1
|
Şenkal-Turhan S, Bulut-Okumuş E, Aydın M, Başak Türkmen N, Taşlıdere A, Şahin F, Yılmaz Ş, Akkuş Süt P, Doğan A. Induced Pluripotent Stem Cell-Derived Parathyroid Organoids Resemble Parathyroid Morphology and Function. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2407567. [PMID: 39331961 DOI: 10.1002/advs.202407567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/12/2024] [Indexed: 09/29/2024]
Abstract
The primary role of the parathyroid glands is to maintain calcium homeostasis through the secretion of parathyroid hormone (PTH). The limited proliferative capacity and differentiation of parathyroid cells hinder the generation of cell therapy options. In this study, parathyroid organoids are successfully generated from human-induced pluripotent stem cells (hiPSCs). At the end of the 20 days of differentiation, the parathyroid organoids exhibited distinct parathyroid morphology. Stereomicroscope, scanning electron microscopy (SEM), and transmission electron microscopy (TEM) analysis demonstrated the 3D arrangement of the cell layers in which intracellular structures of parathyroid cells resemble human parathyroid cellular morphology. Comprehensive molecular analyses, including RNA sequencing (RNA-Seq) and liquid chromatography/mass spectrometry (LC-MS/MS), confirmed the expression of key parathyroid-related markers. Protein expression of CasR, CxCr4, Gcm2, and PTH are observed in parathyroid organoids. Parathyroid organoids secrete PTH, demonstrate active intercellular calcium signaling, and induce osteogenic differentiation via their secretome. The tissue integration potential of parathyroid organoids is determined by transplantation into parathyroidectomized rats. The organoid transplanted animals showed significant elevations in PTH-related markers (CasR, CxCr4, Foxn1, Gcm2, and PTH). PTH secretion is detected in organoid-transplanted animals. The findings represent a significant advancement in parathyroid organoid culture and may offer a cellular therapy for treating PTH-related diseases, including hypoparathyroidism.
Collapse
Affiliation(s)
- Selinay Şenkal-Turhan
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, 34755, Turkey
| | - Ezgi Bulut-Okumuş
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, 34755, Turkey
| | - Muhterem Aydın
- Department of Veterinary Obstetrics and Gynecology, Faculty of Veterinary Medicine, University of Fırat, Elazığ, 23119, Turkey
| | - Neşe Başak Türkmen
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, University of Inonu, Malatya, 44280, Turkey
| | - Aslı Taşlıdere
- Department of Histology and Embryology, Faculty of Medicine, University of Inonu, Malatya, 44280, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, 34755, Turkey
| | - Şahin Yılmaz
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, 34755, Turkey
| | - Pınar Akkuş Süt
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, 34755, Turkey
| | - Ayşegül Doğan
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, 34755, Turkey
| |
Collapse
|
2
|
Wang G, Du Y, Cui X, Xu T, Li H, Dong M, Li W, Li Y, Cai W, Xu J, Li S, Yang X, Wu Y, Chen H, Li X. Directed differentiation of human embryonic stem cells into parathyroid cells and establishment of parathyroid organoids. Cell Prolif 2024; 57:e13634. [PMID: 38494923 PMCID: PMC11294423 DOI: 10.1111/cpr.13634] [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: 10/19/2023] [Revised: 01/25/2024] [Accepted: 03/06/2024] [Indexed: 03/19/2024] Open
Abstract
Differentiation of human embryonic stem cells (hESCs) into human embryonic stem cells-derived parathyroid-like cells (hESC-PT) has clinical significance in providing new therapies for congenital and acquired parathyroid insufficiency conditions. However, a highly reproducible, well-documented method for parathyroid differentiation remains unavailable. By imitating the natural process of parathyroid embryonic development, we proposed a new hypothesis about the in vitro differentiation of parathyroid-like cells. Transcriptome, differentiation marker protein detection and parathyroid hormone (PTH) secretion assays were performed after the completion of differentiation. To optimize the differentiation protocol and further improve the differentiation rate, we designed glial cells missing transcription factor 2 (GCM2) overexpression lentivirus transfection assays and constructed hESCs-derived parathyroid organoids. The new protocol enabled hESCs to differentiate into hESC-PT. HESC-PT cells expressed PTH, GCM2 and CaSR proteins, low extracellular calcium culture could stimulate hESC-PT cells to secrete PTH. hESC-PT cells overexpressing GCM2 protein secreted PTH earlier than their counterpart hESC-PT cells. Compared with the two-dimensional cell culture environment, hESCs-derived parathyroid organoids secreted more PTH. Both GCM2 lentiviral transfection and three-dimensional cultures could make hESC-PT cells functionally close to human parathyroid cells. Our study demonstrated that hESCs could differentiate into hESC-PT in vitro, which paves the road for applying the technology to treat hypoparathyroidism and introduces new approaches in the field of regenerative medicine.
Collapse
Affiliation(s)
- Ge Wang
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yaying Du
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoqing Cui
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Tao Xu
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hanning Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Menglu Dong
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wei Li
- Department of Clinical and Diagnostic SciencesUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Yajie Li
- Department of Rehabilitation, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wenjun Cai
- Department of Rehabilitation, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jia Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Shuyu Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xue Yang
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yonglin Wu
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
3
|
Venkat A, Carlino MJ, Lawton BR, Prasad ML, Amodio M, Gibson CE, Zeiss CJ, Youlten SE, Krishnaswamy S, Krause DS. Single-cell analysis reveals transcriptional dynamics in healthy primary parathyroid tissue. Genome Res 2024; 34:837-850. [PMID: 38977309 PMCID: PMC11293540 DOI: 10.1101/gr.278215.123] [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: 07/01/2023] [Accepted: 06/03/2024] [Indexed: 07/10/2024]
Abstract
Studies on human parathyroids are generally limited to hyperfunctioning glands owing to the difficulty in obtaining normal human tissue. We therefore obtained non-human primate (NHP) parathyroids to provide a suitable alternative for sequencing that would bear a close semblance to human organs. Single-cell RNA expression analysis of parathyroids from four healthy adult M. mulatta reveals a continuous trajectory of epithelial cell states. Pseudotime analysis based on transcriptomic signatures suggests a progression from GCM2 hi progenitors to mature parathyroid hormone (PTH)-expressing epithelial cells with increasing core mitochondrial transcript abundance along pseudotime. We sequenced, as a comparator, four histologically characterized hyperfunctioning human parathyroids with varying oxyphil and chief cell abundance and leveraged advanced computational techniques to highlight similarities and differences from non-human primate parathyroid expression dynamics. Predicted cell-cell communication analysis reveals abundant endothelial cell interactions in the parathyroid cell microenvironment in both human and NHP parathyroid glands. We show abundant RARRES2 transcripts in both human adenoma and normal primate parathyroid cells and use coimmunostaining to reveal high levels of RARRES2 protein (also known as chemerin) in PTH-expressing cells, which could indicate that RARRES2 plays an unrecognized role in parathyroid endocrine function. The data obtained are the first single-cell RNA transcriptome to characterize nondiseased parathyroid cell signatures and to show a transcriptomic progression of cell states within normal parathyroid glands, which can be used to better understand parathyroid cell biology.
Collapse
Affiliation(s)
- Aarthi Venkat
- Computational Biology and Bioinformatics Program, Yale University, New Haven, Connecticut 06511, USA
| | - Maximillian J Carlino
- Yale Stem Cell Center, Yale School of Medicine, New Haven, Connecticut 06520, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Betty R Lawton
- Yale Stem Cell Center, Yale School of Medicine, New Haven, Connecticut 06520, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Manju L Prasad
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut 06520-8023, USA
| | - Matthew Amodio
- Department of Computer Science, Yale University, New Haven, Connecticut 06511, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Courtney E Gibson
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut 06520, USA
| | - Caroline J Zeiss
- Department of Comparative Medicine, Yale School of Medicine, New Haven, Connecticut 06520, USA
| | - Scott E Youlten
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Smita Krishnaswamy
- Computational Biology and Bioinformatics Program, Yale University, New Haven, Connecticut 06511, USA;
- Yale Stem Cell Center, Yale School of Medicine, New Haven, Connecticut 06520, USA
- Department of Computer Science, Yale University, New Haven, Connecticut 06511, USA
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Diane S Krause
- Yale Stem Cell Center, Yale School of Medicine, New Haven, Connecticut 06520, USA;
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut 06510, USA
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut 06520-8023, USA
| |
Collapse
|
4
|
Lee S, Jung HI, Lee J, Kim Y, Chung J, Kim HS, Lim J, Nam KC, Lim YS, Choi HS, Kwak BS. Parathyroid-on-a-chip simulating parathyroid hormone secretion in response to calcium concentration. LAB ON A CHIP 2024; 24:3243-3251. [PMID: 38836406 DOI: 10.1039/d4lc00249k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
The parathyroid gland is an endocrine organ that plays a crucial role in regulating calcium levels in blood serum through the secretion of parathyroid hormone (PTH). Hypoparathyroidism is a chronic disease that can occur due to parathyroid defects, but due to the difficulty of creating animal models of this disease or obtaining human normal parathyroid cells, the evaluation of parathyroid functionality for drug development is limited. Although parathyroid-like cells that secrete PTH have recently been reported, their functionality may be overestimated using traditional culture methods that lack in vivo similarities, particularly vascularization. To overcome these limitations, we obtained parathyroid organoids from tonsil-derived mesenchymal stem cells (TMSCs) and fabricated a parathyroid-on-a-chip, capable of simulating PTH secretion based on calcium concentration. This chip exhibited differences in PTH secretion according to calcium concentration and secreted PTH within the range of normal serum levels. In addition, branches of organoids, which are difficult to observe in animal models, were observed in this chip. This could serve as a guideline for successful engraftment in implantation therapies in the future.
Collapse
Affiliation(s)
- Sunghan Lee
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seadaemun-gu, Seoul, 13722, Republic of Korea
- College of Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyangsi, Gyeonggi-do, 10326, Republic of Korea.
| | - Hyo-Il Jung
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seadaemun-gu, Seoul, 13722, Republic of Korea
- The DABOM Inc., 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jaehun Lee
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seadaemun-gu, Seoul, 13722, Republic of Korea
- College of Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyangsi, Gyeonggi-do, 10326, Republic of Korea.
| | - Youngwon Kim
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seadaemun-gu, Seoul, 13722, Republic of Korea
- College of Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyangsi, Gyeonggi-do, 10326, Republic of Korea.
| | - Jaewoo Chung
- Department of Laboratory Medicine, Dongguk University Ilsan Hospital, 27 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Republic of Korea
| | - Han Su Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, Ewha Womans University, School of Medicine, Seoul 158-710, Republic of Korea
| | - Jiseok Lim
- School of Mechanical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan-si, Gyeongsangbuk-do, 38541, Republic of Korea
- MediSphere Inc., 280, Daehak-ro, Gyeongsan-si, Gyeongsangbuk-do, 38541, Republic of Korea
| | - Ki Chang Nam
- College of Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyangsi, Gyeonggi-do, 10326, Republic of Korea.
| | - Yun-Sung Lim
- Department of Otorhinolaryngology -Head and Neck Surgery, Dongguk University Ilsan Hospital, 27 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Republic of Korea.
| | - Han Seok Choi
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Dongguk University Ilsan Hospital, 27 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Republic of Korea.
| | - Bong Seop Kwak
- College of Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyangsi, Gyeonggi-do, 10326, Republic of Korea.
- MediSphere Inc., 280, Daehak-ro, Gyeongsan-si, Gyeongsangbuk-do, 38541, Republic of Korea
| |
Collapse
|
5
|
Chen C, Gao D, Luo L, Qu R, Hu X, Wang Y, Guo Y. Parathyroid preservation in total endoscopic thyroid surgeries via the mammary areolas approach: Real-world data from a single center. Asian J Surg 2023; 46:5421-5428. [PMID: 37344318 DOI: 10.1016/j.asjsur.2023.05.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/22/2023] [Accepted: 05/31/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Preserving parathyroid glands in situ is crucial to avoid surgical hypoparathyroidism, but it is also one of the greatest challenges during thyroid surgery. Magnified endoscopic imaging has been proposed as a way to improve parathyroid preservation. METHODS 2,603 consecutive patients who underwent thyroid surgery at the First People's Hospital of Zunyi from January 2018 to July 2022 were screened. 1,355 patients were eligible, including 965 endoscopic and 390 open cases. Parathyroid hormone (PTH) loss levels and severe parathyroid injury rates were compared between endoscopic and open cases. Meanwhile, factors that contribute to parathyroid injuries were assessed, including surgical extent, tumor size, carbon nanoparticle guidance, and surgical proficiency. RESULTS PTH loss levels were similar between endoscopic and open cases (P = 0.440). The incidence of severe parathyroid injuries was also comparable (7.8% for endoscopic vs. 6.9% for open, P = 0.592). The endoscopic group had higher rates of autologous parathyroid transplantation (39.5% vs. 24.4%, P = 0.000), while accidental parathyroidectomy rates were similar (11.4% vs. 10.8%, P = 0.739). Among patients who received the same extent of thyroid surgeries, no significant difference was found in PTH loss levels and severe parathyroid injury rates, except for a higher risk of severe parathyroid injuries in endoscopic bilateral thyroidectomy (18.52% vs. 11.52%, P = 0.033). CONCLUSIONS Despite the magnified endoscopic imaging facilitating the identification of parathyroid tissues, endoscopic approaches are not superior to open ones for the in-situ preservation of parathyroid glands. For a bilateral thyroidectomy, open approaches are safer for parathyroid preservation.
Collapse
Affiliation(s)
- Chen Chen
- Breast and Thyroid Center, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, 563000, Zunyi, Guizhou, China.
| | - Dan Gao
- Breast and Thyroid Center, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, 563000, Zunyi, Guizhou, China.
| | - Libo Luo
- Breast and Thyroid Center, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, 563000, Zunyi, Guizhou, China.
| | - Rui Qu
- Breast and Thyroid Center, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, 563000, Zunyi, Guizhou, China.
| | - Xiaochi Hu
- Breast and Thyroid Center, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, 563000, Zunyi, Guizhou, China.
| | - Yixiao Wang
- Department of Surgery, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, 563000, Zunyi, Guizhou, China.
| | - Youming Guo
- Breast and Thyroid Center, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, 563000, Zunyi, Guizhou, China.
| |
Collapse
|
6
|
Nakatsuka R, Kato T, Zhang R, Uemura Y, Sasaki Y, Matsuoka Y, Shirouzu Y, Fujioka T, Yamashita H, Hattori F, Nozaki T, Ogata H, Hitomi H. The Induction of Parathyroid Cell Differentiation from Human Induced Pluripotent Stem Cells Promoted Via TGF-α/EGFR Signaling. Stem Cells Dev 2023; 32:670-680. [PMID: 37639359 DOI: 10.1089/scd.2023.0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023] Open
Abstract
The parathyroid gland plays an essential role in mineral and bone metabolism. Cultivation of physiological human parathyroid cells has yet to be established and the method by which parathyroid cells differentiate from pluripotent stem cells remains uncertain. Therefore, it has been hard to clarify the mechanisms underlying the onset of parathyroid disorders, such as hyperparathyroidism. In this study, we developed a new method of parathyroid cell differentiation from human induced pluripotent stem (iPS) cells. Parathyroid cell differentiation occurred in accordance with embryologic development. Differentiated cells, which expressed the parathyroid hormone, adopted unique cell aggregation similar to the parathyroid gland. In addition, these differentiated cells were identified as calcium-sensing receptor (CaSR)/epithelial cell adhesion molecule (EpCAM) double-positive cells. Interestingly, stimulation with transforming growth factor-α (TGF-α), which is considered a causative molecule of parathyroid hyperplasia, increased the CaSR/EpCAM double-positive cells, but this effect was suppressed by erlotinib, which is an epidermal growth factor receptor (EGFR) inhibitor. These results suggest that TGF-α/EGFR signaling promotes parathyroid cell differentiation from iPS cells in a similar manner to parathyroid hyperplasia.
Collapse
Affiliation(s)
- Ryusuke Nakatsuka
- Department of iPS Stem Cell Regenerative Medicine, Faculty of Medicine, Kansai Medical University, Osaka, Japan
- Department of Pharmacology, Faculty of Dentistry, Osaka Dental University, Osaka, Japan
| | - Tadashi Kato
- Department of iPS Stem Cell Regenerative Medicine, Faculty of Medicine, Kansai Medical University, Osaka, Japan
- Division of Nephrology, Department of Medicine, Showa University, Tokyo, Japan
- Department of Internal Medicine, Showa University Northern Yokohama Hospital, Kanagawa, Japan
| | - Rong Zhang
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Yasushi Uemura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Yuka Sasaki
- Department of Pharmacology, Faculty of Dentistry, Osaka Dental University, Osaka, Japan
| | - Yoshikazu Matsuoka
- Department of iPS Stem Cell Regenerative Medicine, Faculty of Medicine, Kansai Medical University, Osaka, Japan
| | - Yasumasa Shirouzu
- Department of iPS Stem Cell Regenerative Medicine, Faculty of Medicine, Kansai Medical University, Osaka, Japan
| | - Tatsuya Fujioka
- Department of iPS Stem Cell Regenerative Medicine, Faculty of Medicine, Kansai Medical University, Osaka, Japan
| | - Hiromi Yamashita
- Department of iPS Stem Cell Regenerative Medicine, Faculty of Medicine, Kansai Medical University, Osaka, Japan
| | - Fumiyuki Hattori
- Department of iPS Stem Cell Regenerative Medicine, Faculty of Medicine, Kansai Medical University, Osaka, Japan
| | - Tadashige Nozaki
- Department of Pharmacology, Faculty of Dentistry, Osaka Dental University, Osaka, Japan
| | - Hiroaki Ogata
- Department of Internal Medicine, Showa University Northern Yokohama Hospital, Kanagawa, Japan
| | - Hirofumi Hitomi
- Department of iPS Stem Cell Regenerative Medicine, Faculty of Medicine, Kansai Medical University, Osaka, Japan
| |
Collapse
|
7
|
Kano M, Mizuno N, Sato H, Kimura T, Hirochika R, Iwasaki Y, Inoshita N, Nagano H, Kasai M, Yamamoto H, Yamaguchi T, Suga H, Masaki H, Mizutani E, Nakauchi H. Functional calcium-responsive parathyroid glands generated using single-step blastocyst complementation. Proc Natl Acad Sci U S A 2023; 120:e2216564120. [PMID: 37379351 PMCID: PMC10334775 DOI: 10.1073/pnas.2216564120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 04/24/2023] [Indexed: 06/30/2023] Open
Abstract
Patients with permanent hypoparathyroidism require lifelong replacement therapy to avoid life-threatening complications, The benefits of conventional treatment are limited, however. Transplanting a functional parathyroid gland (PTG) would yield better results. Parathyroid gland cells generated from pluripotent stem cells in vitro to date cannot mimic the physiological responses to extracellular calcium that are essential for calcium homeostasis. We thus hypothesized that blastocyst complementation (BC) could be a better strategy for generating functional PTG cells and compensating loss of parathyroid function. We here describe generation of fully functional PTGs from mouse embryonic stem cells (mESCs) with single-step BC. Using CRISPR-Cas9 knockout of Glial cells missing2 (Gcm2), we efficiently produced aparathyroid embryos for BC. In these embryos, mESCs differentiated into endocrinologically mature PTGs that rescued Gcm2-/- mice from neonatal death. The mESC-derived PTGs responded to extracellular calcium, restoring calcium homeostasis on transplantation into mice surgically rendered hypoparathyroid. We also successfully generated functional interspecies PTGs in Gcm2-/- rat neonates, an accomplishment with potential for future human PTG therapy using xenogeneic animal BC. Our results demonstrate that BC can produce functional endocrine organs and constitute a concept in treatment of hypoparathyroidism.
Collapse
Affiliation(s)
- Mayuko Kano
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Metabolism and Endocrinology, Department of Medicine, St. Marianna University School of Medicine, Miyamae-ku, Kawasaki, Kanagawa216-8511, Japan
| | - Naoaki Mizuno
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Hideyuki Sato
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Takaharu Kimura
- Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki305-8577, Japan
| | - Rei Hirochika
- Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki305-8577, Japan
| | - Yasumasa Iwasaki
- Department of Clinical Nutrition, Faculty of Health Science, Suzuka University of Medical Science, Suzuka, Mie510-0293, Japan
- Department of Endocrinology, Metabolism, and Nephrology, Kochi Medical School, Kochi University, Oko-cho, Nankoku, Kochi783-8505, Japan
| | - Naoko Inoshita
- Department of Pathology, Moriyama Memorial Hospital, Edogawa-ku, Tokyo134-0081, Japan
| | - Hisato Nagano
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Department of Plastic and Reconstructive Surgery, National Defense Medical College, Tokorozawa, Saitama359-8513, Japan
| | - Mariko Kasai
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Hiromi Yamamoto
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Tomoyuki Yamaguchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Laboratory of Regenerative Medicine, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo192-0392, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya466-8550, Japan
| | - Hideki Masaki
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Eiji Mizutani
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki305-8577, Japan
| | - Hiromitsu Nakauchi
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
8
|
Karabiyik Acar Ö, Nozhatzadeh GD, Tuncer A, Torun Köse G, Hacihasanoğlu E, Sahin F, Aysan E. Production of parathyroid-like cells from thyroid stem cells in co-culture environment. Medicine (Baltimore) 2022; 101:e32009. [PMID: 36482540 PMCID: PMC9726294 DOI: 10.1097/md.0000000000032009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Parathyroid-like cells were aimed to be developed using cells isolated from thyroid since their embryological origins are the same. METHOD Activin A and sonic hedgehog (Shh) are the proteins used in differentiation (dif) medium. Parathyroid and thyroid cells were cultured in a 3-dimensional environment and divided into five groups: thyroid standard (st) medium, thyroid dif medium, parathyroid st medium, thyroid-parathyroid co-culture st medium, and thyroid-parathyroid co-culture dif medium. Throughout 28 days of incubation, groups were investigated by carrying out the live dead assay, confocal microscopy, real-time PCR, immunohistochemistry and biochemical assays. RESULTS Thyroid-parathyroid co-culture cells grown in dif medium exhibited upregulated expressions of parathormone (PTH) (5.1-fold), PTH1R (3.6-fold), calcium sensing receptor (CaSR) (8.8-fold), and loss of thyroid-specific thyroid transcription factor 1 (TTF1) expression when compared to the thyroid st medium group. PTH secretion decreased by 35% in the parathyroid st medium group and 99.9% in the thyroid-parathyroid co-culture st medium group but decreased only 3.5% in the thyroid-parathyroid co-culture dif medium group on day 28. CONCLUSION Using Activin A and Shh proteins, thyroid stem/progenitor cells were differentiated to parathyroid-like cells successfully in a co-culture environment. A potentially effective novel method for cell differenatiation is co-culture of cells having the same embryological origin.
Collapse
Affiliation(s)
- Özge Karabiyik Acar
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul, Turkey
| | - Gülçin Delal Nozhatzadeh
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul, Turkey
| | - Alperen Tuncer
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Gamze Torun Köse
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Ezgi Hacihasanoğlu
- Department of Pathology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Fikrettin Sahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Erhan Aysan
- Department of General Surgery, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
- * Correspondence: Erhan Aysan, Department of General Surgery, Faculty of Medicine, Yeditepe University, ATA-2 Sitesi, Akasya Cad. No:25, Cengelkoy, Uskudar, Istanbul 34900, Turkey (e-mail: )
| |
Collapse
|
9
|
Noltes ME, Sondorp LH, Kracht L, Antunes IF, Wardenaar R, Kelder W, Kemper A, Szymanski W, Zandee WT, Jansen L, Brouwers AH, Coppes RP, Kruijff S. Patient-derived parathyroid organoids as a tracer and drug-screening application model. Stem Cell Reports 2022; 17:2518-2530. [PMID: 36306782 PMCID: PMC9669499 DOI: 10.1016/j.stemcr.2022.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/29/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022] Open
Abstract
Parathyroid diseases are characterized by dysregulation of calcium homeostasis and alterations in parathyroid hormone (PTH) excretion. The development of parathyroid-targeted treatment and imaging tracers could benefit from in vitro models. Therefore, we aim to establish a patient-derived parathyroid organoid model representing human parathyroid tissue. Hyperplastic parathyroid tissue was dispersed, and parathyroid organoids (PTOs) were cultured and characterized. PTO-derived cells exhibited self-renewal over several passages, indicative of the presence of putative stem cells. Immunofluorescence and RNA sequencing confirmed that PTOs phenocopy hyperplastic parathyroid tissue. Exposure of PTOs to increasing calcium concentrations and PTH-lowering drugs resulted in significantly reduced PTH excretion. PTOs showed specific binding of the imaging tracers 11C-methionine and 99mTc-sestamibi. These data show the functionality of PTOs resembling the parathyroid. This PTO model recapitulates the originating tissue on gene and protein expression and functionality, paving the way for future physiology studies and therapeutic target and tracer discovery.
Collapse
|
10
|
Parathyroid Cell Differentiation from Progenitor Cells and Stem Cells: Development, Molecular Mechanism, Function, and Tissue Engineering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1387:13-24. [PMID: 34981451 DOI: 10.1007/5584_2021_694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Parathyroid glands are endocrine organs which are located posterior to thyroid glands and control secretion of parathyroid hormone (PTH) in order to regulate blood calcium level. PTH maintains calcium homeostasis by acting on the bone, kidney, and small intestine. PTH deficiency leads to chronic hypocalcemia, organ calcinosis, kidney and heart failure, painful muscle spasms, neuromuscular problems, and memory problems. Since parathyroid cells have inadequate proliferation potential in culture conditions, their utilization as a cellular therapy option is very limited. Although studies conducted so far include parathyroid cell differentiation from various cell types, problems related to successful cellular differentiation and transplantation still remain. Recently, parathyroid tissue engineering has attracted attention as a potential treatment for the parathyroid-related diseases caused by hypoparathyroidism. Although major progression is made in the construction of tissue engineering protocols using parathyroid cells and biomaterials, PTH secretion to mimic its spontaneous harmony in the body is a challenge. This chapter comprehensively defines the derivation of parathyroid cells from various cell sources including pluripotent stem cells, molecular mechanisms, and tissue engineering applications.
Collapse
|
11
|
Development of Surgically Transplantable Parathyroid Hormone-Releasing Microbeads. Biomedicines 2022; 10:biomedicines10020440. [PMID: 35203648 PMCID: PMC8962264 DOI: 10.3390/biomedicines10020440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 01/27/2023] Open
Abstract
Hypoparathyroidism is an endocrine disorder that occurs because of the inability to produce parathyroid hormone (PTH) effectively. Previously, we reported the efficacy of tonsil-derived mesenchymal stem cells (TMSCs) differentiated into parathyroid-like cells for the treatment of hypoparathyroidism. Here, we investigated the feasibility of three-dimensional structural microbeads fabricated with TMSCs and alginate, a natural biodegradable polymer, to treat hypoparathyroidism. Alginate microbeads were fabricated by dropping a 2% (w/v) alginate solution containing TMSCs into a 5% CaCl2 solution and then differentiated into parathyroid-like cells using activin A and sonic hedgehog for 7 days. The protein expression of PTH, a specific marker of the parathyroid gland, was significantly higher in differentiated alginate microbeads with TMSCs (Al-dT) compared with in undifferentiated alginate microbeads with TMSCs. For in vivo experiments, we created the hypoparathyroidism animal model by parathyroidectomy (PTX) and implanted alginate microbeads in the dorsal interscapular region. The PTX rats with Al-dT (PTX+Al-dT) showed the highest survival rate and weight change and a gradual increase in serum intact PTH levels. We also detected a higher expression of PTH in retrieved tissues of PTX+Al-dT using immunofluorescence analysis. This study demonstrates that alginate microbeads are potential a new tool as a surgically scalable therapy for treating hypoparathyroidism.
Collapse
|
12
|
Kim JY, Park S, Oh SY, Nam YH, Choi YM, Choi Y, Kim HY, Jung SY, Kim HS, Jo I, Jung SC. Density-Dependent Differentiation of Tonsil-Derived Mesenchymal Stem Cells into Parathyroid-Hormone-Releasing Cells. Int J Mol Sci 2022; 23:ijms23020715. [PMID: 35054901 PMCID: PMC8775366 DOI: 10.3390/ijms23020715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can differentiate into endoderm lineages, especially parathyroid-hormone (PTH)-releasing cells. We have previously reported that tonsil-derived MSC (T-MSC) can differentiate into PTH-releasing cells (T-MSC-PTHCs), which restored the parathyroid functions in parathyroidectomy (PTX) rats. In this study, we demonstrate quality optimization by standardizing the differentiation rate for a better clinical application of T-MSC-PTHCs to overcome donor-dependent variation of T-MSCs. Quantitation results of PTH mRNA copy number in the differentiated cells and the PTH concentration in the conditioned medium confirmed that the differentiation efficiency largely varied depending on the cells from each donor. In addition, the differentiation rate of the cells from all the donors greatly improved when differentiation was started at a high cell density (100% confluence). The large-scale expression profiling of T-MSC-PTHCs by RNA sequencing indicated that those genes involved in exiting the differentiation and the cell cycle were the major pathways for the differentiation of T-MSC-PTHCs. Furthermore, the implantation of the T-MSC-PTHCs, which were differentiated at a high cell density embedded in hyaluronic acid, resulted in a higher serum PTH in the PTX model. This standardized efficiency of differentiation into PTHC was achieved by initiating differentiation at a high cell density. Our findings provide a potential solution to overcome the limitations due to donor-dependent variation by establishing a standardized differentiation protocol for the clinical application of T-MSC therapy in treating hypoparathyroidism.
Collapse
Affiliation(s)
- Ji Yeon Kim
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (J.Y.K.); (S.P.); (Y.H.N.); (Y.C.)
| | - Saeyoung Park
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (J.Y.K.); (S.P.); (Y.H.N.); (Y.C.)
| | - Se-Young Oh
- Departments of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (S.-Y.O.); (Y.M.C.); (I.J.)
| | - Yu Hwa Nam
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (J.Y.K.); (S.P.); (Y.H.N.); (Y.C.)
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 07804, Korea
| | - Young Min Choi
- Departments of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (S.-Y.O.); (Y.M.C.); (I.J.)
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 07804, Korea
| | - Yeonzi Choi
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (J.Y.K.); (S.P.); (Y.H.N.); (Y.C.)
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 07804, Korea
| | - Ha Yeong Kim
- Departments of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (H.Y.K.); (S.Y.J.); (H.S.K.)
| | - Soo Yeon Jung
- Departments of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (H.Y.K.); (S.Y.J.); (H.S.K.)
| | - Han Su Kim
- Departments of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (H.Y.K.); (S.Y.J.); (H.S.K.)
| | - Inho Jo
- Departments of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (S.-Y.O.); (Y.M.C.); (I.J.)
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 07804, Korea
| | - Sung-Chul Jung
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (J.Y.K.); (S.P.); (Y.H.N.); (Y.C.)
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 07804, Korea
- Correspondence: ; Tel.: +82-2-6986-6199
| |
Collapse
|
13
|
Hypoparathyroidism: State of the Art on Cell and Tissue Therapies. Int J Mol Sci 2021; 22:ijms221910272. [PMID: 34638612 PMCID: PMC8508771 DOI: 10.3390/ijms221910272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022] Open
Abstract
Hypoparathyroidism is an endocrine disorder characterized by low serum calcium levels, high serum phosphorus levels, and by inappropriate or absent secretion of the parathyroid hormone (PTH). The most common therapeutic strategy to treat this condition is hormone replacement therapy with calcium and vitamin D but, unfortunately, in the long term this treatment may not be sufficient to compensate for the loss of endocrine function. Glandular autotransplantation is considered the most effective technique in place of replacement therapy. Although it leads to excellent results in most cases, autotransplantation is not always possible. Allograft is a good way to treat patients who have not been able to undergo autograft, but this technique has limited success due to side effects related to tissue rejection. This therapy is supported by systemic immunosuppression, which leads to the onset of serious side effects in patients, with a risk of endocrine toxicity. Today, research on endocrine disorders is focused on discovering alternative graft therapies that can allow optimal results with the fewest possible side effects. In this review, we will make an update on the current state of the art about the cell and tissue therapy as treatment for hypoparathyroidism, to identify which type of therapeutic strategy could be valid for a future clinical use.
Collapse
|
14
|
Mihai R, Thakker RV. MANAGEMENT OF ENDOCRINE DISEASE: Postsurgical hypoparathyroidism: current treatments and future prospects for parathyroid allotransplantation. Eur J Endocrinol 2021; 184:R165-R175. [PMID: 33599211 PMCID: PMC8052514 DOI: 10.1530/eje-20-1367] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/17/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Permanent postsurgical hypoparathyroidism (POSH) is a major complication of anterior neck surgery in general and of thyroid surgery in particular. Depending on diagnostic criteria, up to 10% of patients undergoing bilateral thyroid surgery develop POSH. This leads to a multitude of symptoms that decrease the quality of life and burden the healthcare provision through complex needs for medication and treatment of specific complications, such as seizures and laryngospasm. METHODS Narrative review of current medical treatments for POSH and of the experience accumulated with parathyroid allotransplantation. RESULTS In most patients, POSH is controlled with regular use of calcium supplements and active vitamin D analogues but a significant proportion of patients continue to experience severe symptoms requiring repeated emergency admissions. Replacement therapy with synthetic PTH compounds (PTH1-34, Natpara® and PTH1-84, teriparatide, Forsteo®) has been assessed in multicentre trials, but the use of this medication is restricted by costs and concerns related to the risk of development of osteosarcoma. Based on recent case reports of successful allotransplantation of parathyroid tissue between siblings, there is renewed interest in this technique. Data on selection of donors, parathyroid cell preparation before allotransplantation, site and timing of transplantation, need for immunosuppression and long-term outcomes are reviewed. CONCLUSION A prospective trial to assess the efficacy of parathyroid allotransplantation in patients with severely symptomatic protracted post-surgical hypoparathyroidism is warranted.
Collapse
Affiliation(s)
- Radu Mihai
- Department of Endocrine Surgery, Churchill Cancer Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, Oxfordshire, UK
- Correspondence should be addressed to R Mihai;
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Goliusova DV, Klementieva NV, Panova AV, Mokrysheva NG, Kiselev SL. The Role of Genetic Factors in Endocrine Tissues Development and Its Regulation In Vivo and In Vitro. RUSS J GENET+ 2021. [DOI: 10.1134/s102279542103008x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
16
|
Lawton BR, Martineau C, Sosa JA, Roman S, Gibson CE, Levine MA, Krause DS. Differentiation of PTH-Expressing Cells From Human Pluripotent Stem Cells. Endocrinology 2020; 161:5893997. [PMID: 32810225 PMCID: PMC7505176 DOI: 10.1210/endocr/bqaa141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022]
Abstract
Differentiation of pluripotent stem cells into functional parathyroid-like cells would accelerate development of important therapeutic options for subjects with parathyroid-related disorders, from the design and screening of novel pharmaceutical agents to the development of durable cellular therapies. We have established a highly reproducible directed differentiation approach leading to PTH-expressing cells from human embryonic stem cells and induced pluripotent stem cells. We accomplished this through the comparison of multiple different basal media, the inclusion of the CDK inhibitor PD0332991 in both definitive endoderm and anterior foregut endoderm stages, and a 2-stage pharyngeal endoderm series. This is the first protocol to reproducibly establish PTH-expressing cells from human pluripotent stem cells and represents a first step toward the development of functional parathyroid cells with broad applicability for medicinal and scientific investigation.
Collapse
Affiliation(s)
- Betty R Lawton
- Department of Laboratory Medicine, Yale Stem Cell Center, Yale University, New Haven, Connecticut
| | - Corine Martineau
- Center for Bone Health and Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Julie Ann Sosa
- Department of Surgery, University of California San Francisco, San Francisco, California
| | - Sanziana Roman
- Department of Surgery, University of California San Francisco, San Francisco, California
| | | | - Michael A Levine
- Center for Bone Health and Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Diane S Krause
- Department of Laboratory Medicine, Cell Biology, Yale Stem Cell Center, Yale University, New Haven, Connecticut
- Department of Pathology, Yale Stem Cell Center, Yale University, New Haven, Connecticut
- Correspondence: Diane S. Krause, Yale University, Yale Stem Cell Center, 333 Cedar Street, New Haven, Connecticut 06520-8035, USA. E-mail:
| |
Collapse
|
17
|
Li D, Guo B, Liang Q, Liu Y, Zhang L, Hu N, Zhang X, Yang F, Ruan C. Tissue-engineered parathyroid gland and its regulatory secretion of parathyroid hormone. J Tissue Eng Regen Med 2020; 14:1363-1377. [PMID: 32511868 DOI: 10.1002/term.3080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 11/11/2022]
Abstract
Parathyroid glands (PTGs) are important endocrine organs being mainly responsible for the secretion of parathyroid hormone (PTH) to regulate the balance of calcium (Ca) /phosphorus (P) ions in the body. Once PTGs get injured or removed, their resulting defect or loss of PTH secretion should disturb the level of Ca/P in blood, thus damaging other related organs (bone, kidney, etc.) and even causing death. Recently, tissue-engineered PTGs (TE-PTGs) have attracted lots of attention as a potential treatment for the related diseases of PTGs caused by hypoparathyroidism and hyperparathyroidism, including tetany, muscle cramp, nephrolithiasis, nephrocalcinosis, and osteoporosis. Although great progress has been made in the establishment of TE-PTGs with an effective strategy to integrate the key factors of cells and biomaterials, its regulatory secretion of PTH to mimic its natural rhythms in the body remains a huge challenge. This review comprehensively describes an overview of PTGs from physiology and pathology to cytobiology and tissue engineering. The state of the arts in TE-PTGs and the feasible strategies to regulate PTH secretion behaviors are highlighted to provide an important foundation for further investigation.
Collapse
Affiliation(s)
- Duo Li
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China.,University of Chinese Academy of Sciences, Beijing, PR China
| | - Baochun Guo
- Department of Nephrology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, PR China.,Key Laboratory of Shenzhen Renal Diseases, Shenzhen, PR China
| | - Qingfei Liang
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China.,University of Chinese Academy of Sciences, Beijing, PR China
| | - Yunhui Liu
- University of Chinese Academy of Sciences, Beijing, PR China.,The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Lu Zhang
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Nan Hu
- Department of Nephrology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, PR China.,Key Laboratory of Shenzhen Renal Diseases, Shenzhen, PR China
| | - Xinzhou Zhang
- Department of Nephrology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, PR China.,Key Laboratory of Shenzhen Renal Diseases, Shenzhen, PR China
| | - Fan Yang
- University of Chinese Academy of Sciences, Beijing, PR China.,The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Changshun Ruan
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China.,University of Chinese Academy of Sciences, Beijing, PR China
| |
Collapse
|
18
|
Differentiation of human pluripotent stem cells toward pharyngeal endoderm derivatives: Current status and potential. Curr Top Dev Biol 2020; 138:175-208. [PMID: 32220297 DOI: 10.1016/bs.ctdb.2020.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The pharyngeal apparatus, a transient embryological structure, includes diverse cells from all three germ layers that ultimately contribute to a variety of adult tissues. In particular, pharyngeal endoderm produces cells of the inner ear, palatine tonsils, the thymus, parathyroid and thyroid glands, and ultimobranchial bodies. Each of these structures and organs contribute to vital human physiological processes, including central immune tolerance (thymus) and metabolic homeostasis (parathyroid and thyroid glands, and ultimobranchial bodies). Thus, improper development or damage to pharyngeal endoderm derivatives leads to complicated and severe human maladies, such as autoimmunity, immunodeficiency, hypothyroidism, and/or hypoparathyroidism. To study and treat such diseases, we can utilize human pluripotent stem cells (hPSCs), which differentiate into functionally mature cells in vitro given the proper developmental signals. Here, we discuss current efforts regarding the directed differentiation of hPSCs toward pharyngeal endoderm derivatives. We further discuss model system and therapeutic applications of pharyngeal endoderm cell types produced from hPSCs. Finally, we provide suggestions for improving hPSC differentiation approaches to pharyngeal endoderm derivatives with emphasis on current single cell-omics and 3D culture system technologies.
Collapse
|
19
|
Zhang P, Zhang H, Dong W, Wang Z, Qin Y, Wu C, Dong Q. Differentiation of Rat Adipose-Derived Stem Cells into Parathyroid-Like Cells. Int J Endocrinol 2020; 2020:1860842. [PMID: 32612651 PMCID: PMC7306084 DOI: 10.1155/2020/1860842] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 06/08/2019] [Accepted: 07/04/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND The current treatment for postoperative hypoparathyroidism has shortcomings, such as repeated blood monitoring for dosage adjustment, uncertain long-term efficacy, and the high price of recombinant parathyroid hormone therapy. Adipose-derived stem cells can undergo adipogenic and osteogenic differentiation in vitro and are considered a novel source of parathyroid-like cells, but the idea lacks theoretical basis and feasibility. We aimed at establishing a protocol for differentiating adipose-derived stem cells into parathyroid-like cells for treating hypoparathyroidism. MATERIALS /. METHODS Adipose-derived stem cells were isolated and purified from the inguinal adipose tissue of Sprague Dawley rats. Adipogenic differentiation and osteogenic differentiation of the cells were identified by oil red O and alizarin red S staining, respectively. The adipose-derived stem cells were stimulated by sonic hedgehog (SHH) and activin A. The differentiation of the adipose-derived stem cells to parathyroid-like cells was confirmed by the detection of parathyroid hormone and the related parathyroid markers. RESULTS Adipose-derived stem cells were successfully isolated and purified from the rat adipocytes. The adipogenic and osteogenic differentiation capabilities of the adipose-derived stem cells were determined. SHH and activin A stimulated parathyroid hormone secretion by the adipose-derived stem cells and significantly increased the expression of calcium-sensing receptor (CaSR), parathyroid hormone, and glial cells missing homolog 2 (GCM2) in the cells in a time- and concentration-dependent manner. CONCLUSION We successfully differentiated rat adipose-derived stem cells into parathyroid-like cells, which will pave a new route to curing hypoparathyroidism.
Collapse
Affiliation(s)
- Ping Zhang
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Hao Zhang
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Wenwu Dong
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Zhihong Wang
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Yuan Qin
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Changhao Wu
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Qi Dong
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
- Department of General Surgery, The People's Hospital of China Medical University, Shenyang, Liaoning Province 110016, China
| |
Collapse
|
20
|
Mariniello K, Ruiz-Babot G, McGaugh EC, Nicholson JG, Gualtieri A, Gaston-Massuet C, Nostro MC, Guasti L. Stem Cells, Self-Renewal, and Lineage Commitment in the Endocrine System. Front Endocrinol (Lausanne) 2019; 10:772. [PMID: 31781041 PMCID: PMC6856655 DOI: 10.3389/fendo.2019.00772] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022] Open
Abstract
The endocrine system coordinates a wide array of body functions mainly through secretion of hormones and their actions on target tissues. Over the last decades, a collective effort between developmental biologists, geneticists, and stem cell biologists has generated a wealth of knowledge related to the contribution of stem/progenitor cells to both organogenesis and self-renewal of endocrine organs. This review provides an up-to-date and comprehensive overview of the role of tissue stem cells in the development and self-renewal of endocrine organs. Pathways governing crucial steps in both development and stemness maintenance, and that are known to be frequently altered in a wide array of endocrine disorders, including cancer, are also described. Crucially, this plethora of information is being channeled into the development of potential new cell-based treatment modalities for endocrine-related illnesses, some of which have made it through clinical trials.
Collapse
Affiliation(s)
- Katia Mariniello
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gerard Ruiz-Babot
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Emily C. McGaugh
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - James G. Nicholson
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Angelica Gualtieri
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Maria Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
21
|
Ghanavatinejad F, Fard Tabrizi ZP, Omidghaemi S, Sharifi E, Møller SG, Jami MS. Protein biomarkers of neural system. J Otol 2019; 14:77-88. [PMID: 31467504 PMCID: PMC6712353 DOI: 10.1016/j.joto.2019.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/14/2019] [Accepted: 03/20/2019] [Indexed: 11/30/2022] Open
Abstract
The utilization of biomarkers for in vivo and in vitro research is growing rapidly. This is mainly due to the enormous potential of biomarkers in evaluating molecular and cellular abnormalities in cell models and in tissue, and evaluating drug responses and the effectiveness of therapeutic intervention strategies. An important way to analyze the development of the human body is to assess molecular markers in embryonic specialized cells, which include the ectoderm, mesoderm, and endoderm. Neuronal development is controlled through the gene networks in the neural crest and neural tube, both components of the ectoderm. The neural crest differentiates into several different tissues including, but not limited to, the peripheral nervous system, enteric nervous system, melanocyte, and the dental pulp. The neural tube eventually converts to the central nervous system. This review provides an overview of the differentiation of the ectoderm to a fully functioning nervous system, focusing on molecular biomarkers that emerge at each stage of the cellular specialization from multipotent stem cells to completely differentiated cells. Particularly, the otic placode is the origin of most of the inner ear cell types such as neurons, sensory hair cells, and supporting cells. During the development, different auditory cell types can be distinguished by the expression of the neurogenin differentiation factor1 (Neuro D1), Brn3a, and transcription factor GATA3. However, the mature auditory neurons express other markers including βIII tubulin, the vesicular glutamate transporter (VGLUT1), the tyrosine receptor kinase B and C (Trk B, C), BDNF, neurotrophin 3 (NT3), Calretinin, etc.
Collapse
Affiliation(s)
- Fatemeh Ghanavatinejad
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Science, Shahrekord, Iran
| | - Zahra Pourteymour Fard Tabrizi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Science, Shahrekord, Iran
| | - Shadi Omidghaemi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Science, Shahrekord, Iran
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Simon Geir Møller
- Department of Biological Sciences, St John's University, New York, NY, USA
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Norway
| | - Mohammad-Saeid Jami
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Science, Shahrekord, Iran
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, CA, 90095, USA
| |
Collapse
|
22
|
Cho KA, Lee HJ, Jeong H, Kim M, Jung SY, Park HS, Ryu KH, Lee SJ, Jeong B, Lee H, Kim HS. Tonsil-derived stem cells as a new source of adult stem cells. World J Stem Cells 2019; 11:506-518. [PMID: 31523370 PMCID: PMC6716082 DOI: 10.4252/wjsc.v11.i8.506] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/25/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
Located near the oropharynx, the tonsils are the primary mucosal immune organ. Tonsil tissue is a promising alternative source for the high-yield isolation of adult stem cells, and recent studies have reported the identification and isolation of tonsil-derived stem cells (T-SCs) from waste surgical tissue following tonsillectomies in relatively young donors (i.e., under 10 years old). As such, T-SCs offer several advantages, including superior proliferation and a shorter doubling time compared to bone marrow-derived mesenchymal stem cells (MSCs). T-SCs also exhibit multi-lineage differentiation, including mesodermal, endodermal (e.g., hepatocytes and parathyroid-like cells), and even ectodermal cells (e.g., Schwann cells). To this end, numbers of researchers have evaluated the practical use of T-SCs as an alternative source of autologous or allogenic MSCs. In this review, we summarize the details of T-SC isolation and identification and provide an overview of their application in cell therapy and regenerative medicine.
Collapse
Affiliation(s)
- Kyung-Ah Cho
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07985, South Korea
| | - Hyun Jung Lee
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, South Korea
| | - Hansaem Jeong
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Miri Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Soo Yeon Jung
- Department of Otorhinolaryngology, College of Medicine, Ewha Womans University, Seoul 07985, South Korea
| | - Hae Sang Park
- Department of Otorhinolaryngology, College of Medicine, Hallym University, Chuncheon 24252, South Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul 07985, South Korea
| | - Seung Jin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Byeongmoon Jeong
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, South Korea
| | - Hyukjin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Han Su Kim
- Department of Otorhinolaryngology, College of Medicine, Ewha Womans University, Seoul 07985, South Korea
| |
Collapse
|
23
|
Oh SY, Choi YM, Kim HY, Park YS, Jung SC, Park JW, Woo SY, Ryu KH, Kim HS, Jo I. Application of Tonsil-Derived Mesenchymal Stem Cells in Tissue Regeneration: Concise Review. Stem Cells 2019; 37:1252-1260. [PMID: 31287931 PMCID: PMC6852396 DOI: 10.1002/stem.3058] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/23/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022]
Abstract
Since the discovery of stem cells and multipotency characteristics of mesenchymal stem cells (MSCs), there has been tremendous development in regenerative medicine. MSCs derived from bone marrow have been widely used in various research applications, yet there are limitations such as invasiveness of obtaining samples, low yield and proliferation rate, and questions regarding their practicality in clinical applications. Some have suggested that MSCs from other sources, specifically those derived from palatine tonsil tissues, that is, tonsil‐derived MSCs (TMSCs), could be considered as a new potential therapeutic tool in regenerative medicine due to their superior proliferation rate and differentiation capabilities with low immunogenicity and ease of obtaining. Several studies have determined that TMSCs have differentiation potential not only into the mesodermal lineage but also into the endodermal as well as ectodermal lineages, expanding their potential usage and placing them as an appealing option to consider for future studies in regenerative medicine. In this review, the differentiation capacities of TMSCs and their therapeutic competencies from past studies are addressed. stem cells2019;37:1252–1260
Collapse
Affiliation(s)
- Se-Young Oh
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Young Min Choi
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Ha Yeong Kim
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Yoon Shin Park
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Sung-Chul Jung
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Joo-Won Park
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - So-Youn Woo
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Han Su Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Inho Jo
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
24
|
Feasibility of autologous plasma gel for tonsil-derived stem cell therapeutics in hypoparathyroidism. Sci Rep 2018; 8:11896. [PMID: 30093637 PMCID: PMC6085304 DOI: 10.1038/s41598-018-30454-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 07/30/2018] [Indexed: 12/30/2022] Open
Abstract
Hypoparathyroidism is a deficiency of the parathyroid hormone (PTH) in the body. We previously reported the possibility of treating it using tonsil-derived mesenchymal stem cells (TMSCs) differentiated into PTH-releasing cells. The purpose of this study was to evaluate the feasibility of using autologous plasma gel as scaffold material in treatment of hypoparathyroidism with TMSC. We obtained plasma by venous sampling of autologous blood and centrifuged and fabricated the plasma gel using a sinusoidal pattern heating machine. After we created the hypoparathyroidism animal model, we administered undifferentiated TMSCs and TMSCs differentiated into parathyroid cells at each rat dorsum by intramuscular injection with and without the plasma gel. In the plasma gel groups, intact PTH was detected from on day 21 after TMSC injection; we did not detect intact PTH in the groups that were only transplanted with TMSCs during the entire experimental period. Serum calcium was higher and phosphorous was lower in the TMSC with plasma gel groups than in the groups with TMSCs alone. We detected PTH and chromogranin A in the TMSC-plasma gel-transplanted areas on immunohistochemistry and immunofluorescence stain. Plasma gel can be considered as a cell-delivery scaffold for treating hypoparathyroidism with tonsil-derived mesenchymal stem cells.
Collapse
|
25
|
Hicks G, George R, Sywak M. Short and long-term impact of parathyroid autotransplantation on parathyroid function after total thyroidectomy. Gland Surg 2017; 6:S75-S85. [PMID: 29322025 DOI: 10.21037/gs.2017.09.15] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The most common complication of total thyroidectomy is parathyroid insufficiency. Acute, transient, post-operative hypoparathyroidism increases length of hospitalization, morbidity and cost associated with total thyroidectomy. While permanent hypoparathyroidism poses a significant medical burden with lifetime medication, regular follow up and considerable disease burden related to chronic renal failure and other sequelae. Parathyroid autotransplantation has been demonstrated to result in biochemically functional grafts, leading to the procedures' common use during total thyroidectomy. The clearest indications for parathyroid auto transplantation are inadvertently removed or devascularized parathyroid glands. Some centers utilize routine autotransplantation to reduce the risk of permanent hypoparathyroidism. Novel fluorescence techniques to aid in parathyroid detection during thyroid surgery are under evaluation. This review aims to define the role and impact of parathyroid autotransplantation undertaken during total thyroidectomy.
Collapse
Affiliation(s)
- Gabrielle Hicks
- Endocrine Surgical Unit, University of Sydney, St Leonards, New South Wales, Australia
| | - Robert George
- Endocrine Surgical Unit, University of Sydney, St Leonards, New South Wales, Australia
| | - Mark Sywak
- Endocrine Surgical Unit, University of Sydney, St Leonards, New South Wales, Australia
| |
Collapse
|
26
|
Jo YI, Kim G, Jin YM, Park YJ, Kim HS, Park YS. Intracellular Remodeling and Accumulation of Aberrant Lysosomes in Differentiation of Tonsil-Derived Mesenchymal Stem Cells into Parathyroid-Like Cells. Tissue Eng Regen Med 2017; 14:411-420. [PMID: 30603497 PMCID: PMC6171608 DOI: 10.1007/s13770-017-0042-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 09/19/2016] [Accepted: 09/26/2016] [Indexed: 01/08/2023] Open
Abstract
Differentiation of mesenchymal stem cells (MSC) into a variety of cell lineages such as adipocytes, osteocytes, and chondrocytes is often accompanied up-regulation of autophagy. In our study, we demonstrated that the expression of autophagy-associated proteins (p-Beclin 1, LC3A, LC3B, p-AMPK, p-mTOR and ATG3, ATG7, and ATG12-5) over a period of time was hardly distinguishable from control tonsil-derived MSC (TMSC). Despite the unnoticeable difference in autophagy activation between differentiated TMSC (dTMSC) and the control (cTMSC), we reported significant changes in intracellular compositions in differentiated TMSC into functional parathyroid-like cells secreting parathyroid hormone (PTH). By using transmission electron microscopy (TEM), we observed accumulation of multivesicular bodies (MVB) comprising small, degraded compartments densely accumulated as dark granular or amorphous clumps, multilamellar bodies and lipid droplets in dTMSC. However, no such structures were found in cTMSC. These results suggest that differentiation of TMSC into parathyroid-like cells producing PTH hormone is hardly dependent on autophagy activation in the beginning of our conditions. Furthermore, our results of intracellular remodeling and accumulated endo-lysosomal storage bodies in the later stages of TMSC differentiation present a possible role of the structures in PTH secretion.
Collapse
Affiliation(s)
- Young-Il Jo
- Department of Dental Regenerative Biotechnology, Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
| | - Gyungah Kim
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul, 07985 Republic of Korea
- Ewha Tonsil-Derived Mesenchymal Stem Cells Research Center (ETSRC), School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul, 07985 Republic of Korea
| | - Yoon Mi Jin
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul, 07985 Republic of Korea
- Ewha Tonsil-Derived Mesenchymal Stem Cells Research Center (ETSRC), School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul, 07985 Republic of Korea
| | - Yoon Jeong Park
- Department of Dental Regenerative Biotechnology, Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
| | - Han Su Kim
- Department of Otorhinolaryngology – Head and Neck Surgery, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul, 07985 Republic of Korea
| | - Yoon Shin Park
- Major in Microbiology, School of Biological Sciences, College of Natural Sciences, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, Chungbuk 28644 Republic of Korea
| |
Collapse
|
27
|
Park YS, Lee Y, Jin YM, Kim G, Jung SC, Park YJ, Park KD, Jo I. Sustained release of parathyroid hormone via
in situ
cross‐linking gelatin hydrogels improves the therapeutic potential of tonsil‐derived mesenchymal stem cells for hypoparathyroidism. J Tissue Eng Regen Med 2017; 12:e1747-e1756. [DOI: 10.1002/term.2430] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/25/2017] [Accepted: 02/23/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Yoon Shin Park
- Department of Molecular Medicine, School of MedicineEwha Womans University Seoul Republic of Korea
- Ewha Tonsil‐derived Mesenchymal Stem Cells Research Center (ETSRC), School of MedicineEwha Womans University Seoul Republic of Korea
- School of Biological Sciences, College of Natural SciencesChungbuk National University Cheongju Republic of Korea
| | - Yunki Lee
- Department of Molecular Science and TechnologyAjou University Suwon Republic of Korea
| | - Yoon Mi Jin
- Department of Molecular Medicine, School of MedicineEwha Womans University Seoul Republic of Korea
- Ewha Tonsil‐derived Mesenchymal Stem Cells Research Center (ETSRC), School of MedicineEwha Womans University Seoul Republic of Korea
| | - Gyungah Kim
- Department of Molecular Medicine, School of MedicineEwha Womans University Seoul Republic of Korea
- Ewha Tonsil‐derived Mesenchymal Stem Cells Research Center (ETSRC), School of MedicineEwha Womans University Seoul Republic of Korea
| | - Sung Chul Jung
- Ewha Tonsil‐derived Mesenchymal Stem Cells Research Center (ETSRC), School of MedicineEwha Womans University Seoul Republic of Korea
- Department of Biochemistry, School of MedicineEwha Womans University Seoul Republic of Korea
| | - Yoon Jeong Park
- Department of Dental Regenerative BiotechnologyDental Research Institute, School of Dentistry, Seoul National University Seoul Republic of Korea
| | - Ki Dong Park
- Department of Molecular Science and TechnologyAjou University Suwon Republic of Korea
| | - Inho Jo
- Department of Molecular Medicine, School of MedicineEwha Womans University Seoul Republic of Korea
- Ewha Tonsil‐derived Mesenchymal Stem Cells Research Center (ETSRC), School of MedicineEwha Womans University Seoul Republic of Korea
| |
Collapse
|
28
|
Toledo PC, Rossi RL, Caviedes P. Microencapsulation of Parathyroid Cells for the Treatment of Hypoparathyroidism. Methods Mol Biol 2017; 1479:357-363. [PMID: 27738949 DOI: 10.1007/978-1-4939-6364-5_27] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cell encapsulation is an alternative to avoid rejection of grafted tissue, thus bringing an interesting alternative in cell therapy. It is particularly relevant in ailments where only the implant of small quantities of tissues is warranted. In such circumstances, the use of immunosuppressive therapy in patients implanted with tissues from donors is debatable, yet unavoidable at present in order to prevent rejection and/or sensitization of the host to the tissue, in turn jeopardizing the success of successive implants. Hence, a new line of thought, which aims to provide an immunoprivileged site for the grafted tissue, while at the same time insure its nutrition, as well as its survival and continued function, appears as a most attractive possibility. To achieve these goals, cells or tissues harvested for transplant could be encapsulated in biologically compatible matrices. Among the matrices currently in existence, sodium alginate is the most widely used polymer for tissue encapsulation.In the present chapter, we present a technique used to encapsulate parathyroid tissue, for use as cell transplant therapy in patients with secondary hypoparathyroidism. With this procedure, implanted tissue survives and remains functional for up to 18 months.
Collapse
Affiliation(s)
- Patricio Cabané Toledo
- Head and Neck Surgeon, University of Chile Clinical Hospital, Independencia, Santiago de Chile, Chile.
| | - Ricardo L Rossi
- Department of Surgery of Clínica Alemana de Santiago, Independencia, Santiago de Chile, Chile
| | - Pablo Caviedes
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Independencia, Santiago de Chile, Chile
| |
Collapse
|
29
|
Lee F, Lee JJ, Jan WC, Wu CJ, Chen HH, Cheng SP. Molecular pathways associated with transcriptional alterations in hyperparathyroidism. Oncol Lett 2016; 12:621-626. [PMID: 27347190 DOI: 10.3892/ol.2016.4623] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 05/12/2016] [Indexed: 12/11/2022] Open
Abstract
Hyperparathyroidism is characterized by the oversecretion of parathyroid hormone biochemically and increased cell proliferation histologically. Primary and secondary hyperparathyroidism exhibit distinct pathophysiology but share certain common microscopic features. The present study performed the first genome-wide expression analysis directly comparing the expression profile of primary and secondary hyperparathyroidism. Microarray gene expression analyses were performed in parathyroid tissues from 2 primary hyperparathyroidism patients and 3 secondary hyperparathyroidism patients. Unsupervised hierarchical clustering analysis identified two natural subgroups containing different types of hyperparathyroidism. Combined with additional data extracted from a publicly available database, a meta-signature was constructed to represent an intersection of two sets of differential expression profile. Multiple pathways were identified that are aberrantly regulated in hyperparathyroidism. In primary hyperparathyroidism, dysregulated pathways included cell adhesion molecules, peroxisome proliferator-activated receptor signaling pathway, and neuroactive ligand-receptor interaction. Pathways implicated in secondary hyperparathyroidism included tryptophan metabolism, tight junctions, renin-angiotensin system, steroid hormone biosynthesis, and O-glycan biosynthesis. The present study demonstrates that different pathophysiology is associated with differential gene profiling in hyperparathyroidism. Several pathways are involved in parathyroid dysregulation and may be future targets for therapeutic intervention.
Collapse
Affiliation(s)
- Fang Lee
- Department of Surgery, MacKay Memorial Hospital and MacKay Medical College, Taipei 10449, Taiwan, R.O.C
| | - Jie-Jen Lee
- Department of Surgery, MacKay Memorial Hospital and MacKay Medical College, Taipei 10449, Taiwan, R.O.C.; Department of Nursing, MacKay Junior College of Medicine, Nursing and Management, Tapei 11260, Taiwan, R.O.C.; Department of Pharmacology and Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| | - Woan-Ching Jan
- Department of Nursing, MacKay Junior College of Medicine, Nursing and Management, Tapei 11260, Taiwan, R.O.C
| | - Chih-Jen Wu
- Department of Pharmacology and Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan, R.O.C.; Department of Nephrology, MacKay Memorial Hospital and Mackay Medical College, Taipei 10449, Taiwan, R.O.C
| | - Han-Hsiang Chen
- Department of Pharmacology and Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan, R.O.C.; Department of Nephrology, MacKay Memorial Hospital and Mackay Medical College, Taipei 10449, Taiwan, R.O.C
| | - Shih-Ping Cheng
- Department of Surgery, MacKay Memorial Hospital and MacKay Medical College, Taipei 10449, Taiwan, R.O.C.; Department of Pharmacology and Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| |
Collapse
|
30
|
Park YS, Hwang JY, Jun Y, Jin YM, Kim G, Kim HY, Kim HS, Lee SH, Jo I. Scaffold-free parathyroid tissue engineering using tonsil-derived mesenchymal stem cells. Acta Biomater 2016; 35:215-27. [PMID: 26945633 DOI: 10.1016/j.actbio.2016.03.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/27/2016] [Accepted: 03/01/2016] [Indexed: 01/01/2023]
Abstract
To restore damaged parathyroid function, parathyroid tissue engineering is the best option. Previously, we reported that differentiated tonsil-derived mesenchymal stem cells (dTMSC) restore in vivo parathyroid function, but only if they are embedded in a scaffold. Because of the limited biocompatibility of Matrigel, however, here we developed a more clinically applicable, scaffold-free parathyroid regeneration system. Scaffold-free dTMSC spheroids were engineered in concave microwell plates made of polydimethylsiloxane in control culture medium for the first 7days and differentiation medium (containing activin A and sonic hedgehog) for next 7days. The size of dTMSC spheroids showed a gradual and significant decrease up to day 5, whereafter it decreased much less. Cells in dTMSC spheroids were highly viable (>80%). They expressed high levels of intact parathyroid hormone (iPTH), the parathyroid secretory protein 1, and cell adhesion molecule, N-cadherin. Furthermore, dTMSC spheroids-implanted parathyroidectomized (PTX) rats revealed higher survival rates (50%) over a 3-month period with physiological levels of both serum iPTH (57.7-128.2pg/mL) and ionized calcium (0.70-1.15mmol/L), compared with PTX rats treated with either vehicle or undifferentiated TMSC spheroids. This is the first report of a scaffold-free, human stem cell-based parathyroid tissue engineering and represents a more clinically feasible strategy for hypoparathyroidism treatment than those requiring scaffolds. STATEMENT OF SIGNIFICANCE Herein, we have for the first time developed a scaffold-free parathyroid tissue spheroids using differentiated tonsil-derived mesenchymal stem cells (dTMSC) to restore in vivo parathyroid cell functions. This new strategy is effective, even for long periods (3months), and is thus likely to be more feasible in clinic for hypoparathyroidism treatment. Development of TMSC spheroids may also provide a convenient and efficient scaffold-free platform for researchers investigating conditions involving abnormal calcium homeostasis, such as osteoporosis.
Collapse
|
31
|
Park YS, Kim HS, Jin YM, Yu Y, Kim HY, Park HS, Jung SC, Han KH, Park YJ, Ryu KH, Jo I. Differentiated tonsil-derived mesenchymal stem cells embedded in Matrigel restore parathyroid cell functions in rats with parathyroidectomy. Biomaterials 2015; 65:140-52. [DOI: 10.1016/j.biomaterials.2015.06.044] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 06/24/2015] [Accepted: 06/26/2015] [Indexed: 10/23/2022]
|
32
|
Ikonomou L, Kotton DN. Derivation of Endodermal Progenitors From Pluripotent Stem Cells. J Cell Physiol 2015; 230:246-58. [PMID: 25160562 PMCID: PMC4344429 DOI: 10.1002/jcp.24771] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 08/22/2014] [Indexed: 01/18/2023]
Abstract
Stem and progenitor cells play important roles in organogenesis during development and in tissue homeostasis and response to injury postnatally. As the regenerative capacity of many human tissues is limited, cell replacement therapies hold great promise for human disease management. Pluripotent stem cells such as embryonic stem (ES) cells and induced pluripotent stem (iPS) cells are prime candidates for the derivation of unlimited quantities of clinically relevant cell types through development of directed differentiation protocols, that is, the recapitulation of developmental milestones in in vitro cell culture. Tissue-specific progenitors, including progenitors of endodermal origin, are important intermediates in such protocols since they give rise to all mature parenchymal cells. In this review, we focus on the in vivo biology of embryonic endodermal progenitors in terms of key transcription factors and signaling pathways. We critically review the emerging literature aiming to apply this basic knowledge to achieve the efficient and reproducible in vitro derivation of endodermal progenitors such as pancreas, liver and lung precursor cells.
Collapse
Affiliation(s)
- Laertis Ikonomou
- Center for Regenerative Medicine, Boston University and Boston
Medical Center, Boston, MA, USA
- Boston University Pulmonary Center, Boston University School of
Medicine, Boston, MA, USA
| | - Darrell N. Kotton
- Center for Regenerative Medicine, Boston University and Boston
Medical Center, Boston, MA, USA
- Boston University Pulmonary Center, Boston University School of
Medicine, Boston, MA, USA
| |
Collapse
|
33
|
Park YS, Hwang S, Jin YM, Yu Y, Jung SA, Jung SC, Ryu KH, Kim HS, Jo I. CCN1 secreted by tonsil-derived mesenchymal stem cells promotes endothelial cell angiogenesis via integrin αv β3 and AMPK. J Cell Physiol 2015; 230:140-9. [PMID: 24909560 DOI: 10.1002/jcp.24690] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 05/21/2014] [Indexed: 11/12/2022]
Abstract
CCN1 is highly expressed in cancer cells and has been identified in the secretome of bone marrow-derived mesenchymal stem cells (BM-MSC). Although secreted CCN1 is known to promote angiogenesis, its underlying mechanism remains unclear. Here, we examined whether our recently-established tonsil-derived MSC (T-MSC) secrete CCN1 and, if any, how CCN1 promotes the angiogenesis of human umbilical vein endothelial cells (HUVEC). Compared with untreated control T-MSC, a higher level of CCN1 was secreted by T-MSC treated with activin A and sonic hedgehog, drugs known to induce endodermal differentiation. Expectedly, conditioned medium collected from differentiated T-MSC (DCM) significantly increased HUVEC migration and tube formation compared with that from control T-MSC (CCM), and these stimulatory effects were reversed by neutralization with anti-CCN1 antibody. Treatment with recombinant human CCN1 (rh-CCN1) alone also mimicked the stimulatory effects of DCM. Furthermore, treatment with either DCM or rh-CCN1 increased the phosphorylation of AMP kinase (AMPK), and ectopic expression of siRNA of the AMPK gene inhibited all observed effects of both DCM and rh-CCN1. However, no alteration of intracellular ATP levels or phosphorylation of LKB1, a well-known upstream factor of AMPK activation, was observed under our conditions. Finally, the neutralization of integrin α(v) β(3) with anti-integrin α(v) β(3) antibody almost completely reversed the effects of CCN1 on AMPK phosphorylation, and EC migration and tube formation. Taken together, we demonstrated that T-MSC increase the secretion of CCN1 in response to endodermal differentiation and that integrin α(v) β(3) and AMPK mediate CCN1-induced EC migration and tube formation independent of intracellular ATP levels alteration.
Collapse
Affiliation(s)
- Yoon Shin Park
- Department of Molecular Medicine, School of Medicine, and Global Top 5 Research Program, Ewha Womans University, Mok-6-dong, Yangcheon-gu, Seoul 158-710, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Xenotransplantation of human cultured parathyroid progenitor cells into mouse peritoneum does not induce rejection reaction. Cent Eur J Immunol 2014; 39:279-84. [PMID: 26155136 PMCID: PMC4440013 DOI: 10.5114/ceji.2014.45937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/14/2014] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Parathyroid progenitor cells devoid of immunogenic antigens were used for human allotransplantation. Although there were many potential reasons for the expiry of transplant activity in humans, we decided to exclude a subclinical form of rejection reaction, and test the rejection reaction in an animal model. MATERIAL AND METHODS Experiments were carried out on 40 conventional male mice in their third month of life. The animals were housed in groups of 10 per cage in 4 cages with fitted water dispensers and fed a conventional diet based on standard pellet food. They were divided into four groups of 10 animals each, three experimental groups and one control group. Identified progenitor cells were stored in a cell bank. After testing the phenotype, viability, and absence of immunogenic properties, the cells were transplanted into mouse peritoneum cavity. RESULTS Animals were observed for 9 weeks. At 9 weeks of observation, the mean serum PTH concentration in the experimental groups was 2.0-2.5 pg/ml, while in the control group it did not exceed 1.5 pg/ml. The immunohistochemical assays demonstrated that millions of viable cells with a phenotype identical to the endocrine cells had survived in the peritoneum. Histologic specimens from different internal organs stained for PTH revealed positive cells labelled with anti-PTH Ab in the intestinal lamina, brain, liver, and spleen. CONCLUSIONS In the present paper we have demonstrated that xenotransplantation may be used as a model for an explanation of the immunogenic properties of cells generated from postnatal organs for regenerative therapy.
Collapse
|
35
|
Ryu KH, Cho KA, Park HS, Kim JY, Woo SY, Jo I, Choi YH, Park YM, Jung SC, Chung SM, Choi BO, Kim HS. Tonsil-derived mesenchymal stromal cells: evaluation of biologic, immunologic and genetic factors for successful banking. Cytotherapy 2012; 14:1193-202. [DOI: 10.3109/14653249.2012.706708] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
Agarwal SK, Jothi R. Genome-wide characterization of menin-dependent H3K4me3 reveals a specific role for menin in the regulation of genes implicated in MEN1-like tumors. PLoS One 2012; 7:e37952. [PMID: 22666422 PMCID: PMC3364203 DOI: 10.1371/journal.pone.0037952] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 04/30/2012] [Indexed: 01/07/2023] Open
Abstract
Inactivating mutations in the MEN1 gene predisposing to the multiple endocrine neoplasia type 1 (MEN1) syndrome can also cause sporadic pancreatic endocrine tumors. MEN1 encodes menin, a subunit of MLL1/MLL2-containing histone methyltransferase complexes that trimethylate histone H3 at lysine 4 (H3K4me3). The importance of menin-dependent H3K4me3 in normal and transformed pancreatic endocrine cells is unclear. To study the role of menin-dependent H3K4me3, we performed in vitro differentiation of wild-type as well as menin-null mouse embryonic stem cells (mESCs) into pancreatic islet-like endocrine cells (PILECs). Gene expression analysis and genome-wide H3K4me3 ChIP-Seq profiling in wild-type and menin-null mESCs and PILECs revealed menin-dependent H3K4me3 at the imprinted Dlk1-Meg3 locus in mESCs, and all four Hox loci in differentiated PILECs. Specific and significant loss of H3K4me3 and gene expression was observed for genes within the imprinted Dlk1-Meg3 locus in menin-null mESCs and the Hox loci in menin-null PILECs. Given that the reduced expression of genes within the DLK1-MEG3 locus and the HOX loci is associated with MEN1-like sporadic tumors, our data suggests a possible role for menin-dependent H3K4me3 at these genes in the initiation and progression of sporadic pancreatic endocrine tumors. Furthermore, our investigation also demonstrates that menin-null mESCs can be differentiated in vitro into islet-like endocrine cells, underscoring the utility of menin-null mESC-derived specialized cell types for genome-wide high-throughput studies.
Collapse
Affiliation(s)
- Sunita K. Agarwal
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (SKA); (RJ)
| | - Raja Jothi
- Systems Biology Section, Biostatistics Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
- * E-mail: (SKA); (RJ)
| |
Collapse
|
37
|
Archambeault DR, Tomaszewski J, Childs AJ, Anderson RA, Yao HHC. Testicular somatic cells, not gonocytes, are the major source of functional activin A during testis morphogenesis. Endocrinology 2011; 152:4358-67. [PMID: 21952240 PMCID: PMC3199008 DOI: 10.1210/en.2011-1288] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Proper development of the seminiferous tubules (or testis cords in embryos) is critical for male fertility. Sertoli cells, somatic components of the seminiferous tubules, serve as nurse cells to the male germline, and thus their numbers decide the quantity of sperm output in adulthood. We previously identified activin A, the protein product of the activin βA (Inhba) gene, as a key regulator of murine Sertoli cell proliferation and testis cord expansion during embryogenesis. Although our genetic studies implicated fetal Leydig cells as the primary producers of testicular activin A, gonocytes are another potential source. To investigate the relative contribution of gonocyte-derived activin A to testis morphogenesis, we compared testis development in the Inhba global knockout mouse, which lacks activin A production in all cells (including the gonocytes), and a steroidogenic factor 1 (Sf1)-specific conditional knockout model in which activin A expression in testicular somatic cells is disrupted but gonocyte expression of activin A remains intact. Surprisingly, testis development was comparable in these two models of activin A insufficiency, with similar reductions in Sertoli cell proliferation and minor differences in testis histology. Thus, our findings suggest activin A from male gonocytes is insufficient to promote Sertoli cell proliferation and testis cord expansion in the absence of somatic cell-derived activin A. Evaluation of adult male mice with fetal disruption of activin A revealed reduced testis size, lowered sperm production, altered testicular histology, and elevated plasma FSH levels, defects reminiscent of human cases of androgen-sufficient idiopathic oligozoospermia.
Collapse
Affiliation(s)
- Denise R Archambeault
- Department of Comparative Biosciences, University of Illinois, Urbana, IL 61802, USA
| | | | | | | | | |
Collapse
|
38
|
Woods Ignatoski KM, Bingham EL, Frome LK, Doherty GM. DirectedTrans-Differentiation of Thymus Cells into Parathyroid-Like Cells Without Genetic Manipulation. Tissue Eng Part C Methods 2011; 17:1051-9. [PMID: 21797755 DOI: 10.1089/ten.tec.2011.0170] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Kathleen M. Woods Ignatoski
- Division of Endocrine Surgery, Department of Surgery, University of Michigan Health System, Ann Arbor, Michigan
| | - Evangeline L. Bingham
- Division of Endocrine Surgery, Department of Surgery, University of Michigan Health System, Ann Arbor, Michigan
| | - Lauren K. Frome
- Division of Endocrine Surgery, Department of Surgery, University of Michigan Health System, Ann Arbor, Michigan
| | - Gerard M. Doherty
- Division of Endocrine Surgery, Department of Surgery, University of Michigan Health System, Ann Arbor, Michigan
| |
Collapse
|
39
|
Green MD, Chen A, Nostro MC, d'Souza SL, Schaniel C, Lemischka IR, Gouon-Evans V, Keller G, Snoeck HW. Generation of anterior foregut endoderm from human embryonic and induced pluripotent stem cells. Nat Biotechnol 2011; 29:267-72. [PMID: 21358635 DOI: 10.1038/nbt.1788] [Citation(s) in RCA: 271] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 01/25/2011] [Indexed: 01/08/2023]
Abstract
Directed differentiation of human embryonic stem (hES) cells and human induced pluripotent stem (hiPS) cells captures in vivo developmental pathways for specifying lineages in vitro, thus avoiding perturbation of the genome with exogenous genetic material. Thus far, derivation of endodermal lineages has focused predominantly on hepatocytes, pancreatic endocrine cells and intestinal cells. The ability to differentiate pluripotent cells into anterior foregut endoderm (AFE) derivatives would expand their utility for cell therapy and basic research to tissues important for immune function, such as the thymus; for metabolism, such as thyroid and parathyroid; and for respiratory function, such as trachea and lung. We find that dual inhibition of transforming growth factor (TGF)-β and bone morphogenic protein (BMP) signaling after specification of definitive endoderm from pluripotent cells results in a highly enriched AFE population that is competent to be patterned along dorsoventral and anteroposterior axes. These findings provide an approach for the generation of AFE derivatives.
Collapse
Affiliation(s)
- Michael D Green
- Department of Gene and Cell Medicine and Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Woods Ignatoski KM, Bingham EL, Frome LK, Doherty GM. Differentiation of precursors into parathyroid-like cells for treatment of hypoparathyroidism. Surgery 2010; 148:1186-9; discussion 1189-90. [DOI: 10.1016/j.surg.2010.09.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 09/16/2010] [Indexed: 10/18/2022]
|
41
|
Abstract
Activin was discovered in the 1980s as a gonadal protein that stimulated FSH release from pituitary gonadotropes and was thought of as a reproductive hormone. In the ensuing decades, many additional activities of activin were described and it was found to be produced in a wide variety of cell types at nearly all stages of development. Its signaling and actions are regulated intracellularly and by extracellular antagonists. Over the past 5 years, a number of important advances have been made that clarify our understanding of the structural basis for signaling and regulation, as well as the biological roles of activin in stem cells, embryonic development and in adults. These include the crystallization of activin in complex with the activin type II receptor ActRIIB, or with the binding proteins follistatin and follistatin-like 3, as well as identification of activin's roles in gonadal sex development, follicle development, luteolysis, beta-cell proliferation and function in the islet, stem cell pluripotency and differentiation into different cell types and in immune cells. These advances are reviewed to provide perspective for future studies.
Collapse
Affiliation(s)
- Yin Xia
- Program in Membrane Biology and Division of Nephrology, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | |
Collapse
|