lymphoma cells are identified in the vitreous or retina. Molecular analysis is more useful than vitreal cytokine measurement for SMRL diagnosis.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Enzyme-Linked Immunosorbent Assay
- Female
- Gene Rearrangement, B-Lymphocyte/genetics
- Humans
- Immunoglobulin Heavy Chains/genetics
- Interleukin-10/metabolism
- Interleukin-6/metabolism
- Lymphatic Metastasis
- Lymphoma, B-Cell/diagnosis
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/metabolism
- Lymphoma, T-Cell/diagnosis
- Lymphoma, T-Cell/genetics
- Lymphoma, T-Cell/metabolism
- Male
- Middle Aged
- Polymerase Chain Reaction
- Receptors, Antigen, T-Cell/genetics
- Retinal Neoplasms/diagnosis
- Retinal Neoplasms/genetics
- Retinal Neoplasms/metabolism
- Retrospective Studies
Collapse
Affiliation(s)
- Xiaoguang Cao
- Immunopathology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | |
Collapse
|
15
|
Background. FOOTMARKS OF INNATE IMMUNITY IN THE OVARY AND CYTOKERATIN-POSITIVE CELLS AS POTENTIAL DENDRITIC CELLS 2011. [DOI: 10.1007/978-3-642-16077-6_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
16
|
Griffith TS, Brincks EL, Gurung P, Kucaba TA, Ferguson TA. Systemic immunological tolerance to ocular antigens is mediated by TRAIL-expressing CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2010; 186:791-8. [PMID: 21169546 DOI: 10.4049/jimmunol.1002678] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Systemic immunological tolerance to Ag encountered in the eye restricts the formation of potentially damaging immune responses that would otherwise be initiated at other anatomical locations. We previously demonstrated that tolerance to Ag administered via the anterior chamber (AC) of the eye required Fas ligand-mediated apoptotic death of inflammatory cells that enter the eye in response to the antigenic challenge. Moreover, the systemic tolerance induced after AC injection of Ag was mediated by CD8(+) regulatory T cells. This study examined the mechanism by which these CD8(+) regulatory T cells mediate tolerance after AC injection of Ag. AC injection of Ag did not prime CD4(+) T cells and led to increased TRAIL expression by splenic CD8(+) T cells. Unlike wild-type mice, Trail(-/-) or Dr5(-/-) mice did not develop tolerance to Ag injected into the eye, even though responding lymphocytes underwent apoptosis in the AC of the eyes of these mice. CD8(+) T cells from Trail(-/-) mice that were first injected via the AC with Ag were unable to transfer tolerance to naive recipient wild-type mice, but CD8(+) T cells from AC-injected wild-type or Dr5(-/-) mice could transfer tolerance. Importantly, the transferred wild-type (Trail(+/+)) CD8(+) T cells were also able to decrease the number of infiltrating inflammatory cells into the eye; however, Trail(-/-) CD8(+) T cells were unable to limit the inflammatory cell ingress. Together, our data suggest that "helpless" CD8(+) regulatory T cells generated after AC injection of Ag enforce systemic tolerance in a TRAIL-dependent manner to inhibit inflammation in the eye.
Collapse
Affiliation(s)
- Thomas S Griffith
- Department of Urology, University of Iowa, Iowa City, IA 52242, USA.
| | | | | | | | | |
Collapse
|
17
|
Guardiola-Serrano F, Rossin A, Cahuzac N, Lückerath K, Melzer I, Mailfert S, Marguet D, Zörnig M, Hueber AO. Palmitoylation of human FasL modulates its cell death-inducing function. Cell Death Dis 2010; 1:e88. [PMID: 21368861 PMCID: PMC3035908 DOI: 10.1038/cddis.2010.62] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Fas ligand (FasL) is a transmembrane protein that regulates cell death in Fas-bearing cells. FasL-mediated cell death is essential for immune system homeostasis and the elimination of viral or transformed cells. Because of its potent cytotoxic activity, FasL expression at the cell surface is tightly regulated, for example, via processing by ADAM10 and SPPL2a generating soluble FasL and the intracellular fragments APL (ADAM10-processed FasL form) and SPA (SPPL2a-processed APL). In this study, we report that FasL processing by ADAM10 counteracts Fas-mediated cell death and is strictly regulated by membrane localization, interactions and modifications of FasL. According to our observations, FasL processing occurs preferentially within cholesterol and sphingolipid-rich nanodomains (rafts) where efficient Fas–FasL contact occurs, Fas receptor and FasL interaction is also required for efficient FasL processing, and FasL palmitoylation, which occurs within its transmembrane domain, is critical for efficient FasL-mediated killing and FasL processing.
Collapse
Affiliation(s)
- F Guardiola-Serrano
- University of Nice-Sophia Antipolis, Centre National de la Recherche Scientifique, Equipe labelisée La Ligue, Institute of Developmental Biology and Cancer, UMR, Nice, France
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Yang JR, Liao CH, Pang CY, Huang LLH, Lin YT, Chen YL, Shiue YL, Chen LR. Directed Differentiation into Neural Lineages and Therapeutic Potential of Porcine Embryonic Stem Cells in Rat Parkinson's Disease Model. Cell Reprogram 2010; 12:447-61. [DOI: 10.1089/cell.2009.0078] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Jenn-Rong Yang
- Division of Physiology, Livestock Research Institute, Council of Agriculture, Executive Yuan, Tainan, Taiwan, R.O.C
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan, R.O.C
| | - Chia-Hsin Liao
- Department of Research, Buddhist Tzu-Chi General Hospital, Hualien, Taiwan, R.O.C
- Institute of Medical Science, Buddhist Tzu-Chi University, Hualien, Taiwan, R.O.C
| | - Cheng-Yoong Pang
- Department of Research, Buddhist Tzu-Chi General Hospital, Hualien, Taiwan, R.O.C
- Graduate Institute of Clinical Medicine, College of Medicine, Buddhist Tzu-Chi University, Hualien, Taiwan, R.O.C
| | - Lynn Ling-Huei Huang
- Institute of Biotechnology, National Cheng Kung University, Tainan, Taiwan, R.O.C
- Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan, R.O.C
| | - Yu-Ting Lin
- Division of Physiology, Livestock Research Institute, Council of Agriculture, Executive Yuan, Tainan, Taiwan, R.O.C
| | - Yi-Ling Chen
- Division of Physiology, Livestock Research Institute, Council of Agriculture, Executive Yuan, Tainan, Taiwan, R.O.C
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan, R.O.C
| | - Yow-Ling Shiue
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan, R.O.C
| | - Lih-Ren Chen
- Division of Physiology, Livestock Research Institute, Council of Agriculture, Executive Yuan, Tainan, Taiwan, R.O.C
- Institute of Biotechnology, National Cheng Kung University, Tainan, Taiwan, R.O.C
- Institute of Biotechnology, Southern Taiwan University, Tainan, Taiwan, R.O.C
| |
Collapse
|
19
|
Mital P, Kaur G, Dufour JM. Immunoprotective Sertoli cells: making allogeneic and xenogeneic transplantation feasible. Reproduction 2010; 139:495-504. [DOI: 10.1530/rep-09-0384] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The testis as an immune-privileged site allows long-term survival of allogeneic and xenogeneic transplants. Testicular Sertoli cells (SCs) play a major role in this immunoprotection and have been used to create an ectopic immune-privileged environment that prolongs survival of co-transplanted allogeneic and xenogeneic cells, including pancreatic islets and neurons. Extended survival of such grafts testifies to the immunoprotective properties of SCs. However, there is still variability in the survival rates of the co-grafted cells and rarely are 100% of the grafts protected. This emphasizes the need to learn more about what is involved in creating the optimal immunoprotective milieu. Several parameters including organization of the SCs into tubule-like structures and the production of immunomodulatory factors by SCs, specifically complement inhibitors, cytokines, and cytotoxic lymphocyte inhibitors, are likely important. In addition, an intricate interplay between several of these factors may be responsible for providing the most ideal environment for protection of the co-transplants by SCs. In this review, we will also briefly describe a novel use for the immune-privileged abilities of SCs; engineering them to deliver therapeutic proteins for the treatment of diseases like diabetes and Parkinson's disease. In conclusion, further studies and more detailed analysis of the mechanisms involved in creating the immune-protective environment by SCs may make their application in co-transplantation and as engineered cells clinically feasible.
Collapse
|
20
|
Gurung P, Kucaba TA, Ferguson TA, Griffith TS. Activation-induced CD154 expression abrogates tolerance induced by apoptotic cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:6114-23. [PMID: 19841180 DOI: 10.4049/jimmunol.0901676] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The decision to generate a productive immune response or tolerance often depends on the context in which T cells first see Ag. Using a classical system of tolerance induction, we examined the immunological consequence of Ag encountered in the presence of naive or activated apoptotic cells. Naive apoptotic cells induced tolerance when injected i.v.; however, previously activated apoptotic cells induced immunity. Further analysis revealed a key role for CD154, as tolerance resulted after i.v. injection of either naive or activated apoptotic CD154(-/-) T cells, while coinjection of an agonistic anti-CD40 mAb with naive apoptotic T cells induced robust immunity. Dendritic cells fed activated apoptotic T cells in vitro produced IL-12p40 in a CD154-dependent manner, and the use of IL-12p40(-/-) mice or mAb-mediated neutralization of IL-12 revealed a link between CD154, IL-12, and the ability of activated apoptotic T cells to induce immunity rather than tolerance. Collectively, these results show that CD154 expression on apoptotic T cells can determine the outcome of an immune response to Ag recognized within the context of the apoptotic cells and suggest that the balance between naive and activated apoptotic T cells may dictate whether a productive immune response is encouraged.
Collapse
Affiliation(s)
- Prajwal Gurung
- Department of Urology, University of Iowa, Iowa City, IA 52242-1089, USA
| | | | | | | |
Collapse
|
21
|
Dace DS, Khan AA, Stark JL, Kelly J, Cross AH, Apte RS. Interleukin-10 overexpression promotes Fas-ligand-dependent chronic macrophage-mediated demyelinating polyneuropathy. PLoS One 2009; 4:e7121. [PMID: 19771172 PMCID: PMC2743195 DOI: 10.1371/journal.pone.0007121] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 08/19/2009] [Indexed: 12/20/2022] Open
Abstract
Background Demyelinating polyneuropathy is a debilitating, poorly understood disease that can exist in acute (Guillain-Barré syndrome) or chronic forms. Interleukin-10 (IL-10), although traditionally considered an anti-inflammatory cytokine, has also been implicated in promoting abnormal angiogenesis in the eye and in the pathobiology of autoimmune diseases such as lupus and encephalomyelitis. Principal Findings Overexpression of IL-10 in a transgenic mouse model leads to macrophage-mediated demyelinating polyneuropathy. IL-10 upregulates ICAM-1 within neural tissues, promoting massive macrophage influx, inflammation-induced demyelination, and subsequent loss of neural tissue resulting in muscle weakness and paralysis. The primary insult is to perineural myelin followed by secondary axonal loss. Infiltrating macrophages within the peripheral nerves demonstrate a highly pro-inflammatory signature. Macrophages are central players in the pathophysiology, as in vivo depletion of macrophages using clodronate liposomes reverses the phenotype, including progressive nerve loss and paralysis. Macrophage-mediate demyelination is dependent on Fas-ligand (FasL)-mediated Schwann cell death. Significance These findings mimic the human disease chronic idiopathic demyelinating polyneuropathy (CIDP) and may also promote further understanding of the pathobiology of related conditions such as acute idiopathic demyelinating polyneuropathy (AIDP) or Guillain-Barré syndrome.
Collapse
Affiliation(s)
- Dru S. Dace
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
| | - Aslam A. Khan
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
| | - Jennifer L. Stark
- The National Multiple Sclerosis Society, New York, New York, United States of America
| | - Jennifer Kelly
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
| | - Anne H. Cross
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
| | - Rajendra S. Apte
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
22
|
Taming the tiger by the tail: modulation of DNA damage responses by telomeres. EMBO J 2009; 28:2174-87. [PMID: 19629039 PMCID: PMC2722249 DOI: 10.1038/emboj.2009.176] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 06/03/2009] [Indexed: 11/09/2022] Open
Abstract
Telomeres are by definition stable and inert chromosome ends, whereas internal chromosome breaks are potent stimulators of the DNA damage response (DDR). Telomeres do not, as might be expected, exclude DDR proteins from chromosome ends but instead engage with many DDR proteins. However, the most powerful DDRs, those that might induce chromosome fusion or cell-cycle arrest, are inhibited at telomeres. In budding yeast, many DDR proteins that accumulate most rapidly at double strand breaks (DSBs), have important functions in physiological telomere maintenance, whereas DDR proteins that arrive later tend to have less important functions. Considerable diversity in telomere structure has evolved in different organisms and, perhaps reflecting this diversity, different DDR proteins seem to have distinct roles in telomere physiology in different organisms. Drawing principally on studies in simple model organisms such as budding yeast, in which many fundamental aspects of the DDR and telomere biology have been established; current views on how telomeres harness aspects of DDR pathways to maintain telomere stability and permit cell-cycle division are discussed.
Collapse
|
23
|
Abstract
The healthy immune system makes use of a variety of surveillance mechanisms at different stages of lymphoid development to prevent the occurrence and expansion of potentially harmful autoreactive T cell clones. Disruption of these mechanisms may lead to inappropriate activation of T cells and the development of autoimmune and lymphoproliferative diseases [such as multiple sclerosis, rheumatoid arthritis, lupus erythematosus, diabetes and autoimmune lymphoproliferative syndrome (ALPS)]. Clonal deletion of T cells with high affinities for self-peptide-MHC via programmed cell death (apoptosis) is an essential mechanism leading to self-tolerance. Referred to as negative selection, central tolerance in the thymus serves as the first checkpoint for the developing T cell repertoire and involves the apoptotic elimination of potentially autoreactive T cells clones bearing high affinity T cell receptors (TCR) that recognize autoantigens presented by thymic epithelial cells. Autoreactive T cells that escape negative selection are held in check in the periphery by either functional inactivation ("anergy") or extrathymic clonal deletion, both of which are dependent on the strength and frequency of the TCR signal and the costimulatory context, or by regulatory T cells. This review provides an overview of the different molecular executioners of cell death programs that are vital to intrathymic or extrathymic clonal deletion of T cells. Further, the potential involvement of various apoptotic signaling paradigms are discussed with respect to the genesis and pathophysiology of autoimmune disease.
Collapse
Affiliation(s)
- Martina Gatzka
- Department of Molecular Biology and Biochemistry, Center for Immunology, University of California, Irvine, CA, USA.
| | | |
Collapse
|
24
|
Vasil ML, Stonehouse MJ, Vasil AI, Wadsworth SJ, Goldfine H, Bolcome RE, Chan J. A complex extracellular sphingomyelinase of Pseudomonas aeruginosa inhibits angiogenesis by selective cytotoxicity to endothelial cells. PLoS Pathog 2009; 5:e1000420. [PMID: 19424430 PMCID: PMC2673038 DOI: 10.1371/journal.ppat.1000420] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 04/08/2009] [Indexed: 11/19/2022] Open
Abstract
The hemolytic phospholipase C (PlcHR) expressed by Pseudomonas aeruginosa is the original member of a Phosphoesterase Superfamily, which includes phosphorylcholine-specific phospholipases C (PC-PLC) produced by frank and opportunistic pathogens. PlcHR, but not all its family members, is also a potent sphingomyelinase (SMase). Data presented herein indicate that picomolar (pM) concentrations of PlcHR are selectively lethal to endothelial cells (EC). An RGD motif of PlcHR contributes to this selectivity. Peptides containing an RGD motif (i.e., GRGDS), but not control peptides (i.e., GDGRS), block the effects of PlcHR on calcium signaling and cytotoxicity to EC. Moreover, RGD variants of PlcHR (e.g., RGE, KGD) are significantly reduced in their binding and toxicity, but retain the enzymatic activity of the wild type PlcHR. PlcHR also inhibits several EC-dependent in vitro assays (i.e., EC migration, EC invasion, and EC tubule formation), which represent key processes involved in angiogenesis (i.e., formation of new blood vessels from existing vasculature). Finally, the impact of PlcHR in an in vivo model of angiogenesis in transgenic zebrafish, and ones treated with an antisense morpholino to knock down a key blood cell regulator, were evaluated because in vitro assays cannot fully represent the complex processes of angiogenesis. As little as 2 ng/embryo of PlcHR was lethal to approximately 50% of EGFP-labeled EC at 6 h after injection of embryos at 48 hpf (hours post-fertilization). An active site mutant of PlcHR (Thr178Ala) exhibited 120-fold reduced inhibitory activity in the EC invasion assay, and 20 ng/embryo elicited no detectable inhibitory activity in the zebrafish model. Taken together, these observations are pertinent to the distinctive vasculitis and poor wound healing associated with P. aeruginosa sepsis and suggest that the potent antiangiogenic properties of PlcHR are worthy of further investigation for the treatment of diseases where angiogenesis contributes pathological conditions (e.g., vascularization of tumors, diabetic retinopathy).
Collapse
Affiliation(s)
- Michael L Vasil
- Department of Microbiology, University of Colorado Denver, Anschutz Medical Center, Aurora, Colorado, United States of America.
| | | | | | | | | | | | | |
Collapse
|
25
|
Olson MR, Varga SM. Fas ligand is required for the development of respiratory syncytial virus vaccine-enhanced disease. THE JOURNAL OF IMMUNOLOGY 2009; 182:3024-31. [PMID: 19234198 DOI: 10.4049/jimmunol.0803585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Children immunized with a formalin-inactivated respiratory syncytial virus (RSV) vaccine experienced enhanced disease and exhibited pulmonary eosinophilia upon natural RSV infection. BALB/c mice immunized with either formalin-inactivated RSV or a recombinant vaccinia virus (vacv) expressing the RSV attachment (G) protein develop extensive pulmonary eosinophilia after RSV challenge that mimics the eosinophilic response observed in the children during the 1960s vaccine trials. Fas ligand (FasL) is a major immune effector molecule that can contribute to the clearance of respiratory viruses. However, the role of FasL in the development of RSV vaccine-enhanced disease has not been elucidated. RSV challenge of vacvG-immunized gld mice, that lack functional FasL, results in diminished systemic disease as well as pulmonary eosinophilia. The magnitude of the secondary RSV G-specific CD4 T cell response was diminished in gld mice as compared with wild-type controls. Furthermore, we show that CD4 T cells isolated after RSV challenge of vacvG-immunized gld mice exhibit enhanced expression of Annexin V and caspase 3/7 indicating that FasL is important for either the survival or the expansion of virus-specific secondary effector CD4 T cells. Taken together, these data identify a previously undefined role for FasL in the accumulation of secondary effector CD4 T cells and the development of RSV vaccine-enhanced disease.
Collapse
Affiliation(s)
- Matthew R Olson
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
26
|
Montalvo V, Gery I. Constitutive Pro-Apoptotic Molecules are Differentially Expressed in the Fetal and Adult Mouse Eye. Curr Eye Res 2009; 34:328-32. [DOI: 10.1080/02713680902741688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
27
|
Forrester JV, Xu H, Lambe T, Cornall R. Immune privilege or privileged immunity? Mucosal Immunol 2008; 1:372-81. [PMID: 19079201 DOI: 10.1038/mi.2008.27] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Immune privilege is a concept that has come of age. Where previously it was considered to be a passive phenomenon restricted to certain specialized tissues, it is now viewed as comprising several mechanisms, both active and passive, shared in many aspects with emerging notions of the mechanisms of peripheral tolerance. The relative degrees of immune privilege vary from tissue to tissue depending on the number and strength of each of the mechanisms contained in that tissue. Immune privilege can be generated in non-privileged sites such as the skin and allografts, and is a property of the tissue itself. We therefore propose that, in addition to canonical central and peripheral tolerance mechanisms, there is a third route whereby the organism promotes self-antigen non-reactivity centered on the specific properties of each tissue and varying accordingly (relative degrees of immune privilege). This third mechanism of inducing immunological tolerance, as it is a local tissue phenomenon, might have particular therapeutic significance, for instance in devising strategies for induction of immunity to tumors by disrupting immune privilege or in preventing graft rejection by promoting immune privilege.
Collapse
Affiliation(s)
- J V Forrester
- Department of Ophthalmology, University of Aberdeen, Aberdeen, Scotland.
| | | | | | | |
Collapse
|
28
|
Milia AF, Manetti M, Generini S, Polidori L, Benelli G, Cinelli M, Messerini L, Ibba-Manneschi L, Matucci-Cerinic M. TNFalpha blockade prevents the development of inflammatory bowel disease in HLA-B27 transgenic rats. J Cell Mol Med 2008; 13:164-76. [PMID: 18363845 PMCID: PMC3823044 DOI: 10.1111/j.1582-4934.2008.00310.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Rats transgenic for HLA-B27 and human β2microglobulin (B27TR) develop a multi-systemic disease resembling inflammatory bowel disease (IBD) and spondyloarthritis. TNFα has a crucial role in chronic inflammation. Our objective was to evaluate the effect of anti-TNFα treatment on spontaneous IBD in B27TR. Nine-week-old B27TR received monoclonal anti-TNFα or an isotypic IgG2a,k up to age of 18 weeks. A second group was monitored up to 18 weeks and then randomly assigned to anti-TNFα or IgG2 a,k treatment. Each rat was monitored for clinical IBD manifestations. After sacrifice, the colon was examined for pathological changes. TNFα receptors (TNF-R1, TNF-R2), Fas/Fas-L expression and apoptosis were evaluated. IgG2a,k-treated and untreated B27TR presented signs of IBD at 11 weeks, whereas in anti-TNFα-treated B27TR no IBD signs were detected. In the late treatment, IBD signs improved after 1 week. Histopathological analysis of IgG2a,k-treated B27TR colon showed inflammatory signs that were widely prevented by early anti-TNFα treatment. Late treatment did not significantly reduce inflammation. TNF-R1 was weakly expressed in intestinal epithelial cells of IgG2a,k-treated B27TR, while it was comparable to controls in anti-TNFα-treated animals. TNF-R2 immunopositivity was strongly evident in IgG2a,k-treated B27TR, whereas was absent in anti-TNFα-treated rats. RT-PCR confirmed these results. IgG2a,k-treated B27TR showed, at 18 weeks, few Fas-positive cells and an increase of Fas-L-positive cells. At 27 weeks, Fas-/Fas-L-positive cell number was significantly low. Anti-TNFα treatment increased Fas-L expression, whereas Fas increased only with the early treatment. TNFα blockade is effective in preventing inflammation in early phase of IBD, maintaining the homeostatic balance of apoptosis.
Collapse
Affiliation(s)
- Anna Franca Milia
- Department of Biomedicine, Division of Rheumatology, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Zhang J, Yang K, Tan D, Zeng J, Fine A. IL-1β regulates the mouse Fas ligand expression in corneal endothelial cells. CHINESE SCIENCE BULLETIN-CHINESE 2007. [DOI: 10.1007/s11434-007-0334-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
30
|
Brunlid G, Pruszak J, Holmes B, Isacson O, Sonntag KC. Immature and neurally differentiated mouse embryonic stem cells do not express a functional Fas/Fas ligand system. Stem Cells 2007; 25:2551-8. [PMID: 17615270 PMCID: PMC2951385 DOI: 10.1634/stemcells.2006-0745] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The potential of pluripotent embryonic stem (ES) cells to develop into functional cells or tissue provides an opportunity in the development of new therapies for many diseases including neurodegenerative disorders. The survival of implanted cells usually requires systemic immunosuppression, however, which severely compromises the host immune system, leading to complications in clinical transplantation. An optimal therapy would therefore be the induction of specific tolerance to the donor cells, while otherwise preserving functional immune responses. Fas ligand (FasL) is expressed in activated lymphocytes as well as cells in "immune-privileged" sites including the central nervous system. Its receptor, Fas, is expressed on various immune-reactive cell types, such as activated natural killer and T cells, monocytes, and polymorphic mononucleocytes, which can undergo apoptosis upon interaction with FasL. To render transplanted cells tolerant to host cellular immune responses, we genetically engineered mouse ES cells to express rat FasL (rFasL). The rFasL-expressing ES cells were analyzed for survival during in vitro neurodifferentiation and after transplantation to the rat brain without further immunosuppression. Although control transfected HEK-293T cells expressed functional rFasL, immature and differentiated mouse ES cells did not express the recombinant rFasL surface protein. Furthermore, there was no evidence for functional endogenous Fas and FasL expression on either ES cells or on neural cells after in vitro differentiation. Moreover, implanted rFasL-engineered ES cells did not survive in the rat brains in the absence of the immunosuppressive agent cyclosporine A. Our results indicate that immature and differentiated mouse ES cells do not express a functional Fas/FasL system. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Gabriella Brunlid
- Harvard Medical School, Center for Neuroregeneration Research, Udall Parkinson's Disease Center of Excellence, McLean Hospital, 115 Mill Street, Belmont, Massachusetts 02478, USA
| | | | | | | | | |
Collapse
|
31
|
Liu G, Ma H, Jiang L, Zhao Y. Allograft inflammatory factor-1 and its immune regulation. Autoimmunity 2007; 40:95-102. [PMID: 17453710 DOI: 10.1080/08916930601083946] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The allograft inflammatory factor-1 (AIF-1) is a 17 kDa interferon-gamma (IFN-gamma) inducible Ca(2+)-binding EF-hand protein that is encoded within the HLA class III genomic region. Three proteins including ionized Ca(2+)-binding adaptor 1, microglia response factor-1, and daintain are identical with AIF-1. The expression of AIF-1 was mostly limited to the monocyte/macrophage lineage, and augmented by cytokines such as IFN-gamma. It was assumed that AIF-1 was a novel molecule involved in inflammatory responses, allograft rejection, as well as the activation and function of macrophages. However, it has been reported that AIF-1 is also expressed in macrophages and microglial cells in autoimmune diseases such as experimental autoimmune encephalomyelitis, neuritis and uveitis models, suggesting that AIF-1 may play a pivotal role in autoimmunity. In the present manuscript, the genomic and functional characteristics of AIF-1 family proteins as well as their immune regulatory effects are reviewed.
Collapse
Affiliation(s)
- Guangwei Liu
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beisihuan Xi Road 25, Beijing 100080, China
| | | | | | | |
Collapse
|
32
|
Catalano S, Rizza P, Gu G, Barone I, Giordano C, Marsico S, Casaburi I, Middea E, Lanzino M, Pellegrino M, Andò S. Fas ligand expression in TM4 Sertoli cells is enhanced by estradiol "in situ" production. J Cell Physiol 2007; 211:448-56. [PMID: 17167783 DOI: 10.1002/jcp.20952] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The testis is an immunologically privileged site of the body where Sertoli cells work on to favor local immune tolerance by testicular autoantigens segregation and immunosuppressive factors secretion. Fas/Fas Ligand (FasL) system, expressed prevalently in Sertoli cells, has been considered to be one of the central mechanisms in testis immunological homeostasis. In different cell lines it has been reported that the proapoptotic protein FasL is regulated by 17-beta estradiol (E2). Thus, using as experimental model mouse Sertoli cells TM4, which conserve a large spectrum of functional features present in native Sertoli cells, like aromatase activity, we investigated if estradiol "in situ" production may influence FasL expression. Our results demonstrate that an aromatizable androgen like androst-4-ene-3,17-dione (Delta4) enhanced FasL mRNA, protein content and promoter activity in TM4 cells. The treatment with N(6),2'-O-dibutyryladenosine-3'-5'-cyclic monophosphate [(Bu)(2)cAMP] (simulating FSH action), that is well known to stimulate aromatase activity in Sertoli cells, amplified Delta4 induced FasL expression. Functional studies of mutagenesis, electrophoretic mobility shift (EMSA) and chromatin immunoprecipitation (ChIP) assays revealed that the Sp-1 motif on FasL promoter was required for E2 enhanced FasL expression in TM4 cells. These data let us to recruit FasL among those genes whose expression is up-regulated by E2 through a direct interaction of ERalpha with Sp-1 protein. Finally, evidence that an aromatizable androgen is able to increase FasL expression suggests that E2 production by aromatase activity may contribute to maintain the immunoprivilege status of Sertoli cells.
Collapse
Affiliation(s)
- Stefania Catalano
- Department of Pharmaco-Biology, University of Calabria 87036 Arcavacata di Rende (CS), Calabria, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Belegu V, Oudega M, Gary DS, McDonald JW. Restoring function after spinal cord injury: promoting spontaneous regeneration with stem cells and activity-based therapies. Neurosurg Clin N Am 2007; 18:143-68, xi. [PMID: 17244561 DOI: 10.1016/j.nec.2006.10.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although neural regeneration is an active research field today, no current treatments can aid regeneration after spinal cord injury. This article reviews the feasibility of spinal cord repair and provides an overview of the range of strategies scientists are taking toward regeneration. The major focus of this article is the future role of stem cell transplantation and similar rehabilitative restorative approaches designed to optimize spontaneous regeneration by mobilizing endogenous stem cells and facilitating other cellular mechanisms of regeneration, such as axonal growth and myelination.
Collapse
Affiliation(s)
- Visar Belegu
- The International Center for Spinal Cord Injury, Kennedy Krieger Institute, Department of Neurology, Johns Hopkins University School of Medicine, 707 North Broadway, Room 518, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
34
|
Schulte M, Reiss K, Lettau M, Maretzky T, Ludwig A, Hartmann D, de Strooper B, Janssen O, Saftig P. ADAM10 regulates FasL cell surface expression and modulates FasL-induced cytotoxicity and activation-induced cell death. Cell Death Differ 2007; 14:1040-9. [PMID: 17290285 DOI: 10.1038/sj.cdd.4402101] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The apoptosis-inducing Fas ligand (FasL) is a type II transmembrane protein that is involved in the downregulation of immune reactions by activation-induced cell death (AICD) as well as in T cell-mediated cytotoxicity. Proteolytic cleavage leads to the generation of membrane-bound N-terminal fragments and a soluble FasL (sFasL) ectodomain. sFasL can be detected in the serum of patients with dysregulated inflammatory diseases and is discussed to affect Fas-FasL-mediated apoptosis. Using pharmacological approaches in 293T cells, in vitro cleavage assays as well as loss and gain of function studies in murine embryonic fibroblasts (MEFs), we demonstrate that the disintegrin and metalloprotease ADAM10 is critically involved in the shedding of FasL. In primary human T cells, FasL shedding is significantly reduced after inhibition of ADAM10. The resulting elevated FasL surface expression is associated with increased killing capacity and an increase of T cells undergoing AICD. Overall, our findings suggest that ADAM10 represents an important molecular modulator of FasL-mediated cell death.
Collapse
Affiliation(s)
- M Schulte
- Biochemical Institute, Christian-Albrecht-University, Kiel, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Functionally barricaded immune responses or sites of immune privilege are no longer considered dependent on specific anatomical considerations, but rather, they can develop in any location where immunoregulatory cells congregate and express or release products capable of deviating the host response to foreign antigens. Among the pivotal molecules involved in orchestrating these ectopic sites of immune suppression is transforming growth factor-beta (TGF-beta), a secreted and cell-associated polypeptide with a multiplicity of actions in innate and adaptive immunity. While beneficial in initiating and controlling immune responses and maintaining immune homeostasis, immunosuppressive pathways mediated by TGF-beta may obscure immune surveillance mechanisms, resulting in failure to recognize or respond adequately to self, foreign, or tumor-associated antigens. CD4+CD25+Foxp3+ regulatory T cells represent a dominant purveyor of TGF-beta-mediated suppression and are found in infiltrating tumors and other sites of immune privilege, where they influence CD8+ T cells; CD4+ T-helper (Th)1, Th2, and Th17 cells; natural killer cells; and cells of myeloid lineage to choreograph and/or muck up host defense. Defining the cellular sources, mechanisms of action, and networking that distinguish the dynamic establishment of localized immune privilege is vital for developing strategic approaches to diminish or to embellish these tolerogenic events for therapeutic benefit.
Collapse
Affiliation(s)
- Sharon M Wahl
- Oral Infection and Immunity Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4352, USA.
| | | | | |
Collapse
|
36
|
Arshavsky YI. Alzheimer's disease, brain immune privilege and memory: a hypothesis. J Neural Transm (Vienna) 2006; 113:1697-707. [PMID: 16932992 DOI: 10.1007/s00702-006-0524-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Accepted: 04/28/2006] [Indexed: 12/13/2022]
Abstract
The most distinctive feature of Alzheimer's disease (AD) is the specific degeneration of the neurons involved in memory consolidation, storage, and retrieval. Patients suffering from AD forget basic information about their past, loose linguistic and calculative abilities and communication skills. Thus, understanding the etiology of AD may provide insights into the mechanisms of memory and vice versa. The brain is an immunologically privileged site protected from the organism's immune reactions by the blood-brain barrier (BBB). All risk factors for AD (both cardiovascular and genetic) lead to destruction of the BBB. Evidence emerging from recent literature indicates that AD may have an autoimmune nature associated with BBB impairments. This hypothesis suggests that the process of memory consolidation involves the synthesis of novel macromolecules recognized by the immune system as "non-self" antigens. The objective of this paper is to stimulate new approaches to studies of neural mechanisms underpinning memory consolidation and its breakdown during AD. If the hypothesis on the autoimmune nature of AD is correct, the identification of the putative antigenic macromolecules might be critical to understanding the etiology and prevention of AD, as well as for elucidating cellular mechanisms of learning and memory.
Collapse
Affiliation(s)
- Y I Arshavsky
- Institute for Nonlinear Science, University of California San Diego, La Jolla, CA 92093-0402, USA.
| |
Collapse
|
37
|
Koch KS, Son KH, Maehr R, Pellicciotta I, Ploegh HL, Zanetti M, Sell S, Leffert HL. Immune-privileged embryonic Swiss mouse STO and STO cell-derived progenitor cells: major histocompatibility complex and cell differentiation antigen expression patterns resemble those of human embryonic stem cell lines. Immunology 2006; 119:98-115. [PMID: 16836618 PMCID: PMC1782333 DOI: 10.1111/j.1365-2567.2006.02412.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Embryonic mouse STO (S, SIM; T, 6-thioguanine resistant; O, ouabain resistant) and 3(8)21-enhanced green fluorescent protein (EGFP) cell lines exhibit long-term survival and hepatic progenitor cell behaviour after xenogeneic engraftment in non-immunosuppressed inbred rats, and were previously designated major histocompatibility complex (MHC) class I- and class II-negative lines. To determine the molecular basis for undetectable MHC determinants, the expression and haplotype of H-2K, H-2D, H-2L and I-A proteins were reassessed by reverse transcriptase-polymerase chain reaction (RT-PCR), cDNA sequencing, RNA hybridization, immunoblotting, quantitative RT-PCR (QPCR), immunocytochemistry and flow cytometry. To detect cell differentiation (CD) surface antigens characteristic of stem cells, apoptotic regulation or adaptive immunity that might facilitate progenitor cell status or immune privilege, flow cytometry was also used to screen untreated and cytokine [interferon (IFN)-gamma]-treated cultures. Despite prior PCR genotyping analyses suggestive of H-2q haplotypes in STO, 3(8)21-EGFP and parental 3(8)21 cells, all three lines expressed H-2K cDNA sequences identical to those of d-haplotype BALB/c mice, as well as constitutive and cytokine-inducible H-2K(d) determinants. In contrast, apart from H-2L(d[LOW]) display in 3(8)21 cells, H-2Dd, H-2Ld and I-Ad determinants were undetectable. All three lines expressed constitutive and cytokine-inducible CD34; however, except for inducible CD117([LOW]) expression in 3(8)21 cells, no expression of CD45, CD117, CD62L, CD80, CD86, CD90.1 or CD95L/CD178 was observed. Constitutive and cytokine-inducible CD95([LOW]) expression was detected in STO and 3(8)21 cells, but not in 3(8)21-EGFP cells. MHC (class I(+[LOW])/class II-) and CD (CD34+/CD80-/CD86-/CD95L-) expression patterns in STO and STO cell-derived progenitor cells resemble patterns reported for human embryonic stem cell lines. Whether these patterns reflect associations with mechanisms that are regulatory of immune privilege or functional tissue-specific plasticity is unknown.
Collapse
Affiliation(s)
- Katherine S Koch
- Department of Pharmacology, School of Medicine, University of California at San Diego, La Jolla, CA 92093-0636, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Baccar Harrath A, Yacoubi Loueslati B, Troudi W, Hmida S, Sedkaoui S, Dridi A, Jridi A, Ben Ayed F, Ben Rhomdhane K, Ben Ammar Elgaaied A. HLA class II polymorphism: protective or risk factors to breast cancer in Tunisia? Pathol Oncol Res 2006; 12:79-81. [PMID: 16799707 DOI: 10.1007/bf02893448] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Accepted: 03/18/2006] [Indexed: 01/18/2023]
Abstract
HLA system plays a key role in the tumor cells' escape from immune surveillance. Herein is the first report on the correlation of the susceptibility to breast cancer with HLA class II markers in Tunisia. Molecular typing of HLA-DRB1 and -DQB1 loci was undertaken for 70 Tunisian female patients. Comparison of allele and haplotype distribution between patients and 70 female control subjects reveals a negative association between HLADRB1* 07-DQB1*02 and the incidence of breast cancer in the Tunisian population.
Collapse
Affiliation(s)
- Amal Baccar Harrath
- Laboratory of Molecular Genetics, Immunology and Biotechnology, Faculty of Sciences of Tunis, ElManar University, Tunis, 1060, Tunisia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wallace DJ, Altemare CR, Shen DF, deSmet MD, Buggage RR, Nussenblatt RB, Chan CC. Primary testicular and intraocular lymphomas: two case reports and a review of the literature. Surv Ophthalmol 2006; 51:41-50. [PMID: 16414360 PMCID: PMC1930146 DOI: 10.1016/j.survophthal.2005.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Testicular lymphoma is a rare neoplasm of the testis that is most commonly seen in older patients. It metastasizes preferentially to extranodal sites, including the skin, central nervous system, Waldeyer ring, contralateral testis, and lung. Two case reports of patients with a history of testicular lymphoma who developed involvement of the vitreous and retina are presented. These are interesting cases as the testis, central nervous system, and eye are all immune privileged organs, which may account for occurrence of disease in these sites. Histopathologic examination of diagnostic vitrectomy specimens from both cases showed atypical lymphoid cells with immunoglobulin heavy chain (IgH) gene rearrangements, consistent with the diagnosis of intraocular B-cell lymphoma. The results of a literature review of all reports of ocular involvement with testicular lymphoma are discussed. Patients with testicular lymphoma are at risk for relapse, particularly in the central nervous system. Clinicians should be suspicious for intraocular lymphoma in patients with a history of testicular lymphoma who present with vitritis or retinal lesions.
Collapse
Affiliation(s)
| | | | - De Fen Shen
- National Eye Institute, NEI, Bethesda, Maryland, USA
| | - Marc D. deSmet
- Academic Center University of Amsterdam, the Netherlands
| | | | | | - Chi-Chao Chan
- National Eye Institute, NEI, Bethesda, Maryland, USA
| |
Collapse
|
40
|
Abstract
Ever since the discovery of Medawar, over 50 years ago, that immunological tolerance was an acquired phenomenon that could be manipulated in neonatal mice, the ability to induce therapeutic tolerance against autoantigens, allergens and organ grafts has been a major driving force in immunology. Within the last 20 years we have found that a brief treatment with monoclonal antibodies that block certain functional molecules on the surface of the T cell is able to reprogramme the established immune repertoire of the adult mouse, allowing indefinite acceptance of allografts or effective curing of autoimmune diseases. We are only now just beginning to define many of the regulatory mechanisms that induce and maintain the tolerant state with the aim of being able to safely and reliably apply these technologies to human clinical situations.
Collapse
|
41
|
Boonman ZFHM, van Mierlo GJD, Fransen MF, de Keizer RJW, Jager MJ, Melief CJM, Toes REM. Maintenance of Immune Tolerance Depends on Normal Tissue Homeostasis. THE JOURNAL OF IMMUNOLOGY 2005; 175:4247-54. [PMID: 16177064 DOI: 10.4049/jimmunol.175.7.4247] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ags expressed at immune privileged sites and other peripheral tissues are able to induce T cell tolerance. In this study, we analyzed whether tolerance toward an intraocular tumor expressing a highly immunogenic CTL epitope is maintained, broken, or reverted into immunity in the event the anatomical integrity of the eye is lost. Inoculation of tumor cells into the anterior chamber of the eye of naive B6 mice leads to progressive intraocular tumor growth, an abortive form of CTL activation in the tumor-draining submandibular lymph node, and systemic tolerance as evidenced by the inability of these mice to reject an otherwise benign tumor cell inoculum. Loss of anatomical integrity of the eye as a consequence of phthisis resulted in loss of systemic tolerance and the emergence of effective antitumor immunity against an otherwise lethal tumor challenge. Phthisis was accompanied by dendritic cell maturation and preceded the induction of systemic tumor-specific CTL immunity. Our data show that normal tissue homeostasis and anatomical integrity is required for the maintenance of ocular tolerance and prevention of CTL-mediated immunity. These data also indicate that tissue injury in the absence of viral or microbial infection can act as a switch for the induction of CTL immunity.
Collapse
|
42
|
Pellicer M, Giráldez F, Pumarola F, Barquinero J. [Stem cells for the treatment of hearing loss]. ACTA OTORRINOLARINGOLOGICA ESPANOLA 2005; 56:227-32. [PMID: 15999787 DOI: 10.1016/s0001-6519(05)78606-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
One of the greatest challenges in the treatment of inner ear disorders is to find a cure for the hearing loss caused by the loss of cochlear hair cells or spiral ganglion neurons. The recent discovery of stem cells in the adult inner ear that are capable of differentiating into hair cells, as well as the finding that embryonic stem cells can be converted into hair cells, raise hope for the future development of stem-cell-based treatments.
Collapse
Affiliation(s)
- M Pellicer
- Sección de ORL Pediátrica, Hospital Universitari Vall d'Hebron. Barcelona.
| | | | | | | |
Collapse
|
43
|
Qian J, Lettau M, Podda G, Janssen O. FasL associated factors and their potential role in the regulation of FasL expression. ACTA ACUST UNITED AC 2005. [DOI: 10.1002/sita.200400045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
44
|
Krzyzowska M, Polanczyk M, Bas M, Cymerys J, Schollenberger A, Chiodi F, Niemialtowski M. Mousepox conjunctivitis: the role of Fas/FasL-mediated apoptosis of epithelial cells in virus dissemination. J Gen Virol 2005; 86:2007-2018. [PMID: 15958680 DOI: 10.1099/vir.0.80709-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BALB/c mice infected with the Moscow strain of Ectromelia virus (ECTV-MOS) show a large number of apoptotic cells, and an influx of lymphoid cells in the epithelium and substantia propria of conjunctivae, respectively. The presence of ECTV-MOS antigens in the epithelium of conjunctivae significantly upregulates Fas in the epithelial layer and FasL in the suprabasal layer of conjunctiva. Inhibition of FasL with blocking antibodies in cultures of conjunctival cells isolated from ECTV-MOS-infected BALB/c mice showed that the Fas/FasL pathway is important in apoptosis of ECTV-MOS-infected cells. The results also showed that the presence of cytokines, in particular interferon (IFN)-γ, upregulated expression of Fas. Interleukin (IL) 2, 4, 10 and IFN-γ were produced at the peak of conjunctivitis (at day 15 of infection) with a predominance of IFN-γ and a small, but significant, production of IL4 and IL10 compared with non-infected animals. These results suggest that not only is Fas/FasL expression in conjunctiva involved in elimination of migrating Fas+ cells but also plays an important role in the turnover of conjunctival epithelium and thus may be crucial for ECTV spreading to the surrounding environment.
Collapse
Affiliation(s)
- Malgorzata Krzyzowska
- Immunology Laboratory, Division of Virology, Mycology and Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw Agricultural University, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Magdalena Polanczyk
- Department of Neuroimmunology, School of Medicine, Oregon Health Sciences University, Portland, OR 97201, USA
- Immunology Laboratory, Division of Virology, Mycology and Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw Agricultural University, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Monika Bas
- Immunology Laboratory, Division of Virology, Mycology and Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw Agricultural University, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Joanna Cymerys
- Immunology Laboratory, Division of Virology, Mycology and Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw Agricultural University, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Ada Schollenberger
- Immunology Laboratory, Division of Virology, Mycology and Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw Agricultural University, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Francesca Chiodi
- Microbiology and Tumor Biology Center, Karolinska Institute, Nobels väg 16, S-17177 Stockholm, Sweden
| | - Marek Niemialtowski
- Immunology Laboratory, Division of Virology, Mycology and Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw Agricultural University, Ciszewskiego 8, 02-786 Warsaw, Poland
| |
Collapse
|
45
|
Imarai M, Varela-Nallar L, Figueroa-Gaete C, González P, Valdés D, Velásquez L, Cárdenas H, Maisey K. Fas ligand in the uterus of the non-pregnant mouse induces apoptosis of CD4+ T cells. J Reprod Immunol 2005; 66:13-32. [PMID: 15949559 DOI: 10.1016/j.jri.2005.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2004] [Revised: 01/11/2005] [Accepted: 03/17/2005] [Indexed: 01/25/2023]
Abstract
The role of FasL in the reproductive tissues of the non-pregnant mouse may be the induction of apoptosis of activated T cells for the prevention of unwanted inflammatory responses secondary to infection. This study was undertaken to identify cell and tissue types that typically express FasL in the uterus and oviduct of the mouse and to establish whether FasL on the surface of these cells was able to induce T cell apoptosis. FasL in the mouse uterus and oviduct was demonstrated using three independent methods: RT-PCR, Western blot and immunocytochemistry. The protein was present in the epithelial and mesenchymal cells of the uterus and showed a granular pattern in the apical epithelial portion. Although this suggests the presence of vesicles, surface expression was also detected by flow cytometry of isolated uterine cells. Exogenous administration of estradiol and progesterone had no significant effect on the expression and localization of FasL. The ability of uterine cells to induce FasL-dependent apoptosis of activated CD4+T cells was examined by incubation of phytohemagglutinin-treated T cells with cultured uterine cells. TUNEL and flow cytometric analyses showed that CD4+T cells experienced apoptosis after 5h of co-incubation. Neutralizing antibodies inhibited apoptosis demonstrating that a biologically active FasL is present in the reproductive tissues of the mouse. Results indicate that FasL is a biologically active molecule present in epithelial and mesenchymal cells of the uterus and the oviduct of the non-pregnant mouse that might restrain local immune response by induction of apoptosis of CD4+T lymphocytes.
Collapse
Affiliation(s)
- Mónica Imarai
- Laboratorio de Immunología, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Correo 40, Casilla 33, Santiago, Chile.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Tucker TA, Kundert JA, Bondareva AA, Schmidt EE. Reproductive and neurological Quaking(viable) phenotypes in a severe combined immune deficient mouse background. Immunogenetics 2005; 57:226-31. [PMID: 15900494 PMCID: PMC2604809 DOI: 10.1007/s00251-005-0792-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2004] [Revised: 02/10/2005] [Indexed: 10/25/2022]
Abstract
The quaking(viable) (qkv) mutation, a spontaneous deletion of a multigenic region encompassing roughly 1 Mb at 5.9 cM on the proximal end of mouse chromosome 17, causes severe trembling in all homozygous animals and infertility in all homozygous males. Physiologically, quaking mice exhibit dysmyelination and postmeiotic spermatogenic arrest. Molecular defects in Qkv mice occur in the affected tissues, indicating the primary causes of these pathologies are cell autonomous. However, because both the reproductive and neurological defects are in immune-privileged sites and because some similar pathologies at both sites have been shown to be immune mediated, we tested whether the immune system participates secondarily in manifestation of Qkv phenotypes. The qkv mutation was bred into a severe combined immune-deficient mouse line (SCID; devoid of mature B and T cells) and penetrance of the neurological and the male sterile phenotypes was measured. Results showed that neither defect was ameliorated in the immune-deficient background. We conclude that the Qkv pathologies do not likely involve a B- or T-cell-dependent response against these immune-privileged sites.
Collapse
MESH Headings
- Animals
- Base Sequence
- DNA, Complementary/genetics
- Demyelinating Diseases/genetics
- Demyelinating Diseases/immunology
- Female
- Infertility, Male/genetics
- Infertility, Male/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Quaking/genetics
- Mice, Quaking/immunology
- Mice, Quaking/physiology
- Mice, SCID/genetics
- Mice, SCID/immunology
- Mice, SCID/physiology
- Phenotype
- Pregnancy
- Reproduction/genetics
- Reproduction/immunology
- Spermatogenesis/genetics
- Spermatogenesis/immunology
Collapse
Affiliation(s)
- Tammy A. Tucker
- Veterinary Molecular Biology, Montana State University, Molecular Biosciences Building, 960 Technology Boulevard, Bozeman, MT, 59718, USA
| | - Jean A. Kundert
- Animal Resource Center, Montana State University, Bozeman, MT, 59717, USA
| | - Alla A. Bondareva
- Veterinary Molecular Biology, Montana State University, Molecular Biosciences Building, 960 Technology Boulevard, Bozeman, MT, 59718, USA
- Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N4N1, Canada
| | - Edward E. Schmidt
- Veterinary Molecular Biology, Montana State University, Molecular Biosciences Building, 960 Technology Boulevard, Bozeman, MT, 59718, USA
- e-mail: , Tel.: +1-406-9946375, Fax: +1-406-9944303
| |
Collapse
|
47
|
Linkermann A, Qian J, Lettau M, Kabelitz D, Janssen O. Considering Fas ligand as a target for therapy. Expert Opin Ther Targets 2005; 9:119-34. [PMID: 15757486 DOI: 10.1517/14728222.9.1.119] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
About a decade ago, the death factor Fas ligand (FasL) was identified as the natural trigger of Fas/CD95-dependent apoptosis and as an inducer of Fas-dependent activation-induced cell death. Meanwhile, it is known that this molecule not only contributes to target cell lysis in the immune system but also to the establishment of immune privilege and tumour survival. Because delivering a specific antiproliferative signal to T lymphocytes is of major biomedical interest, the FasL/Fas system has gained much attention over the last few years. However, only recently it became evident that the biology of FasL is more complex than initially anticipated. FasL displays a complex pattern of inducible and constitutive expression associated with a number of different functions as a death factor or a co-stimulatory/accessory molecule in lymphocyte activation. Thus, side effects are likely to occur following systemic administration of, for example, anti-FasL medication, not only because of the constitutive FasL expression on cells within immune privileged tissues and vascular endothelium. In addition, FasL comes in different forms: as a surface molecule, as a protease-shed soluble variant or secreted in vesicles. Because increased levels of soluble FasL (sFasL) have been determined in various immunological and non-immunological diseases, it has been suggested that sFasL might serve as a prognostic or diagnostic marker even though the pathophysiological cause for its enhanced production is hardly known in most cases. This review summarises the current facts and ideas about the clinical and pharmacological potential of FasL and sFasL as targets for therapeutic interventions.
Collapse
Affiliation(s)
- Andreas Linkermann
- Medical Center Schleswig-Holstein Campus Kiel, Institute of Immunology, Michaelisstr. 5, D-24105 Kiel, Germany
| | | | | | | | | |
Collapse
|
48
|
Sonntag KC, Simantov R, Isacson O. Stem cells may reshape the prospect of Parkinson's disease therapy. ACTA ACUST UNITED AC 2005; 134:34-51. [PMID: 15790528 DOI: 10.1016/j.molbrainres.2004.09.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2004] [Indexed: 12/21/2022]
Abstract
The concept of cell replacement to compensate for cell loss and restore functionality has entered several disease entities including neurodegenerative disorders. Recent clinical studies have shown that transplantation of fetal dopaminergic (DA) cells into the brain of Parkinson's disease (PD) patients can reduce disease-associated motor deficits. However, the use of fetal tissue is associated with practical and ethical problems including low efficiency, variability in the clinical outcome and controversy regarding the use of fetuses as donor. An alternative cell resource could be embryonic stem (ES) cells, which can be cultivated in unlimited amounts and which have the potential to differentiate into mature DA cells. Several differentiation protocols have been developed, and some progress has been made in understanding the mechanisms underlying DA specification in ES cell development, but the "holy grail" in this paradigm, which is the production of sufficient amounts of the "right" therapeutic DA cell, has not yet been accomplished. To achieve this goal, several criteria on the transplanted DA cells need to be fulfilled, mainly addressing cell survival, accurate integration in the brain circuitry, normal function, no tumor formation, and no immunogenicity. Here, we summarize the current state of ES cell-derived DA neurogenesis and discuss the aspects involved in generating an optimal cell source for cell replacement in PD.
Collapse
Affiliation(s)
- Kai-Christian Sonntag
- Udall Parkinson's Disease Research Center of Excellence, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478, USA.
| | | | | |
Collapse
|
49
|
Bohana-Kashtan O, Civin CI. Profiling Tumor Counterattack: Do Fas Ligand–Containing Microvesicles Reduce Anticancer Immunity? Clin Cancer Res 2005. [DOI: 10.1158/1078-0432.968.11.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Osnat Bohana-Kashtan
- 1Division of Immunology and Hematopoiesis, Sidney Kimmel Comprehensive Cancer Center, Departments of Oncology and
| | - Curt I. Civin
- 1Division of Immunology and Hematopoiesis, Sidney Kimmel Comprehensive Cancer Center, Departments of Oncology and
- 2Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
50
|
Abstract
Human embryonic stem cells have excellent potential for being the ultimate source of transplantable cells for many different tissues. To enable their clinical use, differentiation protocols should be developed and safety standards must be met. The cells should improve symptoms without generating side effects and their immune rejection must be overcome. Profiling of the immune antigens expressed on the cells has revealed that upon differentiation the cells express molecules of the major histocompatibility complex. Here, we propose ways of overcoming the rejection of human embryonic stem cells after transplantation.
Collapse
Affiliation(s)
- Micha Drukker
- Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, 91904 Jerusalem, Israel.
| | | |
Collapse
|