1
|
Kihara Y, Tanaka Y, Ikeda M, Homma J, Takagi R, Ishigaki K, Yamanouchi K, Honda H, Nagata S, Yamato M. In utero transplantation of myoblasts and adipose-derived mesenchymal stem cells to murine models of Duchenne muscular dystrophy does not lead to engraftment and frequently results in fetal death. Regen Ther 2022; 21:486-493. [PMID: 36313392 PMCID: PMC9596598 DOI: 10.1016/j.reth.2022.10.003] [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: 08/23/2022] [Revised: 09/19/2022] [Accepted: 10/09/2022] [Indexed: 11/07/2022] Open
Abstract
Introduction Duchenne muscular dystrophy (DMD) is a progressive disease that leads to damage of muscle and myocardium due to genetic abnormalities in the dystrophin gene. In utero cell transplantation that might facilitate allogenic transplantation is worth considering to treat this disease. Methods We performed allogeneic in utero transplantation of GFP-positive myoblasts and adipose-derived mesenchymal stem cells into murine DMD model animals. The transplantation route in this study was fetal intraperitoneal transplantation and transplacental transplantation. Transplanted animals were examined at 4-weeks old by immunofluorescence staining and RT-qPCR. Results No GFP-positive cells were found by immunofluorescence staining of skeletal muscle and no GFP mRNA was detected by RT-qPCR in any animal, transplantation method and cell type. Compared with previous reports, myoblast transplantation exhibited an equivalent mortality rate, but adipose-derived stem cell (ASC) transplantation produced a higher mortality rate. Conclusions In utero transplantation of myoblasts or ASCs to murine models of DMD does not lead to engraftment and, in ASC transplantation primarily, frequently results in fetal death.
Collapse
Affiliation(s)
- Yuki Kihara
- Department of Pediatrics, Tokyo Women’s Medical University, School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan,Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Yukie Tanaka
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | - Masanari Ikeda
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | - Jun Homma
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Ryo Takagi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Keiko Ishigaki
- Department of Pediatrics, Tokyo Women’s Medical University, School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Keitaro Yamanouchi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | - Hiroaki Honda
- Human Disease Models, Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, Japan
| | - Satoru Nagata
- Department of Pediatrics, Tokyo Women’s Medical University, School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan,Corresponding author. Fax: +81 3-3359-6046.
| |
Collapse
|
2
|
Lederer CW, Koniali L, Buerki-Thurnherr T, Papasavva PL, La Grutta S, Licari A, Staud F, Bonifazi D, Kleanthous M. Catching Them Early: Framework Parameters and Progress for Prenatal and Childhood Application of Advanced Therapies. Pharmaceutics 2022; 14:pharmaceutics14040793. [PMID: 35456627 PMCID: PMC9031205 DOI: 10.3390/pharmaceutics14040793] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 01/19/2023] Open
Abstract
Advanced therapy medicinal products (ATMPs) are medicines for human use based on genes, cells or tissue engineering. After clear successes in adults, the nascent technology now sees increasing pediatric application. For many still untreatable disorders with pre- or perinatal onset, timely intervention is simply indispensable; thus, prenatal and pediatric applications of ATMPs hold great promise for curative treatments. Moreover, for most inherited disorders, early ATMP application may substantially improve efficiency, economy and accessibility compared with application in adults. Vindicating this notion, initial data for cell-based ATMPs show better cell yields, success rates and corrections of disease parameters for younger patients, in addition to reduced overall cell and vector requirements, illustrating that early application may resolve key obstacles to the widespread application of ATMPs for inherited disorders. Here, we provide a selective review of the latest ATMP developments for prenatal, perinatal and pediatric use, with special emphasis on its comparison with ATMPs for adults. Taken together, we provide a perspective on the enormous potential and key framework parameters of clinical prenatal and pediatric ATMP application.
Collapse
Affiliation(s)
- Carsten W. Lederer
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.K.); (P.L.P.); (M.K.)
- Correspondence: ; Tel.: +357-22-392764
| | - Lola Koniali
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.K.); (P.L.P.); (M.K.)
| | - Tina Buerki-Thurnherr
- Empa, Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland;
| | - Panayiota L. Papasavva
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.K.); (P.L.P.); (M.K.)
| | - Stefania La Grutta
- Institute of Translational Pharmacology, IFT National Research Council, 90146 Palermo, Italy;
| | - Amelia Licari
- Pediatric Clinic, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy;
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, 50005 Hradec Králové, Czech Republic;
| | - Donato Bonifazi
- Consorzio per Valutazioni Biologiche e Farmacologiche (CVBF) and European Paediatric Translational Research Infrastructure (EPTRI), 70122 Bari, Italy;
| | - Marina Kleanthous
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.K.); (P.L.P.); (M.K.)
| |
Collapse
|
3
|
Mohamedaly S, Alkhani A, Nijagal A. The relative abundance of monocyte subsets determines susceptibility to perinatal hepatic inflammation. JOURNAL OF CLINICAL & CELLULAR IMMUNOLOGY 2020; 11:602. [PMID: 36304699 PMCID: PMC9603689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The devastating consequences of perinatal liver inflammation contribute to a pressing need to develop therapeutics for the diseases that underly this condition. Biliary atresia (BA) is a perinatal inflammatory disease of the liver that results in obliterative cholangiopathy and rapidly progresses to liver failure, requiring transplantation. The ability to develop targeted therapies requires an understanding of the immune mechanisms that mitigate perinatal liver inflammation. This article reviews our recent findings demonstrating that in a murine model of perinatal hepatic inflammation, Ly6cLo non-classical monocytes express a pro-reparative transcriptomic profile and that the relative abundance of Ly6cLo monocytes promotes resolution of perinatal liver inflammation, rendering neonatal pups resistant to disease. We also examine the lineage relationship between monocyte subsets, reviewing data that suggests classical monocytes are a precursor for non-classical monocytes, and the alternative possibility that separate progenitors exist for each subset. Although a precursor-product relationship between classical and non-classical monocytes might exist in certain environments, we argue that they may also arise from separate progenitors, which is evident by sustained Ly6cLo non-classical monocyte expansion when Ly6cHi monocytes are absent. An improved understanding of monocyte subsets and their developmental trajectories during perinatal hepatic inflammation will provide insight into how therapies directed at controlling monocyte function may help alleviate the devastating consequences of diseases like BA.
Collapse
Affiliation(s)
| | | | - Amar Nijagal
- ‡ Corresponding Author: Amar Nijagal, MD, Assistant Professor of Surgery, Division of Pediatric Surgery, 513 Parnassus Avenue, HSW 1652, Campus Box 0570, University of CA, San Francisco, San Francisco, CA 94143-0570, Office: 415-476-4086; Fax: 415-476-2314,
| |
Collapse
|
4
|
Ly6c Lo non-classical monocytes promote resolution of rhesus rotavirus-mediated perinatal hepatic inflammation. Sci Rep 2020; 10:7165. [PMID: 32346042 PMCID: PMC7188847 DOI: 10.1038/s41598-020-64158-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/11/2020] [Indexed: 12/02/2022] Open
Abstract
Perinatal hepatic inflammation can have devastating consequences. Monocytes play an important role in the initiation and resolution of inflammation, and their diverse functions can be attributed to specific cellular subsets: pro-inflammatory or classical monocytes (Ly6cHi) and pro-reparative or non-classical monocytes (Ly6cLo). We hypothesized that inherent differences in Ly6cHi classical monocytes and Ly6cLo non-classical monocytes determine susceptibility to perinatal hepatic inflammation in late gestation fetuses and neonates. We found an anti-inflammatory transcriptional profile expressed by Ly6cLo non-classical monocytes, and a physiologic abundance of these cells in the late gestation fetal liver. Unlike neonatal pups, late gestation fetuses proved to be resistant to rhesus rotavirus (RRV) mediated liver inflammation. Furthermore, neonatal pups were rendered resistant to RRV-mediated liver injury when Ly6cLo non-classical monocytes were expanded. Pharmacologic inhibition of Ly6cLo non-classical monocytes in this setting restored susceptibility to RRV-mediated disease. These data demonstrate that Ly6cLo monocytes promote resolution of perinatal liver inflammation in the late gestation fetus, where there is a physiologic expansion of non-classical monocytes, and in the neonatal liver upon experimental expansion of these cells. Therapeutic strategies directed towards enhancing Ly6cLo non-classical monocyte function may mitigate the detrimental effects of perinatal liver inflammation.
Collapse
|
5
|
Frascoli M, Coniglio L, Witt R, Jeanty C, Fleck-Derderian S, Myers DE, Lee TH, Keating S, Busch MP, Norris PJ, Tang Q, Cruz G, Barcellos LF, Gomez-Lopez N, Romero R, MacKenzie TC. Alloreactive fetal T cells promote uterine contractility in preterm labor via IFN-γ and TNF-α. Sci Transl Med 2019; 10:10/438/eaan2263. [PMID: 29695455 DOI: 10.1126/scitranslmed.aan2263] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 12/13/2017] [Accepted: 04/05/2018] [Indexed: 12/15/2022]
Abstract
Healthy pregnancy is the most successful form of graft tolerance, whereas preterm labor (PTL) may represent a breakdown in maternal-fetal tolerance. Although maternal immune responses have been implicated in pregnancy complications, fetal immune responses against maternal antigens are often not considered. To examine the fetal immune system in the relevant clinical setting, we analyzed maternal and cord blood in patients with PTL and healthy term controls. We report here that the cord blood of preterm infants has higher amounts of inflammatory cytokines and a greater activation of dendritic cells. Moreover, preterm cord blood is characterized by the presence of a population of central memory cells with a type 1 T helper phenotype, which is absent in term infants, and an increase in maternal microchimerism. T cells from preterm infants mount a robust proliferative, proinflammatory response to maternal antigens compared to term infants yet fail to respond to third-party antigens. Furthermore, we show that T cells from preterm infants stimulate uterine myometrial contractility through interferon-γ and tumor necrosis factor-α. In parallel, we found that adoptive transfer of activated T cells directly into mouse fetuses resulted in pregnancy loss. Our findings indicate that fetal inflammation and rejection of maternal antigens can contribute to the signaling cascade that promotes uterine contractility and that aberrant fetal immune responses should be considered in the pathogenesis of PTL.
Collapse
Affiliation(s)
- Michela Frascoli
- Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Lacy Coniglio
- Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Russell Witt
- Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Cerine Jeanty
- Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Department of Surgery, University of California, San Francisco, CA 94143, USA
| | | | - Dana E Myers
- Obstetrics and Gynecology, University of California, San Francisco, CA 94143, USA
| | - Tzong-Hae Lee
- Blood Systems Research Institute, San Francisco, CA 94118, USA
| | - Sheila Keating
- Blood Systems Research Institute, San Francisco, CA 94118, USA
| | - Michael P Busch
- Blood Systems Research Institute, San Francisco, CA 94118, USA
| | - Philip J Norris
- Blood Systems Research Institute, San Francisco, CA 94118, USA
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Giovanna Cruz
- Division of Epidemiology, Genetic Epidemiology and Genomics Laboratory, School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Lisa F Barcellos
- Division of Epidemiology, Genetic Epidemiology and Genomics Laboratory, School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)/National Institutes of Health (NIH)/U.S. Department of Health and Human Services (DHHS), Bethesda, MD 20892, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA.,Department of Microbiology, Immunology, and Biochemistry, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)/National Institutes of Health (NIH)/U.S. Department of Health and Human Services (DHHS), Bethesda, MD 20892, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Tippi C MacKenzie
- Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, CA 94143, USA. .,Department of Surgery, University of California, San Francisco, CA 94143, USA.,Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
6
|
Kumar P, Gao K, Wang C, Pivetti C, Lankford L, Farmer D, Wang A. In Utero Transplantation of Placenta-Derived Mesenchymal Stromal Cells for Potential Fetal Treatment of Hemophilia A. Cell Transplant 2019; 27:130-139. [PMID: 29562772 PMCID: PMC6434487 DOI: 10.1177/0963689717728937] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Hemophilia A (HA) is an X-linked recessive disorder caused by mutations in the factor VIII (FVIII) gene leading to deficient blood coagulation. The current standard of care is frequent infusions of plasma-derived FVIII or recombinant B-domain-deleted FVIII (BDD-FVIII). While this treatment is effective, many patients eventually develop FVIII inhibitors that limit the effectiveness of the infused FVIII. As a monogenic disorder, HA is an ideal target for gene or cell-based therapy. Several studies have investigated allogeneic stem cell therapy targeting in utero or postnatal treatment of HA but have not been successful in completely correcting HA. Autologous in utero transplantation of mesenchymal stem cells is promising for treatment of HA due to the naive immune status of the fetal environment as well as its potential to prevent transplant rejection and long-term FVIII inhibitor formation. HA can be diagnosed by chorionic villus sampling performed during the first trimester (10 to 13 wk) of gestation. In this study, we used an established protocol and isolated placenta-derived mesenchymal stromal cells (PMSCs) from first trimester chorionic villus tissue and transduced them with lentiviral vector encoding the BDD-FVIII gene. We show that gene-modified PMSCs maintain their immunophenotype and multipotency, express, and secrete high levels of active FVIII. PMSCs were then transplanted at embryonic day 14.5 (E14.5) into wild-type fetuses from time-mated pregnant mice. Four days after birth, pups were checked for engraftment, and varying levels of expression of human green fluorescent protein were found in the organs tested. This study shows feasibility of the approach to obtain PMSCs from first trimester chorionic villus tissue, genetically modify them with the FVIII gene, and transplant them in utero for cell-mediated gene therapy of HA. Future studies will involve evaluation of long-term engraftment, phenotypic correction in HA mice, and prevention of FVIII inhibitor development by this approach.
Collapse
Affiliation(s)
- Priyadarsini Kumar
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| | - Kewa Gao
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA.,2 Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Chuwang Wang
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA.,2 Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Christopher Pivetti
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| | - Lee Lankford
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| | - Diana Farmer
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| | - Aijun Wang
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
7
|
Mattar CNZ, Tan YW, Johana N, Biswas A, Tan LG, Choolani M, Bakkour S, Johnson M, Chan JKY, Flake AW. Fetoscopic versus Ultrasound-Guided Intravascular Delivery of Maternal Bone Marrow Cells in Fetal Macaques: A Technical Model for Intrauterine Haemopoietic Cell Transplantation. Fetal Diagn Ther 2019; 46:175-186. [PMID: 30661073 DOI: 10.1159/000493791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/14/2018] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Significant limitations with existing treatments for major haemoglobinopathies motivate the development of effective intrauterine therapy. We assessed the feasibility of fetoscopic and ultrasound-guided intrauterine haemopoietic cell transplantation (IUHCT) in macaque fetuses in early gestation when haemopoietic and immunological ontogeny is anticipated to enable long-term donor cell engraftment. MATERIAL AND METHODS Fluorescent-labelled bone marrow-derived mononuclear cells from 10 pregnant Macaca fascicularis were injected into their fetuses at E71-114 (18.9-170.0E+6 cells/fetus) by fetoscopic intravenous (n = 7) or ultrasound (US)-guided intracardiac injections, with sacrifice at 24 h to examine donor-cell distribution. RESULTS Operating times ranged from 35 to 118 min. Chorionic membrane tenting and intrachorionic haemorrhage were observed only with fetoscopy (n = 2). Labelled cells were stereoscopically visualised in lung, spleen, liver, and placenta. Donor-cell chimerism was highest in liver, spleen, and heart by flow cytometry, placenta by unique polymorphism qPCR, and was undetected in blood. Chimerism was 2-3 log-fold lower in individual organs by qPCR than by flow cytometry. DISCUSSION Both fetoscopic and US-guided IUHCT were technically feasible, but fetoscopy caused more intraoperative complications in our pilot series. The discrepancy in chimerism detection predicts the challenges in long-term surveillance of donor-cell chimerism. Further studies of long-term outcomes in the non-human primate are valuable for the development of clinical protocols for IUHCT.
Collapse
Affiliation(s)
- Citra N Z Mattar
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yi-Wan Tan
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Nuryanti Johana
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Arijit Biswas
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lay-Geok Tan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mahesh Choolani
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sonia Bakkour
- Blood Systems Research Institute, San Francisco, California, USA
| | - Mark Johnson
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jerry K Y Chan
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore, .,Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore, Singapore,
| | - Alan W Flake
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Ahn NJ, Stratigis JD, Coons BE, Flake AW, Nah-Cederquist HD, Peranteau WH. Intravenous and Intra-amniotic In Utero Transplantation in the Murine Model. J Vis Exp 2018. [PMID: 30371676 DOI: 10.3791/58047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
In utero transplantation (IUT) is a unique and versatile mode of therapy that can be used to introduce stem cells, viral vectors, or any other substances early in the gestation. The rationale behind IUT for therapeutic purposes is based on the small size of the fetus, the fetal immunologic immaturity, the accessibility and proliferative nature of the fetal stem or progenitor cells, and the potential to treat a disease or the onset of symptoms prior to birth. Taking advantage of these normal developmental properties of the fetus, the delivery of hematopoietic stem cells (HSC) via an IUT has the potential to treat congenital hematologic disorders such as sickle cell disease, without the required myeloablative or immunosuppressive conditioning required for postnatal HSC transplants. Similarly, the accessibility of progenitor cells in multiple organs during development potentially allows for a more efficient targeting of stem/progenitor cells following an IUT of viral vectors for gene therapy or genome editing. Additionally, IUT can be used to study normal developmental processes including, but not limited to, the development of immunologic tolerance. The murine model provides a valuable and affordable means to understanding the potential and limitations of IUT prior to pre-clinical large animal studies and an eventual clinical application. Here, we describe a protocol for performing an IUT in the murine fetus through intravenous and intra-amniotic routes. This protocol has been used successfully to elucidate the necessary conditions and mechanisms behind in utero hematopoietic stem cell transplantation, tolerance induction, and in utero gene therapy.
Collapse
Affiliation(s)
- Nicholas J Ahn
- Division of General, Thoracic, and Fetal Surgery, Center for Fetal Research, Children's Hospital of Philadelphia
| | - John D Stratigis
- Division of General, Thoracic, and Fetal Surgery, Center for Fetal Research, Children's Hospital of Philadelphia
| | - Barbara E Coons
- Division of General, Thoracic, and Fetal Surgery, Center for Fetal Research, Children's Hospital of Philadelphia
| | - Alan W Flake
- Division of General, Thoracic, and Fetal Surgery, Center for Fetal Research, Children's Hospital of Philadelphia
| | - Hyun-Duck Nah-Cederquist
- Division of Plastic Reconstructive Surgery, Center for Fetal Research, Children's Hospital of Philadelphia
| | - William H Peranteau
- Division of General, Thoracic, and Fetal Surgery, Center for Fetal Research, Children's Hospital of Philadelphia;
| |
Collapse
|
9
|
Systemic multilineage engraftment in mice after in utero transplantation with human hematopoietic stem cells. Blood Adv 2018; 2:69-74. [PMID: 29344586 DOI: 10.1182/bloodadvances.2017011585] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/10/2017] [Indexed: 01/22/2023] Open
Abstract
IUHCT of human cord blood-derived CD34+ cells into fetal NSG mice results in systemic multilineage engraftment with human cells.Preconditioning with in utero injection of an anti-c-Kit receptor antibody (ACK2) results in an improved rate of engraftment.
Collapse
|
10
|
Beaudin AE, Boyer SW, Perez-Cunningham J, Hernandez GE, Derderian SC, Jujjavarapu C, Aaserude E, MacKenzie T, Forsberg EC. A Transient Developmental Hematopoietic Stem Cell Gives Rise to Innate-like B and T Cells. Cell Stem Cell 2016; 19:768-783. [PMID: 27666010 DOI: 10.1016/j.stem.2016.08.013] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 06/01/2016] [Accepted: 08/15/2016] [Indexed: 12/24/2022]
Abstract
The generation of distinct hematopoietic cell types, including tissue-resident immune cells, distinguishes fetal from adult hematopoiesis. However, the mechanisms underlying differential cell production to generate a layered immune system during hematopoietic development are unclear. Using an irreversible lineage-tracing model, we identify a definitive hematopoietic stem cell (HSC) that supports long-term multilineage reconstitution upon transplantation into adult recipients but does not persist into adulthood in situ. These HSCs are fully multipotent, yet they display both higher lymphoid cell production and greater capacity to generate innate-like B and T lymphocytes as compared to coexisting fetal HSCs and adult HSCs. Thus, these developmentally restricted HSCs (drHSCs) define the origin and generation of early lymphoid cells that play essential roles in establishing self-recognition and tolerance, with important implications for understanding autoimmune disease, allergy, and rejection of transplanted organs.
Collapse
Affiliation(s)
- Anna E Beaudin
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Scott W Boyer
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jessica Perez-Cunningham
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Gloria E Hernandez
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - S Christopher Derderian
- Eli and Edythe Broad Center of Regeneration Medicine and Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Chethan Jujjavarapu
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Eric Aaserude
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Tippi MacKenzie
- Eli and Edythe Broad Center of Regeneration Medicine and Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
11
|
Frascoli M, Jeanty C, Fleck S, Moradi PW, Keating S, Mattis AN, Tang Q, MacKenzie TC. Heightened Immune Activation in Fetuses with Gastroschisis May Be Blocked by Targeting IL-5. THE JOURNAL OF IMMUNOLOGY 2016; 196:4957-66. [PMID: 27183609 DOI: 10.4049/jimmunol.1502587] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/18/2016] [Indexed: 12/20/2022]
Abstract
The development of the fetal immune system during pregnancy is a well-orchestrated process with important consequences for fetal and neonatal health, but prenatal factors that affect immune activation are poorly understood. We hypothesized that chronic fetal inflammation may lead to alterations in development of the fetal immune system. To test this hypothesis, we examined neonates with gastroschisis, a congenital abdominal wall defect that leads to exposure of the fetal intestines to amniotic fluid, with resultant intestinal inflammation. We determined that patients with gastroschisis show high systemic levels of inflammatory cytokines and chemokines such as eotaxin, as well as earlier activation of CD4(+) and CD8(+) effector and memory T cells in the cord blood compared with controls. Additionally, increased numbers of T cells and eosinophils infiltrate the serosa and mucosa of the inflamed intestines. Using a mouse model of gastroschisis, we observed higher numbers of eosinophils and both type 2 and type 3 innate lymphoid cells (ILC2 and ILC3), specifically in the portion of organs exposed to the amniotic fluid. Given the role of IL-5 produced by ILC2 in regulating eosinophil development and survival, we determined that maternal or fetal administration of the anti-IL-5 neutralizing Ab, or a depleting Ab against ILCs, can both effectively reduce intestinal eosinophilia. Thus, a congenital anomaly causing chronic inflammation can alter the composition of circulating and tissue-resident fetal immune cells. Given the high rate of prenatal and neonatal complications in these patients, such changes have clinical significance and might become targets for fetal therapy.
Collapse
Affiliation(s)
- Michela Frascoli
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143; Department of Surgery, University of California San Francisco, San Francisco, CA 94143
| | - Cerine Jeanty
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143; Department of Surgery, University of California San Francisco, San Francisco, CA 94143
| | - Shannon Fleck
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143
| | - Patriss W Moradi
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143; Department of Surgery, University of California San Francisco, San Francisco, CA 94143
| | - Sheila Keating
- Blood Systems Research Institute, San Francisco, CA 94118; and
| | - Aras N Mattis
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143; Department of Pathology, University of California San Francisco, San Francisco, CA 94143
| | - Qizhi Tang
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143
| | - Tippi C MacKenzie
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143; Department of Surgery, University of California San Francisco, San Francisco, CA 94143; Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143;
| |
Collapse
|
12
|
Ravikumar M, Sunil S, Black J, Barkauskas DS, Haung AY, Miller RH, Selkirk SM, Capadona JR. The roles of blood-derived macrophages and resident microglia in the neuroinflammatory response to implanted intracortical microelectrodes. Biomaterials 2014; 35:8049-64. [PMID: 24973296 DOI: 10.1016/j.biomaterials.2014.05.084] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 05/28/2014] [Indexed: 12/23/2022]
Abstract
Resident microglia and blood-borne macrophages have both been implicated to play a dominant role in mediating the neuroinflammatory response affecting implanted intracortical microelectrodes. However, the distinction between each cell type has not been demonstrated due to a lack of discriminating cellular markers. Understanding the subtle differences of each cell population in mediating neuroinflammation can aid in determining the appropriate therapeutic approaches to improve microelectrode performance. Therefore, the goal of this study is to characterize the role of infiltrating blood-derived cells, specifically macrophages, in mediating neuroinflammation following intracortical microelectrode implantation. Interestingly, we found no correlation between microglia and neuron populations at the microelectrode-tissue interface. On the other hand, blood-borne macrophages consistently dominated the infiltrating cell population following microelectrode implantation. Most importantly, we found a correlation between increased populations of blood-derived cells (including the total macrophage population) and neuron loss at the microelectrode-tissue interface. Specifically, the total macrophage population was greatest at two and sixteen weeks post implantation, at the same time points when we observed the lowest densities of neuronal survival in closest proximity to the implant. Together, our results suggest a dominant role of infiltrating macrophages, and not resident microglia, in mediating neurodegeneration following microelectrode implantation.
Collapse
Affiliation(s)
- Madhumitha Ravikumar
- Department of Biomedical Engineering, Case Western Reserve University, School of Engineering, 2071 Martin Luther King Jr. Drive, Wickenden Bldg, Cleveland OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Rehabilitation Research and Development, Spinal Cord Injury Division, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland OH 44106, USA
| | - Smrithi Sunil
- Department of Biomedical Engineering, Case Western Reserve University, School of Engineering, 2071 Martin Luther King Jr. Drive, Wickenden Bldg, Cleveland OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Rehabilitation Research and Development, Spinal Cord Injury Division, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland OH 44106, USA
| | - James Black
- Department of Biomedical Engineering, Case Western Reserve University, School of Engineering, 2071 Martin Luther King Jr. Drive, Wickenden Bldg, Cleveland OH 44106, USA
| | - Deborah S Barkauskas
- Department of Biomedical Engineering, Case Western Reserve University, School of Engineering, 2071 Martin Luther King Jr. Drive, Wickenden Bldg, Cleveland OH 44106, USA; Department of Pediatrics, Case Western Reserve University School of Medicine, Wolstein Research Building 6528, 2103 Cornell Rd, Cleveland, OH 44106, USA
| | - Alex Y Haung
- Department of Pediatrics, Case Western Reserve University School of Medicine, Wolstein Research Building 6528, 2103 Cornell Rd, Cleveland, OH 44106, USA
| | - Robert H Miller
- Department of Neurosciences, Case Western Reserve University, School of Medicine, 10900 Euclid Ave, Sears Tower Bldg, Cleveland OH 44106, USA
| | - Stephen M Selkirk
- Department of Neurology, Case Western Reserve University, School of Medicine, 11100 Euclid Avenue, Sears Tower Bldg, Cleveland OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Rehabilitation Research and Development, Spinal Cord Injury Division, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland OH 44106, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, School of Engineering, 2071 Martin Luther King Jr. Drive, Wickenden Bldg, Cleveland OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Rehabilitation Research and Development, Spinal Cord Injury Division, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland OH 44106, USA.
| |
Collapse
|
13
|
In utero depletion of fetal hematopoietic stem cells improves engraftment after neonatal transplantation in mice. Blood 2014; 124:973-80. [PMID: 24879814 DOI: 10.1182/blood-2014-02-550327] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Although in utero hematopoietic cell transplantation is a promising strategy to treat congenital hematopoietic disorders, levels of engraftment have not been therapeutic for diseases in which donor cells have no survival advantage. We used an antibody against the murine c-Kit receptor (ACK2) to deplete fetal host hematopoietic stem cells (HSCs) and increase space within the hematopoietic niche for donor cell engraftment. Fetal mice were injected with ACK2 on embryonic days 13.5 to 14.5 and surviving pups were transplanted with congenic hematopoietic cells on day of life 1. Low-dose ACK2 treatment effectively depleted HSCs within the bone marrow with minimal toxicity and the antibody was cleared from the serum before the neonatal transplantation. Chimerism levels were significantly higher in treated pups than in controls; both myeloid and lymphoid cell chimerism increased because of higher engraftment of HSCs in the bone marrow. To test the strategy of repeated HSC depletion and transplantation, some mice were treated with ACK2 postnatally, but the increase in engraftment was lower than that seen with prenatal treatment. We demonstrate a successful fetal conditioning strategy associated with minimal toxicity. Such strategies could be used to achieve clinically relevant levels of engraftment to treat congenital stem cell disorders.
Collapse
|
14
|
Evans TA, Barkauskas DS, Myers JT, Hare EG, You JQ, Ransohoff RM, Huang AY, Silver J. High-resolution intravital imaging reveals that blood-derived macrophages but not resident microglia facilitate secondary axonal dieback in traumatic spinal cord injury. Exp Neurol 2014; 254:109-20. [PMID: 24468477 PMCID: PMC3954731 DOI: 10.1016/j.expneurol.2014.01.013] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/12/2013] [Accepted: 01/11/2014] [Indexed: 10/25/2022]
Abstract
After traumatic spinal cord injury, functional deficits increase as axons die back from the center of the lesion and the glial scar forms. Axonal dieback occurs in two phases: an initial axon intrinsic stage that occurs over the first several hours and a secondary phase which takes place over the first few weeks after injury. Here, we examine the secondary phase, which is marked by infiltration of macrophages. Using powerful time-lapse multi-photon imaging, we captured images of interactions between Cx3cr1(+/GFP) macrophages and microglia and Thy-1(YFP) axons in a mouse dorsal column crush spinal cord injury model. Over the first few weeks after injury, axonal retraction bulbs within the lesion are static except when axonal fragments are lost by a blebbing mechanism in response to physical contact followed by phagocytosis by mobile Cx3Cr1(+/GFP) cells. Utilizing a radiation chimera model to distinguish marrow-derived cells from radio-resistant CNS-resident microglia, we determined that the vast majority of accumulated cells in the lesion are derived from the blood and only these are associated with axonal damage. Interestingly, CNS-resident Cx3Cr1(+/GFP) microglia did not increasingly accumulate nor participate in neuronal destruction in the lesion during this time period. Additionally, we found that the blood-derived cells consisted mainly of singly labeled Ccr2(+/RFP) macrophages, singly labeled Cx3Cr1(+/GFP) macrophages and a small population of double-labeled cells. Since all axon destructive events were seen in contact with a Cx3Cr1(+/GFP) cell, we infer that the CCR2 single positive subset is likely not robustly involved in axonal dieback. Finally, in our model, deletion of CCR2, a chemokine receptor, did not alter the position of axons after dieback. Understanding the in vivo cellular interactions involved in secondary axonal injury may lead to clinical treatment candidates involving modulation of destructive infiltrating blood monocytes.
Collapse
Affiliation(s)
- Teresa A Evans
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Deborah S Barkauskas
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Jay T Myers
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Elisabeth G Hare
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Jing Qiang You
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Richard M Ransohoff
- Department of Neurosciences, Neuroinflammation Research Center, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | - Alex Y Huang
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
15
|
Wegorzewska M, Nijagal A, Wong CM, Le T, Lescano N, Tang Q, MacKenzie TC. Fetal intervention increases maternal T cell awareness of the foreign conceptus and can lead to immune-mediated fetal demise. THE JOURNAL OF IMMUNOLOGY 2014; 192:1938-45. [PMID: 24415782 DOI: 10.4049/jimmunol.1302403] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fetal interventions to diagnose and treat congenital anomalies are growing in popularity but often lead to preterm labor. The possible contribution of the maternal adaptive immune system to postsurgical pregnancy complications has not been explored. We recently showed that fetal intervention in mice increases maternal T cell trafficking into the fetus and hypothesized that this process also may lead to increased maternal T cell recognition of the foreign conceptus and subsequent breakdown in maternal-fetal tolerance. In this study, we show that fetal intervention in mice results in accumulation of maternal T cells in the uterus and that these activated cells can produce effector cytokines. In adoptive transfer experiments, maternal T cells specific for a fetal alloantigen proliferate after fetal intervention, escape apoptosis, and become enriched compared with endogenous T cells in the uterus and uterine-draining lymph nodes. Finally, we demonstrate that such activation and accumulation can have a functional consequence: in utero transplantation of hematopoietic cells carrying the fetal alloantigen leads to enhanced demise of semiallogeneic fetuses within a litter. We further show that maternal T cells are necessary for this phenomenon. These results suggest that fetal intervention enhances maternal T cell recognition of the fetus and that T cell activation may be a culprit in postsurgical pregnancy complications. Our results have clinical implications for understanding and preventing complications associated with fetal surgery such as preterm labor.
Collapse
Affiliation(s)
- Marta Wegorzewska
- Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143
| | | | | | | | | | | | | |
Collapse
|
16
|
Haddad-Tóvolli R, Szabó NE, Zhou X, Alvarez-Bolado G. Genetic manipulation of the mouse developing hypothalamus through in utero electroporation. J Vis Exp 2013. [PMID: 23912701 DOI: 10.3791/50412] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Genetic modification of specific regions of the developing mammalian brain is a very powerful experimental approach. However, generating novel mouse mutants is often frustratingly slow. It has been shown that access to the mouse brain developing in utero with reasonable post-operatory survival is possible. Still, results with this procedure have been reported almost exclusively for the most superficial and easily accessible part of the developing brain, i.e. the cortex. The thalamus, a narrower and more medial region, has proven more difficult to target. Transfection into deeper nuclei, especially those of the hypothalamus, is perhaps the most challenging and therefore very few results have been reported. Here we demonstrate a procedure to target the entire hypothalamic neuroepithelium or part of it (hypothalamic regions) for transfection through electroporation. The keys to our approach are longer narcosis times, injection in the third ventricle, and appropriate kind and positioning of the electrodes. Additionally, we show results of targeting and subsequent histological analysis of the most recessed hypothalamic nucleus, the mammillary body.
Collapse
|
17
|
Direct and indirect antigen presentation lead to deletion of donor-specific T cells after in utero hematopoietic cell transplantation in mice. Blood 2013; 121:4595-602. [PMID: 23610372 DOI: 10.1182/blood-2012-10-463174] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
In utero hematopoietic cell transplantation (IUHCTx) is a promising method to induce donor-specific tolerance but the mechanisms of antigen presentation that educate host T cells and the relative importance of deletion vs regulation in this setting are unknown. We studied the roles of direct and indirect antigen presentation (mediated by donor- and host-derived antigen-presenting cells [APCs], respectively) in a mouse model of IUHCTx. We found that IUHCTx leads to precocious maturation of neonatal host dendritic cells (DCs) and that there is early differentiation of donor-derived DCs, even after transplantation of a stem cell source without mature APCs. We next performed allogeneic IUHCTx into donor-specific T-cell receptor transgenic mice and confirmed that both direct and indirect antigen presentation lead to clonal deletion of effector T cells in chimeras. Deletion did not persist when chimerism was lost. Importantly, although the percentage of regulatory T cells (Tregs) after IUHCTx increased, there was no expansion in Treg numbers. In wild-type mice, there was a similar deletion of effector cells without expansion of donor-specific Tregs. Thus, tolerance induction after IUHCTx depends on both direct and indirect antigen presentation and is secondary to thymic deletion, without de novo Treg induction.
Collapse
|
18
|
Landsman L, Nijagal A, Whitchurch TJ, VanderLaan RL, Zimmer WE, MacKenzie TC, Hebrok M. Pancreatic mesenchyme regulates epithelial organogenesis throughout development. PLoS Biol 2011; 9:e1001143. [PMID: 21909240 PMCID: PMC3167782 DOI: 10.1371/journal.pbio.1001143] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Accepted: 07/28/2011] [Indexed: 01/10/2023] Open
Abstract
Genetic disruption of the pancreatic mesenchyme reveals that it is critical for the expansion of epithelial progenitors and for the proliferation of insulin-producing beta cells. The developing pancreatic epithelium gives rise to all endocrine and exocrine cells of the mature organ. During organogenesis, the epithelial cells receive essential signals from the overlying mesenchyme. Previous studies, focusing on ex vivo tissue explants or complete knockout mice, have identified an important role for the mesenchyme in regulating the expansion of progenitor cells in the early pancreas epithelium. However, due to the lack of genetic tools directing expression specifically to the mesenchyme, the potential roles of this supporting tissue in vivo, especially in guiding later stages of pancreas organogenesis, have not been elucidated. We employed transgenic tools and fetal surgical techniques to ablate mesenchyme via Cre-mediated mesenchymal expression of Diphtheria Toxin (DT) at the onset of pancreas formation, and at later developmental stages via in utero injection of DT into transgenic mice expressing the Diphtheria Toxin receptor (DTR) in this tissue. Our results demonstrate that mesenchymal cells regulate pancreatic growth and branching at both early and late developmental stages by supporting proliferation of precursors and differentiated cells, respectively. Interestingly, while cell differentiation was not affected, the expansion of both the endocrine and exocrine compartments was equally impaired. To further elucidate signals required for mesenchymal cell function, we eliminated β-catenin signaling and determined that it is a critical pathway in regulating mesenchyme survival and growth. Our study presents the first in vivo evidence that the embryonic mesenchyme provides critical signals to the epithelium throughout pancreas organogenesis. The findings are novel and relevant as they indicate a critical role for the mesenchyme during late expansion of endocrine and exocrine compartments. In addition, our results provide a molecular mechanism for mesenchymal expansion and survival by identifying β-catenin signaling as an essential mediator of this process. These results have implications for developing strategies to expand pancreas progenitors and β-cells for clinical transplantation. Embryonic development is a highly complex process that requires tight orchestration of cellular proliferation, differentiation, and migration as cells grow within loosely aggregated mesenchyme and more organized epithelial sheets to form organs and tissues. In addition to intrinsic cell-autonomous signals, these events are further regulated by environmental cues provided by neighboring cells. Prior work demonstrated a critical role for the surrounding mesenchyme in guiding epithelial growth during the early stages of pancreas development. However, it remained unclear whether the mesenchyme also guided the later stages of pancreas organogenesis when the functional exocrine and endocrine cells are formed. Here, we show that specific genetic ablation of the mesenchyme at distinct developmental stages in vivo results in the formation of a smaller, misshapen pancreas. Loss of the mesenchyme profoundly impairs the expansion of both endocrine and exocrine pancreatic progenitors, as well as the proliferative capacity of maturing cells, including insulin-producing beta-cells. Thus, our studies reveal unappreciated roles for the mesenchyme in guiding the formation of the epithelial pancreas throughout development. The results suggest that identifying the specific mesenchymal signals might help to optimize cell culture protocols that aim to achieve the differentiation of stem cells into insulin-producing beta cells.
Collapse
Affiliation(s)
- Limor Landsman
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America
| | - Amar Nijagal
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Surgery, University of California, San Francisco, San Francisco, California, United States of America
| | - Theresa J. Whitchurch
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America
| | - Renee L. VanderLaan
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America
| | - Warren E. Zimmer
- Department of Systems Biology and Translational Medicine, Texas A&M Health Science Center, College Station, Texas, United States of America
| | - Tippi C. MacKenzie
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Surgery, University of California, San Francisco, San Francisco, California, United States of America
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|