1
|
Mohamed Rasheed ZB, Nordin F, Wan Kamarul Zaman WS, Tan YF, Abd Aziz NH. Autologous Human Mesenchymal Stem Cell-Based Therapy in Infertility: New Strategies and Future Perspectives. BIOLOGY 2023; 12:108. [PMID: 36671799 PMCID: PMC9855776 DOI: 10.3390/biology12010108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/12/2023]
Abstract
Infertility could be associated with a few factors including problems with physical and mental health, hormonal imbalances, lifestyles, and genetic factors. Given that there is a concern about the rise of infertility globally, increased focus has been given to its treatment for the last several decades. Traditional assisted reproductive technology (ART) has been the prime option for many years in solving various cases of infertility; however, it contains significant risks and does not solve the fundamental problem of infertility such as genetic disorders. Attention toward the utilization of MSCs has been widely regarded as a promising option in the development of stem-cell-based infertility treatments. This narrative review briefly presents the challenges in the current ART treatment of infertility and the various potential applications of autologous MSCs in the treatment of these reproductive diseases.
Collapse
Affiliation(s)
- Zahirrah Begam Mohamed Rasheed
- UKM Medical Molecular Biology Institute (UMBI), Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Fazlina Nordin
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | | | - Yuen-Fen Tan
- PPUKM-MAKNA Cancer Center, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, WPKL, Kuala Lumpur 56000, Malaysia
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Bandar Sungai Long, Kajang 43000, Malaysia
| | - Nor Haslinda Abd Aziz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- Research Laboratory of UKM Specialist Children’s Hospital, UKM Specialist Children’s Hospital, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
2
|
Bhartiya D, Jha N, Tripathi A, Tripathi A. Very small embryonic-like stem cells have the potential to win the three-front war on tissue damage, cancer, and aging. Front Cell Dev Biol 2023; 10:1061022. [PMID: 36684436 PMCID: PMC9846763 DOI: 10.3389/fcell.2022.1061022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/14/2022] [Indexed: 01/05/2023] Open
Abstract
The concept of dedifferentiation and reprogramming of mature somatic cells holds much promise for the three-front "war" against tissue damage, cancer, and aging. It was hoped that reprogramming human somatic cells into the induced pluripotent state, along with the use of embryonic stem cells, would transform regenerative medicine. However, despite global efforts, clinical applications remain a distant dream, due to associated factors such as genomic instability, tumorigenicity, immunogenicity, and heterogeneity. Meanwhile, the expression of embryonic (pluripotent) markers in multiple cancers has baffled the scientific community, and it has been suggested that somatic cells dedifferentiate and "reprogram" into the pluripotent state in vivo to initiate cancer. It has also been suggested that aging can be reversed by partial reprogramming in vivo. However, better methods are needed; using vectors or Yamanaka factors in vivo, for example, is dangerous, and many potential anti-aging therapies carry the same risks as those using induced pluripotent cells, as described above. The present perspective examines the potential of endogenous, pluripotent very small embryonic-like stem cells (VSELs). These cells are naturally present in multiple tissues; they routinely replace diseased tissue and ensure regeneration to maintain life-long homeostasis, and they have the ability to differentiate into adult counterparts. Recent evidence suggests that cancers initiate due to the selective expansion of epigenetically altered VSELs and their blocked differentiation. Furthermore, VSEL numbers have been directly linked to lifespan in studies of long- and short-lived transgenic mice, and VSEL dysfunction has been found in the ovaries of aged mice. To conclude, a greater interest in VSELs, with their potential to address all three fronts of this war, could be the "light at the end of the tunnel."
Collapse
|
3
|
Altered Biology of Testicular VSELs and SSCs by Neonatal Endocrine Disruption Results in Defective Spermatogenesis, Reduced Fertility and Tumor Initiation in Adult Mice. Stem Cell Rev Rep 2021; 16:893-908. [PMID: 32592162 DOI: 10.1007/s12015-020-09996-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Reproductive health of men has declined in recent past with reduced sperm count and increased incidence of infertility and testicular cancers mainly attributed to endocrine disruption in early life. Present study aims to evaluate whether testicular stem cells including very small embryonic-like stem cells (VSELs) and spermatogonial stem cells (SSCs) get affected by endocrine disruption and result in pathologies in adult life. Effect of treatment on mice pups with estradiol (20 μg on days 5-7) and diethylstilbestrol (DES, 2 μg on days 1-5) was studied on VSELs, SSCs and spermatogonial cells in adult life. Treatment affected spermatogenesis, tubules in Stage VIII & sperm count were reduced along with reduction of meiotic (4n) cells and markers (Prohibitin, Scp3, Protamine). Enumeration of VSELs by flow cytometry (2-6 μm, 7AAD-, LIN-CD45-SCA-1+) and qRT-PCR using specific transcripts for VSELs (Oct-4a, Sox-2, Nanog, Stella, Fragilis), SSCs (tOct-4, Gfra-1, Gpr-125) and early germ cells (Mvh, Dazl) showed several-fold increase but transition from c-Kit negative to c-Kit positive spermatogonial cells was blocked on D100 after treatment. Transcripts specific for apoptosis (Bcl2, Bax) remained unaffected but tumor suppressor (p53) and epigenetic regulator (NP95) transcripts showed marked disruption. 9 of 10 mice exposed to DES showed tumor-like changes. To conclude, endocrine disruption resulted in a tilt towards excessive self-renewal of VSELs (leading to testicular cancer after DES treatment) and blocked differentiation (reduced numbers of c-Kit positive cells, meiosis, sperm count and fertility). Understanding the underlying basis for infertility and cancer initiation from endogenous stem cells through murine modelling will hopefully improve human therapies in future.
Collapse
|
4
|
Ezawa M, Kouno F, Kubo H, Sakuma T, Yamamoto T, Kinoshita T. Pou5f3.3 is involved in establishment and maintenance of hematopoietic cells during Xenopus development. Tissue Cell 2021; 72:101531. [PMID: 33798831 DOI: 10.1016/j.tice.2021.101531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/05/2021] [Accepted: 03/15/2021] [Indexed: 11/30/2022]
Abstract
Three POU family class V gene homologues are expressed in the development of Xenopus. In contrast to the expression of Pou5f3.1 and Pou5f3.2 in organogenesis, Pou5f3.3 is expressed during oogenesis in ovary. We investigated the expression and function of Pou5f3.3 in organogenesis of Xenopus laevis. RT-PCR and immunohistochemical analysis indicated that Pou5f3.3 was expressed in a small number of adult liver cells and blood cells. Immunocytochemical investigation proved that Bmi1, a marker for hematopoietic progenitor cells, was co-expressed in Pou5f3.3-expressing small spherical cells in the peripheral blood. In anemic induction by intraperitoneal injection of phenyl hydrazine, the number of Pou5f3.3-expressing cells significantly increased within 3 days after phenyl hydrazine injection. In CRISPR/Cas mutagenesis of Pou5f3.3, Bmi1-positive hematopoietic progenitor cell count decreased in the hematopoietic dorsal-lateral plate (DLP) region, resulting in a considerable reduction in peripheral blood cells. CRISPR/Cas-induced hematopoietic deficiency was completely rescued by Pou5f3.3 supplementation, but not by Pou5f3.1 or Pou5f3.2. Transplantation experiments using the H2B-GFP transgenic line demonstrated that DLP-derived Pou5f3.3-positive and Bmi1-positive cells were translocated into the liver and bone through the bloodstream. These results suggest that Pou5f3.3 plays an essential role in the establishment and maintenance of hematopoietic progenitor cells during Xenopus development.
Collapse
Affiliation(s)
- Minami Ezawa
- Department of Life Science, Faculty of Science, Rikkyo University, 3-34-1 Nishi-Ikebukuro, Toshima-ku, Tokyo, 171-8501, Japan
| | - Fumika Kouno
- Department of Life Science, Faculty of Science, Rikkyo University, 3-34-1 Nishi-Ikebukuro, Toshima-ku, Tokyo, 171-8501, Japan
| | - Hideo Kubo
- Department of Membrane Biochemistry, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Tetsushi Sakuma
- Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Takashi Yamamoto
- Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Tsutomu Kinoshita
- Department of Life Science, Faculty of Science, Rikkyo University, 3-34-1 Nishi-Ikebukuro, Toshima-ku, Tokyo, 171-8501, Japan.
| |
Collapse
|
5
|
Ventura-Carmenate Y, Alkaabi FM, Castillo-Aleman YM, Villegas-Valverde CA, Ahmed YM, Sanna P, Almarzooqi AA, Abdelrazik A, Torres-Zambrano GM, Wade-Mateo M, Quesada-Saliba D, Abdel Hadi L, Bencomo-Hernandez AA, Rivero-Jimenez RA. Safety and efficacy of autologous non-hematopoietic enriched stem cell nebulization in COVID-19 patients: a randomized clinical trial, Abu Dhabi 2020. TRANSLATIONAL MEDICINE COMMUNICATIONS 2021; 6:25. [PMID: 34746417 PMCID: PMC8563822 DOI: 10.1186/s41231-021-00101-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/05/2021] [Indexed: 05/04/2023]
Abstract
BACKGROUND The novel SARS-CoV-2 has caused the coronavirus disease 2019 (COVID-19) pandemic. Currently, with insufficient worldwide vaccination rates, identifying treatment solutions to reduce the impact of the virus is urgently needed. METHOD An adaptive, multicentric, open-label, and randomized controlled phase I/II clinical trial entitled the "SENTAD-COVID Study" was conducted by the Abu Dhabi Stem Cells Center under exceptional conditional approval by the Emirates Institutional Review Board (IRB) for COVID-19 Research Committee from April 4th to July 31st, 2020, using an autologous peripheral blood non-hematopoietic enriched stem cell cocktail (PB-NHESC-C) administered by compressor (jet) nebulization as a complement to standard care therapy. The primary endpoints include safety and efficacy assessments, adverse events, the mortality rate within 28 days, and the time to clinical improvement as measured by a 2-point reduction on a seven-category ordinal scale or discharge from the hospital whichever occurred first. RESULTS The study included a total of 139 randomized COVID-19 patients, with 69 in the experimental group and 70 in the control group (standard care). Overall survival was 94.20% for the cocktail-treated group vs. 90.27% for the control group. Adverse events were reported in 50 (72.46%) patients receiving PB-NHESC-C and 51 (72.85%) in the control group (p = 0.9590), with signs and symptoms commonly found in COVID-19. After the first 9 days of the intervention, 67.3% of cocktail-treated patients recovered and were released from hospitals compared to 53.1% (RR = 0.84; 95% CI, 0.56-1.28) in the control group. Improvement, i.e., at least a 2-point reduction in the severity scale, was more frequently observed in cocktail-treated patients (42.0%) than in controls (17.0%) (RR = 0.69; 95% CI, 0.56-0.88). CONCLUSIONS Cocktail treatment improved clinical outcomes without increasing adverse events. Thus, the nebulization of PB-NHESC-C was safe and effective for treatment in most of these patients. TRIAL REGISTRATION ClinicalTrials.gov. NCT04473170. It was retrospectively registered on July 16th, 2020.
Collapse
Affiliation(s)
- Yendry Ventura-Carmenate
- Abu Dhabi Stem Cells Center, Al Misahah Street, Villa No. 25, Rowdhat, Zone-1, POB 4600, Abu Dhabi City, United Arab Emirates
| | | | - Yandy Marx Castillo-Aleman
- Abu Dhabi Stem Cells Center, Al Misahah Street, Villa No. 25, Rowdhat, Zone-1, POB 4600, Abu Dhabi City, United Arab Emirates
| | | | - Yasmine Maher Ahmed
- Abu Dhabi Stem Cells Center, Al Misahah Street, Villa No. 25, Rowdhat, Zone-1, POB 4600, Abu Dhabi City, United Arab Emirates
| | - Pierdanilo Sanna
- Abu Dhabi Stem Cells Center, Al Misahah Street, Villa No. 25, Rowdhat, Zone-1, POB 4600, Abu Dhabi City, United Arab Emirates
| | | | - Abeer Abdelrazik
- Abu Dhabi Stem Cells Center, Al Misahah Street, Villa No. 25, Rowdhat, Zone-1, POB 4600, Abu Dhabi City, United Arab Emirates
| | - Gina Marcela Torres-Zambrano
- Abu Dhabi Stem Cells Center, Al Misahah Street, Villa No. 25, Rowdhat, Zone-1, POB 4600, Abu Dhabi City, United Arab Emirates
| | | | - David Quesada-Saliba
- Miami Dade College, Mathematics Department Chair, Wolfson Campus, Miami, FL 33132 USA
| | - Loubna Abdel Hadi
- Abu Dhabi Stem Cells Center, Al Misahah Street, Villa No. 25, Rowdhat, Zone-1, POB 4600, Abu Dhabi City, United Arab Emirates
| | | | - Rene Antonio Rivero-Jimenez
- Abu Dhabi Stem Cells Center, Al Misahah Street, Villa No. 25, Rowdhat, Zone-1, POB 4600, Abu Dhabi City, United Arab Emirates
| |
Collapse
|
6
|
D’Oronzo S, Silvestris E, Lovero D, Cafforio P, Duda L, Cormio G, Paradiso A, Palmirotta R, Silvestris F. DEAD-Box Helicase 4 (Ddx4) + Stem Cells Sustain Tumor Progression in Non-Serous Ovarian Cancers. Int J Mol Sci 2020; 21:6096. [PMID: 32847044 PMCID: PMC7503840 DOI: 10.3390/ijms21176096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/14/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
DEAD-Box Helicase 4 (Ddx4)+ ovarian stem cells are able to differentiate into several cell types under appropriate stimuli. Ddx4 expression has been correlated with poor prognosis of serous ovarian cancer (OC), while the potential role of Ddx4+ cells in non-serous epithelial OC (NS-EOC) is almost unexplored. The aim of this study was to demonstrate the presence of Ddx4+ cells in NS-EOC and investigate the effect of follicle-stimulating hormone (FSH) on this population. Increased Ddx4 expression was demonstrated in samples from patients with advanced NS-EOC, compared to those with early-stage disease. Under FSH stimulation, OC-derived Ddx4+ cells differentiated into mesenchymal-like (ML) cells, able to deregulate genes involved in cell migration, invasiveness, stemness and chemoresistance in A2780 OC cells. This effect was primarily induced by ML-cells deriving from advanced NS-EOC, suggesting that a tumor-conditioned germ cell niche inhabits its microenvironment and is able to modulate, in a paracrine manner, tumor cell behavior through transcriptome modulation.
Collapse
Affiliation(s)
- Stella D’Oronzo
- Department of Biomedical Sciences and Human Oncology–Section of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro, 70121 Bari, Italy; (D.L.); (P.C.); (R.P.)
- I.R.C.C.S-Giovanni Paolo II Cancer Institute, 70124 Bari, Italy; (E.S.); (G.C.); (A.P.)
| | - Erica Silvestris
- I.R.C.C.S-Giovanni Paolo II Cancer Institute, 70124 Bari, Italy; (E.S.); (G.C.); (A.P.)
| | - Domenica Lovero
- Department of Biomedical Sciences and Human Oncology–Section of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro, 70121 Bari, Italy; (D.L.); (P.C.); (R.P.)
| | - Paola Cafforio
- Department of Biomedical Sciences and Human Oncology–Section of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro, 70121 Bari, Italy; (D.L.); (P.C.); (R.P.)
| | - Loren Duda
- Department of Pathology, University of Foggia, 71122 Foggia, Italy;
| | - Gennaro Cormio
- I.R.C.C.S-Giovanni Paolo II Cancer Institute, 70124 Bari, Italy; (E.S.); (G.C.); (A.P.)
- Department of Biomedical Sciences and Human Oncology–Section of 2nd Unit of Obstetrics and Gynecology, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Angelo Paradiso
- I.R.C.C.S-Giovanni Paolo II Cancer Institute, 70124 Bari, Italy; (E.S.); (G.C.); (A.P.)
| | - Raffaele Palmirotta
- Department of Biomedical Sciences and Human Oncology–Section of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro, 70121 Bari, Italy; (D.L.); (P.C.); (R.P.)
| | - Franco Silvestris
- Department of Biomedical Sciences and Human Oncology–Section of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro, 70121 Bari, Italy; (D.L.); (P.C.); (R.P.)
| |
Collapse
|
7
|
Pluripotent Stem (VSELs) and Progenitor (EnSCs) Cells Exist in Adult Mouse Uterus and Show Cyclic Changes Across Estrus Cycle. Reprod Sci 2020; 28:278-290. [PMID: 32710237 DOI: 10.1007/s43032-020-00250-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/11/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022]
Abstract
We have earlier reported pluripotent, very small embryonic-like stem cells (VSELs) and slightly bigger endometrial stem cells (EnSCs) in adult mouse uterus and their regulation by gonadotropin and steroid hormones. VSELs can differentiate into cells of all three lineages in vitro; however, they neither expand readily in vitro nor compliment a developing embryo. In the present study, a robust protocol is described to enrich uterine stem/progenitor cells along with their characterization and variation across estrus cycle. After enzymatic digestion of adult mouse uterus, single-cell suspension obtained was spun at 1000 rpm (250 g) to pellet majority of cells. Stem cells remain buoyant at this speed and were pelleted by spinning supernatant at 3000 rpm (1000 g). Spherical, darkly stained VSELs (2-6 μm) with high nucleo-cytoplasmic ratio and EnSCs (> 6 μm) expressed OCT-4, NANOG, SSEA-1, SCA-1, and c-KIT. OCT-4-positive cells co-expressed SSEA-1, ERα, ERβ, PR, and FSHR. Transcripts specific for pluripotent state (Oct-4, Oct-4a, Sox-2, Nanog), primordial germ cells (Stella, Fragilis), and receptors for pituitary and steroid hormones (ERα, ERβ, PR, FSHR 1 and 3) were studied by RT-PCR in 3000 rpm pellet. Cell pellet collected at 3000 rpm showed 10-fold enrichment of VSELs (2-6 μm, viable cells with surface phenotype of LIN-CD45-SCA-1+) by flow cytometry and upregulation of pluripotent transcripts by qRT-PCR compared with 1000 rpm pellet. VSELs were maximal during estrus and metestrus phases of estrus cycle. To conclude, VSELs/EnSCs can be enriched from adult uterus using the strategy described here, vary in numbers across estrus cycle, and are vulnerable to endocrine disruption as they express steroid receptors.
Collapse
|
8
|
Markers of Regenerative Processes in Patients with Bipolar Disorder: A Case-control Study. Brain Sci 2020; 10:brainsci10070408. [PMID: 32629800 PMCID: PMC7408571 DOI: 10.3390/brainsci10070408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 11/16/2022] Open
Abstract
Progress in medical science has allowed the discovery of many factors affecting the pathogenesis of bipolar disorder, and among the most recent research directions are found regenerative and inflammatory processes. The role of regenerative processes remains particularly poorly explored, but available data encourage further research, which may explain the pathogenesis of bipolar disorder (BD). The aim of this study was to evaluate the mobilization of stem cells into peripheral blood, in patients with bipolar disorder during stable phase, not treated with lithium salts. The study included 30 unrelated individuals with the diagnosis of bipolar disorder, with disease duration of at least 10 years, not treated with lithium salts for at least five years prior to the study. The control group consisted of 30 healthy subjects, matched for age, sex, body mass index (BMI), origin, socio-demographic factors and nicotine use. Blood samples underwent cytometric analyses to assess concentrations of: Very Small Embryonic Like (VSEL) CD34+, VSEL AC133+, HSC CD34+, HSC AC133+. There were no significant differences in stem cell levels between patients with BD and healthy controls. However, the level of VSEL cells AC133 + was significantly higher in type I BD patients compared to healthy controls. Our results indicate a disturbance in regenerative processes in patients with bipolar disorder.
Collapse
|
9
|
Kaushik A, Bhartiya D. Pluripotent Very Small Embryonic-Like Stem Cells in Adult Testes - An Alternate Premise to Explain Testicular Germ Cell Tumors. Stem Cell Rev Rep 2019; 14:793-800. [PMID: 30238242 DOI: 10.1007/s12015-018-9848-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Developmental exposure to endocrine disruptors has resulted in the increased incidence of infertility and testicular germ cell tumors (T2GCT) in young men residing in developed countries. Unlike T1GCT (infants and young children) and T3GCT (aged men), T2GCT arise from CIS/GCNIS that develops from pre-CIS. Pre-CIS represents undifferentiated, growth-arrested gonocytes that persist in fetal testes due to endocrine disruption. However, whether pre-CIS truly exist, do CIS develop into T2GCT, why no CIS in T1GCT/T3GCT, why germ cell tumors (GCT) also occur along midline at extra-gonadal sites, why T1GCT show partial erasure and T2GCT show complete erasure of genomic imprints are open questions that are awaiting answers. We propose that rather than pre-CIS, pluripotent, very small embryonic-like stem cells (VSELs) get affected by exposure to endocrine disruption. Since VSELs are developmentally equivalent to primordial germ cells (PGCs), T2GCT cells show complete erasure of genomic imprints and CIS represents growth-arrested clonally expanding stem/progenitor cells. PGCs/VSELs migrate along the midline to various organs and this explains why GCT occur along the midline, T1GCT show partial erasure of imprints as they develop from migrating PGCs. T3GCT possibly reflects effects of aging due to compromised differentiation and expansion of pre-meiotic spermatocytes. Absent spermatogenesis in pre-pubertal and aged testes explains absence of CIS in T1GCT and T3GCT. Endocrine disruptors possibly alter epigenetic state of VSELs and thus rather than maintaining normal tissue homeostasis, VSELs undergo increased proliferation and compromised differentiation resulting in reduced sperm count, infertility and TGCT. This newly emerging understanding offers alternate premise to explain TGCT and warrants further exploration.
Collapse
Affiliation(s)
- Ankita Kaushik
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India
| | - Deepa Bhartiya
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India.
| |
Collapse
|
10
|
Bujko K, Kucia M, Ratajczak J, Ratajczak MZ. Hematopoietic Stem and Progenitor Cells (HSPCs). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1201:49-77. [PMID: 31898781 DOI: 10.1007/978-3-030-31206-0_3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hematopoietic stem/progenitor cells (HSPCs) isolated from bone marrow have been successfully employed for 50 years in hematological transplantations. Currently, these cells are more frequently isolated from mobilized peripheral blood or umbilical cord blood. In this chapter, we overview several topics related to these cells including their phenotype, methods for isolation, and in vitro and in vivo assays to evaluate their proliferative potential. The successful clinical application of HSPCs is widely understood to have helped establish the rationale for the development of stem cell therapies and regenerative medicine.
Collapse
Affiliation(s)
- Kamila Bujko
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Magda Kucia
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland.
| |
Collapse
|
11
|
Suman S, Domingues A, Ratajczak J, Ratajczak MZ. Potential Clinical Applications of Stem Cells in Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1201:1-22. [PMID: 31898779 DOI: 10.1007/978-3-030-31206-0_1] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The field of regenerative medicine is looking for a pluripotent/multipotent stem cell able to differentiate across germ layers and be safely employed in therapy. Unfortunately, with the exception of hematopoietic stem/progenitor cells (HSPCs) for hematological applications, the current clinical results with stem cells are somewhat disappointing. The potential clinical applications of the more primitive embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have so far been discouraging, as both have exhibited several problems, including genomic instability, a risk of teratoma formation, and the possibility of rejection. Therefore, the only safe stem cells that have so far been employed in regenerative medicine are monopotent stem cells, such as the abovementioned HSPCs or mesenchymal stem cells (MSCs) isolated from postnatal tissues. However, their monopotency, and therefore limited differentiation potential, is a barrier to their broader application in the clinic. Interestingly, results have accumulated indicating that adult tissues contain rare, early-development stem cells known as very small embryonic-like stem cells (VSELs), which can differentiate into cells from more than one germ layer. This chapter addresses different sources of stem cells for potential clinical application and their advantages and problems to be solved.
Collapse
Affiliation(s)
- Suman Suman
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Alison Domingues
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland.
| |
Collapse
|
12
|
Plausible Links Between Metabolic Networks, Stem Cells, and Longevity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1201:355-388. [PMID: 31898793 DOI: 10.1007/978-3-030-31206-0_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aging is an inevitable consequence of life, and all multicellular organisms undergo a decline in tissue and organ functions as they age. Several well-known risk factors, such as obesity, diabetes, and lack of physical activity that lead to the cardiovascular system, decline and impede the function of vital organs, ultimately limit overall life span. Over recent years, aging research has experienced an unparalleled growth, particularly with the discovery and recognition of genetic pathways and biochemical processes that control to some extent the rate of aging.In this chapter, we focus on several aspects of stem cell biology and aging, beginning with major cellular hallmarks of aging, endocrine regulation of aging and its impact on stem cell compartment, and mechanisms of increased longevity. We then discuss the role of epigenetic modifications associated with aging and provide an overview on a most recent search of antiaging modalities.
Collapse
|
13
|
Changes in cell fate determine the regenerative and functional capacity of the developing kidney before and after release of obstruction. Clin Sci (Lond) 2018; 132:2519-2545. [PMID: 30442812 DOI: 10.1042/cs20180623] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/23/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022]
Abstract
Congenital obstructive nephropathy is a major cause of chronic kidney disease (CKD) in children. The contribution of changes in the identity of renal cells to the pathology of obstructive nephropathy is poorly understood. Using a partial unilateral ureteral obstruction (pUUO) model in genetically modified neonatal mice, we traced the fate of cells derived from the renal stroma, cap mesenchyme, ureteric bud (UB) epithelium, and podocytes using Foxd1Cre, Six2Cre, HoxB7Cre, and Podocyte.Cre mice respectively, crossed with double fluorescent reporter (membrane-targetted tandem dimer Tomato (mT)/membrane-targetted GFP (mG)) mice. Persistent obstruction leads to a significant loss of tubular epithelium, rarefaction of the renal vasculature, and decreased renal blood flow (RBF). In addition, Forkhead Box D1 (Foxd1)-derived pericytes significantly expanded in the interstitial space, acquiring a myofibroblast phenotype. Degeneration of Sine Oculis Homeobox Homolog 2 (Six2) and HoxB7-derived cells resulted in significant loss of glomeruli, nephron tubules, and collecting ducts. Surgical release of obstruction resulted in striking regeneration of tubules, arterioles, interstitium accompanied by an increase in blood flow to the level of sham animals. Contralateral kidneys with remarkable compensatory response to kidney injury showed an increase in density of arteriolar branches. Deciphering the mechanisms involved in kidney repair and regeneration post relief of obstruction has potential therapeutic implications for infants and children and the growing number of adults suffering from CKD.
Collapse
|
14
|
Gounari E, Daniilidis A, Tsagias N, Michopoulou A, Kouzi K, Koliakos G. Isolation of a novel embryonic stem cell cord blood-derived population with in vitro hematopoietic capacity in the presence of Wharton's jelly-derived mesenchymal stromal cells. Cytotherapy 2018; 21:246-259. [PMID: 30522805 DOI: 10.1016/j.jcyt.2018.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/06/2018] [Accepted: 11/13/2018] [Indexed: 01/16/2023]
Abstract
BACKGROUND Recent studies highlight the existence of a population of cord blood (CB)-derived stem cells that bare embryonic features (very small embryonic-like stem cells [VSELs]) as the most primitive CB-stem cell population. In the present study, we present for the first time a novel and high purity isolation method of VSELs with in vitro hematopoietic capacity in the presence of Wharton's jelly-derived mesenchymal stromal cells (WJ-MSCs). METHODS The experimental procedure includes isolation upon gradually increased centrifugation spins and chemotaxis to Stromal cell-derived factor 1a (SDF-1a). Τhis cell population is characterized with flow cytometry, alkaline phosphatase (ALP) staining and qRT-PCR. The functional role of the isolated VSELs is assayed following co-culture with WJ-MSCs or bone marrow-derived mesenchymal stromal cells (BM-MSCs), whereas the stimulation of the quiescent VSEL population is verified via cell cycle analysis. The in vitro hematopoietic capacity is evaluated in methylcellulose cultures and also through induction of erythroid differentiation. RESULTS The final isolated subpopulation is characterized as a small-sized CD45/Lineage-/CXCR4+/CD133+/SSEA-4+cell population, positive in ALP staining and overexpressing the Oct3/4, Nanog and Sox-2 transcription factors. Upon the co-culture with MSCs, a stimulation of the quiescent VSEL population is observed. An impressive increase in the co-expression of the CD34+/CD45+ markers is observed following the co-culture with the WJ-MSCs, which is confirmed by the intense clonogenic ability suggesting in vitro differentiation toward all of the hematopoietic cell lineages and successful differentiation toward erythrocytes. DISCUSSION Conclusively, we propose a novel, rapid and rather simplified isolation method of CB-VSELs, capable of in vitro hematopoiesis.
Collapse
Affiliation(s)
- Eleni Gounari
- Biohellenika Biotechnology Company, Thessaloniki, Greece; Department of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Angelos Daniilidis
- 2nd Department of Obstetrics and Gynecology, Hippokratio General Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Anna Michopoulou
- Biohellenika Biotechnology Company, Thessaloniki, Greece; Department of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kokkona Kouzi
- Biohellenika Biotechnology Company, Thessaloniki, Greece; Department of Histology Embryology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George Koliakos
- Biohellenika Biotechnology Company, Thessaloniki, Greece; Department of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
15
|
Li R, Miao J, Tabaran AF, O’Sullivan MG, Anderson KJ, Scott PM, Wang Z, Cormier RT. A novel cancer syndrome caused by KCNQ1-deficiency in the golden Syrian hamster. J Carcinog 2018; 17:6. [PMID: 30450013 PMCID: PMC6187935 DOI: 10.4103/jcar.jcar_5_18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/31/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The golden Syrian hamster is an emerging model organism. To optimize its use, our group has made the first genetically engineered hamsters. One of the first genes that we investigated is KCNQ1 which encodes for the KCNQ1 potassium channel and also has been implicated as a tumor suppressor gene. MATERIALS AND METHODS We generated KCNQ1 knockout (KO) hamsters by CRISPR/Cas9-mediated gene targeting and investigated the effects of KCNQ1-deficiency on tumorigenesis. RESULTS By 70 days of age seven of the eight homozygous KCNQ1 KOs used in this study began showing signs of distress, and on necropsy six of the seven ill hamsters had visible cancers, including T-cell lymphomas, plasma cell tumors, hemangiosarcomas, and suspect myeloid leukemias. CONCLUSIONS None of the hamsters in our colony that were wild-type or heterozygous for KCNQ1 mutations developed cancers indicating that the cancer phenotype is linked to KCNQ1-deficiency. This study is also the first evidence linking KCNQ1-deficiency to blood cancers.
Collapse
Affiliation(s)
- Rong Li
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, USA
| | - Jinxin Miao
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, USA
| | - Alexandru-Flaviu Tabaran
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Minnesota, Duluth, MN, USA
- Comparative Pathology Shared Resource, Masonic Cancer Center, University of Minnesota, Duluth, MN, USA
| | - M. Gerard O’Sullivan
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Minnesota, Duluth, MN, USA
- Comparative Pathology Shared Resource, Masonic Cancer Center, University of Minnesota, Duluth, MN, USA
| | - Kyle J. Anderson
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, USA
| | - Patricia M. Scott
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, USA
| | - Zhongde Wang
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, USA
| | - Robert T. Cormier
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, USA
| |
Collapse
|
16
|
Ratajczak MZ, Ciechanowicz AK, Kucharska-Mazur J, Samochowiec J. Stem cells and their potential clinical applications in psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:3-9. [PMID: 28435007 PMCID: PMC5623088 DOI: 10.1016/j.pnpbp.2017.04.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/17/2017] [Accepted: 04/19/2017] [Indexed: 12/23/2022]
Abstract
The robustness of stem cells is one of the major factors that directly impacts life quality and life span. Evidence has accumulated that changes in the stem cell compartment affect human mental health and serve as an indicator of psychiatric problems. It is well known that stem cells continuously replace differentiated cells and tissues that are used up during life, although this replacement occurs at a different pace in the various organs. However, the participation of local neural stem cells in regeneration of the central nervous system is controversial. It is known that low numbers of stem cells circulate continuously in peripheral blood (PB) and lymph and undergo a circadian rhythm in their PB level, with the peak occurring early in the morning and the nadir at night, and recent evidence suggests that the number and pattern of circulating stem cells in PB changes in psychotic disorders. On the other hand, progress in the creation of induced pluripotent stem cells (iPSCs) from patient somatic cells provides valuable tools with which to study changes in gene expression in psychotic patients. We will discuss the various potential sources of stem cells that are currently employed in regenerative medicine and the mechanisms that explain some of their beneficial effects as well as the emerging problems with stem cell therapies. However, the main question remains: Will it be possible in the future to modulate the stem cell compartment to reverse psychiatric problems?
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute, 500 South Floyd Street, James Graham Brown Cancer Center, University of Louisville, Louisville 40202, KY, USA; Department of Regenerative Medicine Warsaw Medical University, Warsaw, Poland.
| | | | | | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
17
|
Parte SC, Smolenkov A, Batra SK, Ratajczak MZ, Kakar SS. Ovarian Cancer Stem Cells: Unraveling a Germline Connection. Stem Cells Dev 2017; 26:1781-1803. [PMID: 29078734 PMCID: PMC5725638 DOI: 10.1089/scd.2017.0153] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/24/2017] [Indexed: 12/11/2022] Open
Abstract
Ovarian cancer is most lethal among gynecological cancers with often fatal consequences due to lack of effective biomarkers and relapse, which propels ovarian cancer research into unique directions to establish solid targeted therapeutics. "Ovarian stem cells" expressing germline pluripotent markers serve as novel paradigm with potential to address infertility, menopause, and probably influence tumor initiation. Cancer stem cells (CSCs) pose vital role in tumor recurrence and hence it is extremely important to study them with respect to ovarian stem cells across various cancer stages and normal ovaries. Pluripotent (OCT4, NANOG, SOX2, SSEA1, and SSEA4), germline (IFITM3, VASA/DDX4), and cancer stem (CD44, LGR5) cell specific markers were characterized for protein and mRNA expression in tumor tissues to understand their distribution in the surface epithelium and ovarian cortex in benign, borderline, and high-grade malignant stages. To elucidate whether pluripotent ovarian germline stem cells and CSCs are common subset of stem cells in tumor tissues, VASA was colocalized with known pluripotent stem (OCT4, SSEA1, SSEA4) and CSC (CD44, LGR5) specific markers by confocal microscopy. Single, smaller spherical (≤5 μm), and larger elliptical fibroblast like (≥10 μm) cells (also in clusters or multiples) were detected implying probable functional behavioral significance of cells in tumor initiation and metastasis across various cancer stages. Cells revealed characteristic staining pattern in ovarian surface epithelium (OSE) and cortex regions exclusive for each marker. Co-expression studies revealed specific subpopulations existing simultaneously in OSE and cortex and that a dynamic hierarchy of (cancer) stem cells with germline properties prevails in normal ovaries and cancer stages. Novel insights into CSC biology with respect to ovarian and germline stem cell perspective were obtained. Understanding molecular signatures and distribution within ovarian tissue may enable identification of precise tumor-initiating CSC populations and signaling pathways thus improving their efficient targeting and strategies to prevent their dissemination causing fatal relapse.
Collapse
Affiliation(s)
- Seema C. Parte
- Department of Physiology, University of Louisville, Louisville, Kentucky
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Andrei Smolenkov
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Mariusz Z. Ratajczak
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
- Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Sham S. Kakar
- Department of Physiology, University of Louisville, Louisville, Kentucky
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| |
Collapse
|
18
|
Ratajczak MZ, Ratajczak J, Suszynska M, Miller DM, Kucia M, Shin DM. A Novel View of the Adult Stem Cell Compartment From the Perspective of a Quiescent Population of Very Small Embryonic-Like Stem Cells. Circ Res 2017; 120:166-178. [PMID: 28057792 PMCID: PMC5221475 DOI: 10.1161/circresaha.116.309362] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 12/12/2022]
Abstract
Evidence has accumulated that adult hematopoietic tissues and other organs contain a population of dormant stem cells (SCs) that are more primitive than other, already restricted, monopotent tissue-committed SCs (TCSCs). These observations raise several questions, such as the developmental origin of these cells, their true pluripotent or multipotent nature, which surface markers they express, how they can be efficiently isolated from adult tissues, and what role they play in the adult organism. The phenotype of these cells and expression of some genes characteristic of embryonic SCs, epiblast SCs, and primordial germ cells suggests their early-embryonic deposition in developing tissues as precursors of adult SCs. In this review, we will critically discuss all these questions and the concept that small dormant SCs related to migratory primordial germ cells, described as very small embryonic-like SCs, are deposited during embryogenesis in bone marrow and other organs as a backup population for adult tissue-committed SCs and are involved in several processes related to tissue or organ rejuvenation, aging, and cancerogenesis. The most recent results on successful ex vivo expansion of human very small embryonic-like SC in chemically defined media free from feeder-layer cells open up new and exciting possibilities for their application in regenerative medicine.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- From the Department of Medicine, Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, KY (M.Z.R., J.R., M.S., D.M.M., M.K.); Department of Regenerative Medicine, Warsaw Medical University, Poland (M.Z.R., M.K.); and Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea (D.-M.S.).
| | - Janina Ratajczak
- From the Department of Medicine, Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, KY (M.Z.R., J.R., M.S., D.M.M., M.K.); Department of Regenerative Medicine, Warsaw Medical University, Poland (M.Z.R., M.K.); and Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea (D.-M.S.)
| | - Malwina Suszynska
- From the Department of Medicine, Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, KY (M.Z.R., J.R., M.S., D.M.M., M.K.); Department of Regenerative Medicine, Warsaw Medical University, Poland (M.Z.R., M.K.); and Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea (D.-M.S.)
| | - Donald M Miller
- From the Department of Medicine, Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, KY (M.Z.R., J.R., M.S., D.M.M., M.K.); Department of Regenerative Medicine, Warsaw Medical University, Poland (M.Z.R., M.K.); and Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea (D.-M.S.)
| | - Magda Kucia
- From the Department of Medicine, Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, KY (M.Z.R., J.R., M.S., D.M.M., M.K.); Department of Regenerative Medicine, Warsaw Medical University, Poland (M.Z.R., M.K.); and Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea (D.-M.S.)
| | - Dong-Myung Shin
- From the Department of Medicine, Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, KY (M.Z.R., J.R., M.S., D.M.M., M.K.); Department of Regenerative Medicine, Warsaw Medical University, Poland (M.Z.R., M.K.); and Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea (D.-M.S.)
| |
Collapse
|
19
|
Bhartiya D, Shaikh A, Anand S, Patel H, Kapoor S, Sriraman K, Parte S, Unni S. Endogenous, very small embryonic-like stem cells: critical review, therapeutic potential and a look ahead. Hum Reprod Update 2016; 23:41-76. [PMID: 27614362 DOI: 10.1093/humupd/dmw030] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/27/2016] [Accepted: 08/04/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Both pluripotent very small embryonic-like stem cells (VSELs) and induced pluripotent stem (iPS) cells were reported in 2006. In 2012, a Nobel Prize was awarded for iPS technology whereas even today the very existence of VSELs is not well accepted. The underlying reason is that VSELs exist in low numbers, remain dormant under homeostatic conditions, are very small in size and do not pellet down at 250-280g. The VSELs maintain life-long tissue homeostasis, serve as a backup pool for adult stem cells and are mobilized under stress conditions. An imbalance in VSELs function (uncontrolled proliferation) may result in cancer. SEARCH METHODS The electronic database 'Medline/Pubmed' was systematically searched with the subject heading term 'very small embryonic-like stem cells'. OBJECTIVE AND RATIONALE The most primitive stem cells that undergo asymmetric cell divisions to self-renew and give rise to progenitors still remain elusive in the hematopoietic system and testes, while the presence of stem cells in ovary is still being debated. We propose to review the available literature on VSELs, the methods of their isolation and characterization, their ontogeny, how they compare with embryonic stem (ES) cells, primordial germ cells (PGCs) and iPS cells, and their role in maintaining tissue homeostasis. The review includes a look ahead on how VSELs will result in paradigm shifts in basic reproductive biology. OUTCOMES Adult tissue-specific stem cells including hematopoietic, spermatogonial, ovarian and mesenchymal stem cells have good proliferation potential and are indeed committed progenitors (with cytoplasmic OCT-4), which arise by asymmetric cell divisions of pluripotent VSELs (with nuclear OCT-4). VSELs are the most primitive stem cells and postulated to be an overlapping population with the PGCs. Rather than migrating only to the gonads, PGCs migrate and survive in various adult body organs throughout life as VSELs. VSELs express both pluripotent and PGC-specific markers and are epigenetically and developmentally more mature compared with ES cells obtained from the inner cell mass of a blastocyst-stage embryo. As a result, VSELs readily differentiate into three embryonic germ layers and spontaneously give rise to both sperm and oocytes in vitro. Like PGCs, VSELs do not divide readily in culture, nor produce teratoma or integrate in the developing embryo. But this property of being relatively quiescent allows endogenous VSELs to survive various kinds of toxic insults. VSELs that survive oncotherapy can be targeted to induce endogenous regeneration of non-functional gonads. Transplanting healthy niche (mesenchymal) cells have resulted in improved gonadal function and live births. WIDER IMPLICATIONS Being quiescent, VSELs possibly do not accumulate genomic (nuclear or mitochondrial) mutations and thus may be ideal endogenous, pluripotent stem cell candidates for regenerative and reproductive medicine. The presence of VSELs in adult gonads and the fact that they survive oncotherapy may obviate the need to bank gonadal tissue for fertility preservation prior to oncotherapy. VSELs and their ability to undergo spermatogenesis/neo-oogenesis in the presence of a healthy niche will help identify newer strategies toward fertility restoration in cancer survivors, delaying menopause and also enabling aged mothers to have better quality eggs.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Ambreen Shaikh
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Sandhya Anand
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Hiren Patel
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Sona Kapoor
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Kalpana Sriraman
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India.,The Foundation for Medical Research, 84-A, RG Thadani Marg, Worli, Mumbai 400018, India
| | - Seema Parte
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India.,Department of Physiology, James Graham Brown Cancer Centre, University of Louisville School of Medicine, 2301 S 3rd St, Louisville, KY 40202, USA
| | - Sreepoorna Unni
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India.,Inter Disciplinary Studies Department, University College, Zayed University, Academic City, PO Box 19282, Dubai, United Arab Emirates
| |
Collapse
|
20
|
Maj M, Schneider G, Ratajczak J, Suszynska M, Kucia M, Ratajczak MZ. The cell cycle- and insulin-signaling-inhibiting miRNA expression pattern of very small embryonic-like stem cells contributes to their quiescent state. Exp Biol Med (Maywood) 2015; 240:1107-1111. [PMID: 25966979 PMCID: PMC4935278 DOI: 10.1177/1535370215584940] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/23/2015] [Indexed: 11/15/2022] Open
Abstract
Murine Oct4(+), very small embryonic-like stem cells (VSELs), are a quiescent stem cell population that requires a supportive co-culture layer to proliferate and/or to differentiate in vitro. Gene expression studies have revealed that the quiescence of these cells is due to changes in expression of parentally imprinted genes, including genes involved in cell cycle regulation and insulin and insulin-like growth factor signaling (IIS). To investigate the role of microRNAs (miRNAs) in VSEL quiescence, we performed miRNA studies in highly purified VSELs and observed a unique miRNA expression pattern in these cells. Specifically, we observed significant differences in the expression of certain miRNA species (relative to a reference cell population), including (i) miRNA-25_1 and miRNA-19 b, whose downregulation has the effect of upregulating cell cycle checkpoint genes and (ii) miRNA-675-3 p and miRNA-675-5 p, miRNA-292-5 p, miRNA-184, and miRNA-125 b, whose upregulation attenuates IIS. These observations are important for understanding the biology of these cells and for developing efficient ex vivo expansion strategies for VSELs isolated from adult tissues.
Collapse
Affiliation(s)
- Magdalena Maj
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Gabriela Schneider
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Janina Ratajczak
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Malwina Suszynska
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Magda Kucia
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA Department of Regenerative Medicine, Warsaw Medical University, Warsaw 02-091, Poland
| | - Mariusz Z Ratajczak
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA Department of Regenerative Medicine, Warsaw Medical University, Warsaw 02-091, Poland
| |
Collapse
|
21
|
Bhartiya D, Patel H. Very small embryonic-like stem cells are involved in pancreatic regeneration and their dysfunction with age may lead to diabetes and cancer. Stem Cell Res Ther 2015; 6:96. [PMID: 25976079 PMCID: PMC4432983 DOI: 10.1186/s13287-015-0084-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mouse pancreas has a remarkable ability to regenerate after partial pancreatectomy, and several investigators have studied the underlying mechanisms involved in this regeneration process; however, the field remains contentious. Elegant lineage-tracing studies undertaken over a decade have generated strong evidence against neogenesis from stem cells and in favor of reduplication of pre-existing islets. Ductal epithelium has also been implicated during regeneration. We recently provided direct evidence for the possible involvement of very small embryonic-like stem cells (VSELs) during regeneration after partial pancreatectomy in mice. VSELs were first reported in pancreas in 2008 and are mobilized in large numbers after treating mice with streptozotocin and in patients with pancreatic cancer. VSELs can be detected in mouse pancreas as small-sized LIN−/CD45−/SCA-1+ cells (3 to 5 μm), present in small numbers (0.6%), which express nuclear Oct-4 (octamer-binding transcription factor 4) and other pluripotent markers along with their immediate descendant ‘progenitors’, which are slightly bigger and co-express Oct-4 and PDX-1. VSELs and the progenitors get mobilized in large numbers after partial pancreatectomy and regenerate both pancreatic islets and acinar cells. In this review, we deliberate upon possible reasons why VSELs have eluded scientists so far. Because of their small size, VSELs are probably unknowingly and inadvertently discarded during processing. Similar to menopause and related loss of ovarian function, type 2 diabetes mellitus occurs because of a decline in beta-cell function possibly resulting from an age-related compromised niche which does not allow VSELs to maintain normal homeostasis. As suggested earlier for ovarian cancers, the presence of Oct-4 and other pluripotent markers in pancreatic cancers is suggestive of VSELs as the possible cancer-initiating stem cells. Several issues raised in the review require urgent confirmation and thus provide scope for further research before arriving at a consensus on the fundamental role played by VSELs in normal pancreas biology and during regeneration, aging, and cancer. In the future, such understanding may allow manipulation of endogenous VSELs to our advantage in patients with diabetes and also to treat cancer.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, JM Street, Parel, Mumbai, 400012, India.
| | - Hiren Patel
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, JM Street, Parel, Mumbai, 400012, India.
| |
Collapse
|
22
|
Stem Cells in Regenerative Therapy. Bioengineering (Basel) 2015. [DOI: 10.1007/978-3-319-10798-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
23
|
Grymula K, Tarnowski M, Piotrowska K, Suszynska M, Mierzejewska K, Borkowska S, Fiedorowicz K, Kucia M, Ratajczak MZ. Evidence that the population of quiescent bone marrow-residing very small embryonic/epiblast-like stem cells (VSELs) expands in response to neurotoxic treatment. J Cell Mol Med 2014; 18:1797-1806. [PMID: 24895014 PMCID: PMC4162847 DOI: 10.1111/jcmm.12315] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/02/2014] [Indexed: 12/18/2022] Open
Abstract
The concept that bone marrow (BM)-derived cells may participate in neural regeneration remains controversial, and the identity of the specific cell type(s) involved remains unknown. We recently reported that the adult murine BM contains a highly mobile population of Sca-1(+) Lin(-) CD45(-) cells known as very small embryonic/epiblast-like stem cells (VSELs) that express several markers of pluripotency such as Oct-4. In the BM microenvironment, these cells are kept quiescent because of epigenetic modification of certain paternally imprinted genes. However, as reported, these cells can be mobilized in mice in an experimental model of stroke and express several genes involved in neurogenesis while circulating in peripheral blood (PB). In the current work, we employed a model of toxic brain damage, which is induced by administration of kainic acid, to see not only whether VSELs can be mobilized into PB in response to this neurotoxin, but, more importantly, whether they proliferate and expand in BM tissue. We report here for the first time that brain damage leads to activation and expansion of the BM pool of quiescent VSELs, which precedes their subsequent egress into PB. Harnessing these cells in neural tissue regeneration is currently one of the challenges in regenerative medicine.
Collapse
Affiliation(s)
- Katarzyna Grymula
- Department of Physiology, Pomeranian Medical UniversitySzczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology, Pomeranian Medical UniversitySzczecin, Poland
| | | | - Malwina Suszynska
- Department of Physiology, Pomeranian Medical UniversitySzczecin, Poland
| | | | - Sylwia Borkowska
- Department of Physiology, Pomeranian Medical UniversitySzczecin, Poland
| | | | - Magda Kucia
- Department of Physiology, Pomeranian Medical UniversitySzczecin, Poland
- Stem Cell Institute at James Graham Brown Cancer Center, University of LouisvilleLouisville, KY, USA
| | - Mariusz Z Ratajczak
- Department of Physiology, Pomeranian Medical UniversitySzczecin, Poland
- Stem Cell Institute at James Graham Brown Cancer Center, University of LouisvilleLouisville, KY, USA
| |
Collapse
|
24
|
Ratajczak MZ, Marycz K, Poniewierska-Baran A, Fiedorowicz K, Zbucka-Kretowska M, Moniuszko M. Very small embryonic-like stem cells as a novel developmental concept and the hierarchy of the stem cell compartment. Adv Med Sci 2014; 59:273-280. [PMID: 25170822 DOI: 10.1016/j.advms.2014.08.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/06/2014] [Accepted: 08/04/2014] [Indexed: 01/14/2023]
Abstract
Our current understanding of stem cells suffers from a lack of precision, as the stem cell compartment is a broad continuum between early stages of development and adult postnatal tissues, and it is not fully understood how this transition occurs. The definition of stem cell pluripotency is adapted from embryology and excludes the possibility that some early-development stem cells with pluri- and/or multipotential differentiation potential may reside in postnatal tissues in a dormant state in which they are protected from uncontrolled proliferation and thus do not form teratomas or have the ability to complement blastocyst development. We will discuss the concept that a population of very small embryonic-like stem cells (VSELs) could be a link between early-development stages and adult stem cell compartments and reside in a quiescent state in adult tissues. The epigenetic mechanism identified that changes expression of certain genes involved in insulin/insulin-like growth factor signaling (IIS) in VSELs, on the one hand, keeps these cells quiescent in adult tissues and, on the other hand, provides a novel view of the stem cell compartment, IIS, tissue/organ rejuvenation, aging, and cancerogenesis.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA; Department of Physiology, Pomeranian Medical University, Szczecin, Poland.
| | - Krzysztof Marycz
- University of Environmental and Life Sciences, Electron Microscopy Laboratory, Wroclaw, Poland; Wroclaw Research Centre EIT+, Wroclaw, Poland
| | - Agata Poniewierska-Baran
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA; Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | | | - Monika Zbucka-Kretowska
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland; Department of Allergology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
25
|
Suszynska M, Poniewierska-Baran A, Gunjal P, Ratajczak J, Marycz K, Kakar SS, Kucia M, Ratajczak MZ. Expression of the erythropoietin receptor by germline-derived cells - further support for a potential developmental link between the germline and hematopoiesis. J Ovarian Res 2014; 7:66. [PMID: 24982693 PMCID: PMC4074848 DOI: 10.1186/1757-2215-7-66] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 06/11/2014] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Expressing several markers of migrating primordial germ cells (PGCs), the rare population of quiescent, bone marrow (BM)-residing very small embryonic-like stem cells (VSELs) can be specified like PGCs into hematopoietic stem/progenitor cells (HSPCs). These two properties of VSELs support the possibility of a developmental origin of HSPCs from migrating PGCs. METHODS To address a potential link between VSELs and germ line cells we analyzed by RT-PCR and FACS expression of erythropoietin receptor (EpoR) on murine bone marrow- and human umbilical cord blood-derived VSELs, murine and human teratocarcinoma cell lines and human ovarian cancer cells. A proper gating strategy and immunostaining excluded from FACS analysis potential contamination by erythroblasts. Furthermore, the transwell chemotaxis assays as well as adhesion and signaling studies were performed to demonstrate functionality of erythropoietin - EpoR axes on these cells. RESULTS We report here that murine and human VSELs as well as murine and human teratocarcinoma cell lines and ovarian cancer cell lines share a functional EpoR. CONCLUSIONS Our data provide more evidence of a potential developmental link between germline cells, VSELs, and HSCs and sheds more light on the developmental hierarchy of the stem cell compartment in adult tissues.
Collapse
Affiliation(s)
- Malwina Suszynska
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202, USA ; Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Agata Poniewierska-Baran
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202, USA ; Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Pranesh Gunjal
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202, USA
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202, USA ; Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | | | - Sham S Kakar
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202, USA
| | - Magda Kucia
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202, USA ; Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202, USA ; Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
26
|
Suszynska M, Zuba-Surma EK, Maj M, Mierzejewska K, Ratajczak J, Kucia M, Ratajczak MZ. The proper criteria for identification and sorting of very small embryonic-like stem cells, and some nomenclature issues. Stem Cells Dev 2014; 23:702-713. [PMID: 24299281 PMCID: PMC3967357 DOI: 10.1089/scd.2013.0472] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/27/2013] [Indexed: 01/14/2023] Open
Abstract
Evidence has accumulated that both murine and human adult tissues contain early-development stem cells with a broader differentiation potential than other adult monopotent stem cells. These cells, being pluripotent or multipotent, exist at different levels of specification and most likely represent overlapping populations of cells that, depending on the isolation strategy, ex vivo expansion protocol, and markers employed for their identification, have been given different names. In this review, we will discuss a population of very small embryonic-like stem cells (VSELs) in the context of other stem cells that express pluripotent/multipotent markers isolated from adult tissues as well as review the most current, validated working criteria on how to properly identify and isolate these very rare cells. VSELs have been successfully purified in several laboratories; however, a few have failed to isolate them, which has raised some unnecessary controversy in the field. Therefore, in this short review, we will address the most important reasons that some investigators have experienced problems in isolating these very rare cells and discuss some still unresolved challenges which should be overcome before these cells can be widely employed in the clinic.
Collapse
Affiliation(s)
- Malwina Suszynska
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
- Department of Physiology Pomeranian Medial University, Szczecin, Poland
| | - Ewa K. Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Magdalena Maj
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | | | - Janina Ratajczak
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
- Department of Physiology Pomeranian Medial University, Szczecin, Poland
| | - Magda Kucia
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
- Department of Physiology Pomeranian Medial University, Szczecin, Poland
| | - Mariusz Z. Ratajczak
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
- Department of Physiology Pomeranian Medial University, Szczecin, Poland
| |
Collapse
|
27
|
Ratajczak MZ, Zuba-Surma E, Wojakowski W, Suszynska M, Mierzejewska K, Liu R, Ratajczak J, Shin DM, Kucia M. Very small embryonic-like stem cells (VSELs) represent a real challenge in stem cell biology: recent pros and cons in the midst of a lively debate. Leukemia 2014; 28:473-484. [PMID: 24018851 PMCID: PMC3948156 DOI: 10.1038/leu.2013.255] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 08/28/2013] [Indexed: 02/07/2023]
Abstract
The concept that adult tissue, including bone marrow (BM), contains early-development cells with broader differentiation potential has again been recently challenged. In response, we would like to review the accumulated evidence from several independent laboratories that adult tissues, including BM, harbor a population of very rare stem cells that may cross germ layers in their differentiation potential. Thus, the BM stem cell compartment hierarchy needs to be revisited. These dormant, early-development cells that our group described as very small embryonic-like stem cells (VSELs) most likely overlap with similar populations of stem cells that have been identified in adult tissues by other investigators as the result of various experimental strategies and have been given various names. As reported, murine VSELs have some pluripotent stem cell characteristics. Moreover, they display several epiblast/germline markers that suggest their embryonic origin and developmental deposition in adult BM. Moreover, at the molecular level, changes in expression of parentally imprinted genes (for example, Igf2-H19) and resistance to insulin/insulin-like growth factor signaling (IIS) regulates their quiescent state in adult tissues. In several emergency situations related to organ damage, VSELs can be activated and mobilized into peripheral blood, and in appropriate animal models they contribute to tissue organ/regeneration. Interestingly, their number correlates with lifespan in mice, and they may also be involved in some malignancies. VSELs have been successfully isolated in several laboratories; however, some investigators experience problems with their isolation.
Collapse
Affiliation(s)
- M Z Ratajczak
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - E Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - W Wojakowski
- Third Department of Cardiology, Silesian Medical University, Katowice, Poland
| | - M Suszynska
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - K Mierzejewska
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - R Liu
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - J Ratajczak
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - D M Shin
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - M Kucia
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
28
|
Shin DM, Suszynska M, Mierzejewska K, Ratajczak J, Ratajczak MZ. Very small embryonic-like stem-cell optimization of isolation protocols: an update of molecular signatures and a review of current in vivo applications. Exp Mol Med 2013; 45:e56. [PMID: 24232255 PMCID: PMC3849570 DOI: 10.1038/emm.2013.117] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 07/22/2013] [Indexed: 01/09/2023] Open
Abstract
As the theory of stem cell plasticity was first proposed, we have explored an alternative hypothesis for this phenomenon: namely that adult bone marrow (BM) and umbilical cord blood (UCB) contain more developmentally primitive cells than hematopoietic stem cells (HSCs). In support of this notion, using multiparameter sorting we were able to isolate small Sca1(+)Lin(-)CD45(-) cells and CD133(+)Lin(-)CD45(-) cells from murine BM and human UCB, respectively, which were further enriched for the detection of various early developmental markers such as the SSEA antigen on the surface and the Oct4 and Nanog transcription factors in the nucleus. Similar populations of cells have been found in various organs by our team and others, including the heart, brain and gonads. Owing to their primitive cellular features, such as the high nuclear/cytoplasm ratio and the presence of euchromatin, they are called very small embryonic-like stem cells (VSELs). In the appropriate in vivo models, VSELs differentiate into long-term repopulating HSCs, mesenchymal stem cells (MSCs), lung epithelial cells, cardiomyocytes and gametes. In this review, we discuss the most recent data from our laboratory and other groups regarding the optimal isolation procedures and describe the updated molecular characteristics of VSELs.
Collapse
Affiliation(s)
- Dong-Myung Shin
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Malwina Suszynska
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Kasia Mierzejewska
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| |
Collapse
|
29
|
Wnt/β-catenin signaling triggers neuron reprogramming and regeneration in the mouse retina. Cell Rep 2013; 4:271-86. [PMID: 23850287 DOI: 10.1016/j.celrep.2013.06.015] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 03/01/2013] [Accepted: 06/11/2013] [Indexed: 12/11/2022] Open
Abstract
Cell-fusion-mediated somatic-cell reprogramming can be induced in culture; however, whether this process occurs in mammalian tissues remains enigmatic. Here, we show that upon activation of Wnt/β-catenin signaling, mouse retinal neurons can be transiently reprogrammed in vivo back to a precursor stage. This occurs after their spontaneous fusion with transplanted hematopoietic stem and progenitor cells (HSPCs). Moreover, we demonstrate that retinal damage is essential for cell-hybrid formation in vivo. Newly formed hybrids can proliferate, commit to differentiation toward a neuroectodermal lineage, and finally develop into terminally differentiated neurons. This results in partial regeneration of the damaged retinal tissue, with functional rescue. Following retinal damage and induction of Wnt/β-catenin signaling, cell-fusion-mediated reprogramming also occurs after endogenous recruitment of bone-marrow-derived cells in the eyes. Our data demonstrate that in vivo reprogramming of terminally differentiated retinal neurons after their fusion with HSPCs is a potential mechanism for tissue regeneration.
Collapse
|
30
|
Gao L, Thilakavathy K, Nordin N. A plethora of human pluripotent stem cells. Cell Biol Int 2013; 37:875-87. [DOI: 10.1002/cbin.10120] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 04/15/2013] [Indexed: 12/28/2022]
Affiliation(s)
- Liyang Gao
- Clinical Genetics Unit; Department of Obstetrics & Gynaecology; Faculty of Medicine & Health Sciences; Universiti Putra Malaysia; 43400; UPM Serdang; Selangor; Malaysia
| | | | | |
Collapse
|
31
|
Twigger AJ, Hodgetts S, Filgueira L, Hartmann PE, Hassiotou F. From breast milk to brains: the potential of stem cells in human milk. J Hum Lact 2013; 29:136-9. [PMID: 23515086 DOI: 10.1177/0890334413475528] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Alecia-Jane Twigger
- School of Anatomy, Physiology and Human Biology, Faculty of Science, The University of Western Australia, Perth, WA, Australia
| | | | | | | | | |
Collapse
|
32
|
Kassmer SH, Krause DS. Very small embryonic-like cells: biology and function of these potential endogenous pluripotent stem cells in adult tissues. Mol Reprod Dev 2013; 80:677-90. [PMID: 23440892 DOI: 10.1002/mrd.22168] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 02/17/2013] [Indexed: 01/15/2023]
Abstract
Very small embryonic-like cells (VSELs), found in murine bone marrow and other adult tissues, are small, non-hematopoietic cells expressing markers of pluripotent embryonic and primordial germ cells. A similar cell type in humans has begun to be characterized, though with a slightly different phenotype and surface markers. Consistent with expression of pluripotency genes, murine VSELs differentiate into cell types from three germ-layer lineages in vitro, though pluripotency has yet to be shown at the single-cell level or in vivo. VSELs appear to be quiescent under steady state conditions, apparently due to partially erased imprinting and overexpression of cell cycle inhibitory genes. In vivo, VSELs can enter the cell cycle under stress conditions, but which factors regulate quiescence versus proliferation and self-renewal versus differentiation are as yet unknown, and in vitro conditions that induce proliferation and self-renewal have yet to be defined. Future experiments are needed to address whether a VSEL niche actively regulates quiescence in vivo or quiescence is cell autonomous under steady state conditions. Insights into these mechanisms may help to address whether or not VSELs could play a role in regenerative medicine in the future.
Collapse
Affiliation(s)
- Susannah H Kassmer
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
33
|
Ratajczak MZ, Mierzejewska K, Ratajczak J, Kucia M. CD133 Expression Strongly Correlates with the Phenotype of Very Small Embryonic-/Epiblast-Like Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 777:125-141. [PMID: 23161080 PMCID: PMC5565199 DOI: 10.1007/978-1-4614-5894-4_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CD133 antigen (prominin-1) is a useful cell surface marker of very small embryonic-like stem cells (VSELs). Antibodies against it, conjugated to paramagnetic beads or fluorochromes, are thus powerful biological tools for their isolation from human umbilical cord blood, mobilized peripheral blood, and bone marrow. VSELs are described with the following characteristics: (1) are slightly smaller than red blood cells; (2) display a distinct morphology, typified by a high nuclear/cytoplasmic ratio and an unorganized euchromatin; (3) become mobilized during stress situations into peripheral blood; (4) are enriched in the CD133(+)Lin(-)CD45(-) cell fraction in humans; and (5) express markers of pluripotent stem cells (e.g., Oct-4, Nanog, and stage-specific embryonic antigen-4). The most recent in vivo data from our and other laboratories demonstrated that human VSELs exhibit some characteristics of long-term repopulating hematopoietic stem cells and are at the top of the hierarchy in the mesenchymal lineage. However, still more labor is needed to characterize better at a molecular level these rare cells.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, 40202 Rm. 107, Louisville, KY, USA,
| | | | | | | |
Collapse
|
34
|
Samardzija C, Quinn M, Findlay JK, Ahmed N. Attributes of Oct4 in stem cell biology: perspectives on cancer stem cells of the ovary. J Ovarian Res 2012; 5:37. [PMID: 23171809 PMCID: PMC3536609 DOI: 10.1186/1757-2215-5-37] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 10/30/2012] [Indexed: 01/05/2023] Open
Abstract
Epithelial ovarian cancer (EOC) remains the most lethal of all the gynaecological malignancies with drug resistance and recurrence remaining the major therapeutic barrier in the management of the disease. Although several studies have been undertaken to understand the mechanisms responsible for chemoresistance and subsequent recurrence in EOC, the exact mechanisms associated with chemoresistance/recurrence continue to remain elusive. Recent studies have shown that the parallel characteristics commonly seen between embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSC) are also shared by a relatively rare population of cells within tumors that display stem cell-like features. These cells, termed 'cancer initiating cells' or 'cancer stem cells (CSCs)' have been shown not only to display increased self renewal and pluripotent abilities as seen in ESCs and iPSCs, but are also highly tumorigenic in in vivo mouse models. Additionally, these CSCs have been implicated in tumor recurrence and chemoresistance, and when isolated have consistently shown to express the master pluripotency and embryonic stem cell regulating gene Oct4. This article reviews the involvement of Oct4 in cancer progression and chemoresistance, with emphasis on ovarian cancer. Overall, we highlight why ovarian cancer patients, who initially respond to conventional chemotherapy subsequently relapse with recurrent chemoresistant disease that is essentially incurable.
Collapse
Affiliation(s)
- Chantel Samardzija
- Women's Cancer Research Centre, Royal Women's Hospital, 20 Flemington Road, Parkville, VIC, 3052, Australia.
| | | | | | | |
Collapse
|
35
|
Ratajczak MZ, Shin DM, Liu R, Mierzejewska K, Ratajczak J, Kucia M, Zuba-Surma EK. Very small embryonic/epiblast-like stem cells (VSELs) and their potential role in aging and organ rejuvenation--an update and comparison to other primitive small stem cells isolated from adult tissues. Aging (Albany NY) 2012; 4:235-246. [PMID: 22498452 PMCID: PMC3371759 DOI: 10.18632/aging.100449] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 04/05/2012] [Indexed: 02/07/2023]
Abstract
Very small embryonic-like stem cells (VSELs) are a population of developmentally early stem cells residing in adult tissues. These rare cells, which are slightly smaller than red blood cells, i) become mobilized during stress situations into peripheral blood, ii) are enriched in the Sca1+Lin-CD45- cell fraction in mice and the CD133+ Lin-CD45- cell fraction in humans, iii) express markers of pluripotent stem cells (e.g., Oct4, Nanog, and SSEA), and iv) display a distinct morphology characterized by a high nuclear/cytoplasmic ratio and undifferentiated chromatin. Recent evidence indicates that murine VSELs are kept quiescent in adult tissues and protected from teratoma formation by epigenetic modification of imprinted genes that regulate insulin/insulin like growth factor signaling (IIS). The successful reversal of these epigenetic changes in VSELs that render them quiescent will be crucial for efficient expansion of these cells. The most recent data in vivo from our and other laboratories demonstrated that both murine and human VSELs exhibit some characteristics of long-term repopulating hematopoietic stem cells (LT-HSCs), are at the top of the hierarchy in the mesenchymal lineage, and may differentiate into organ-specific cells (e.g., cardiomyocytes). Moreover, as recently demonstrated the number of these cells positively correlates in several murine models with longevity. Finally, while murine BM-derived VSELs have been extensively characterized more work is needed to better characterize these small cells at the molecular level in humans.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, Louisville, KT, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Borlongan CV, Glover LE, Tajiri N, Kaneko Y, Freeman TB. The great migration of bone marrow-derived stem cells toward the ischemic brain: therapeutic implications for stroke and other neurological disorders. Prog Neurobiol 2011; 95:213-28. [PMID: 21903148 PMCID: PMC3185169 DOI: 10.1016/j.pneurobio.2011.08.005] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 08/10/2011] [Accepted: 08/15/2011] [Indexed: 02/08/2023]
Abstract
Accumulating laboratory studies have implicated the mobilization of bone marrow (BM)-derived stem cells in brain plasticity and stroke therapy. This mobilization of bone cells to the brain is an essential concept in regenerative medicine. Over the past ten years, mounting data have shown the ability of bone marrow-derived stem cells to mobilize from BM to the peripheral blood (PB) and eventually enter the injured brain. This homing action is exemplified in BM stem cell mobilization following ischemic brain injury. Various BM-derived cells, such as hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs) and very small embryonic-like cells (VSELs) have been demonstrated to exert therapeutic benefits in stroke. Here, we discuss the current status of these BM-derived stem cells in stroke therapy, with emphasis on possible cellular and molecular mechanisms of action that mediate the cells' beneficial effects in the ischemic brain. When possible, we also discuss the relevance of this therapeutic regimen in other central nervous system (CNS) disorders.
Collapse
Affiliation(s)
- Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, USA.
| | | | | | | | | |
Collapse
|
37
|
Zuba-Surma EK, Wojakowski W, Ratajczak MZ, Dawn B. Very small embryonic-like stem cells: biology and therapeutic potential for heart repair. Antioxid Redox Signal 2011; 15:1821-34. [PMID: 21194389 PMCID: PMC3159118 DOI: 10.1089/ars.2010.3817] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Very small embryonic-like stem cells (VSELs) represent a population of extremely small nonhematopoietic pluripotent cells that are negative for lineage markers and express Sca-1 in mice and CD133 in humans. Their embryonic-like characteristics include the expression of markers of pluripotency; the ability to give rise to cellular derivatives of all three germ-layers; and the ability to form embryoid-like bodies. Indeed, quiescent VSELs may represent the remnants of epiblast-derived cells in adult organs. After tissue injury, including acute myocardial infarction (MI), bone marrow-derived VSELs are mobilized into the peripheral blood and home to the damaged organ. Given the ability of VSELs to differentiate into cardiomyocytes and endothelial cells, and their ability to secrete various cardioprotective growth factors/cytokines, VSELs may serve as an ideal cellular source for cardiac repair. Consistently, transplantation of VSELs after an acute MI improves left ventricular (LV) structure and function, and these benefits remain stable during long-term follow-up. Although the mechanisms remain under investigation, effects of secreted factors, regeneration of cellular constituents, and stimulation of endogenous stem/progenitors may play combinatorial roles. The purpose of this review is to summarize the current evidence regarding the biologic features of VSELs, and to discuss their potential as cellular substrates for therapeutic cardiac repair.
Collapse
Affiliation(s)
- Ewa K Zuba-Surma
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | | | | | | |
Collapse
|
38
|
|
39
|
Nam H, Kim J, Park J, Park JC, Kim JW, Seo BM, Lee JC, Lee G. Expression profile of the stem cell markers in human Hertwig's epithelial root sheath/Epithelial rests of Malassez cells. Mol Cells 2011; 31:355-60. [PMID: 21359676 PMCID: PMC3933961 DOI: 10.1007/s10059-011-0045-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 01/14/2011] [Accepted: 01/14/2011] [Indexed: 12/12/2022] Open
Abstract
Hertwig's epithelial root sheath/Epithelial rests of Malassez (HERS/ERM) cells are unique epithelial cells in the periodontal ligament. They remain in periodontal tissues through-out the adult life, and it is expected that their functional role is to maintain the homeostasis of the periodontium through reciprocal interactions with other periodontal cells. In this study, we investigated whether HERS/ERM cells have primitive stem cell characteristics: those of embryonic stem cells as well as of epithelial stem cells. Primary HERS/ERM cells had typical epithelial cell morphology and characteristics and they maintained for more than five passages. They expressed epithelial stem cell-related genes: ABCG2, ANp63, p75, EpCAM, and Bmi-1. Moreover, the expression of embryonic stem cell markers such as Oct-4, Nanog, and SSEA-4 were detected. Next, we investigated whether the expression of these stem cell markers was maintained during the sub-culture process. HERS/ERM cells showed different expression levels of these stemness genes at each passage, but their expression was maintained throughout the passages. Taken together, our data suggest that a primary culture of HERS/ERM cells contains a population of primitive stem cells that express epithelial stem cell markers and embryonic stem cell markers. Furthermore, these cell populations were maintained during the sub-culturing process in our culture conditions. Therefore, our findings suggest that there is a strong possibility of accomplishing cementum tissue engineering with HERS/ERM cells.
Collapse
Affiliation(s)
| | | | | | - Joo-Cheol Park
- Department of Oral Histology-Developmental Biology, School of Dentistry, Seoul National University, Seoul 110-749, Korea
| | - Jung-Wook Kim
- Department of Pediatric Dentistry, School of Dentistry, Seoul National University, Seoul 110-749, Korea
| | - Byoung-Moo Seo
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul 110-749, Korea
| | - Jae Cheoun Lee
- Children’s Dental Center and CDC Baby Tooth Stem Cell Bank, Seoul 135-953, Korea
| | | |
Collapse
|
40
|
Parte S, Bhartiya D, Telang J, Daithankar V, Salvi V, Zaveri K, Hinduja I. Detection, characterization, and spontaneous differentiation in vitro of very small embryonic-like putative stem cells in adult mammalian ovary. Stem Cells Dev 2011; 20:1451-64. [PMID: 21291304 PMCID: PMC3148829 DOI: 10.1089/scd.2010.0461] [Citation(s) in RCA: 182] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The present study was undertaken to detect, characterize, and study differentiation potential of stem cells in adult rabbit, sheep, monkey, and menopausal human ovarian surface epithelium (OSE). Two distinct populations of putative stem cells (PSCs) of variable size were detected in scraped OSE, one being smaller and other similar in size to the surrounding red blood cells in the scraped OSE. The smaller 1-3 μm very small embryonic-like PSCs were pluripotent in nature with nuclear Oct-4 and cell surface SSEA-4, whereas the bigger 4-7 μm cells with cytoplasmic localization of Oct-4 and minimal expression of SSEA-4 were possibly the tissue committed progenitor stem cells. Pluripotent gene transcripts of Oct-4, Oct-4A, Nanog, Sox-2, TERT, and Stat-3 in human and sheep OSE were detected by reverse transcriptase-polymerase chain reaction. The PSCs underwent spontaneous differentiation into oocyte-like structures, parthenote-like structures, embryoid body-like structures, cells with neuronal-like phenotype, and embryonic stem cell-like colonies, whereas the epithelial cells transformed into mesenchymal phenotype by epithelial-mesenchymal transition in 3 weeks of OSE culture. Germ cell markers like c-Kit, DAZL, GDF-9, VASA, and ZP4 were immuno-localized in oocyte-like structures. In conclusion, as opposed to the existing view of OSE being a bipotent source of oocytes and granulosa cells, mammalian ovaries harbor distinct very small embryonic-like PSCs and tissue committed progenitor stem cells population that have the potential to develop into oocyte-like structures in vitro, whereas mesenchymal fibroblasts appear to form supporting granulosa-like somatic cells. Research at the single-cell level, including complete gene expression profiling, is required to further confirm whether postnatal oogenesis is a conserved phenomenon in adult mammals.
Collapse
Affiliation(s)
- Seema Parte
- Department of Stem Cell Biology, National Institute for Research in Reproductive Health, Mumbai, India
| | | | | | | | | | | | | |
Collapse
|
41
|
Identification of Very Small Embryonic/Epiblast-Like Stem Cells (VSELs) Circulating in Peripheral Blood During Organ/Tissue Injuries. Methods Cell Biol 2011; 103:31-54. [DOI: 10.1016/b978-0-12-385493-3.00003-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
42
|
Fernández-Medarde A, Santos E. The RasGrf family of mammalian guanine nucleotide exchange factors. Biochim Biophys Acta Rev Cancer 2010; 1815:170-88. [PMID: 21111786 DOI: 10.1016/j.bbcan.2010.11.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 11/14/2010] [Indexed: 12/31/2022]
Abstract
RasGrf1 and RasGrf2 are highly homologous mammalian guanine nucleotide exchange factors which are able to activate specific Ras or Rho GTPases. The RasGrf genes are preferentially expressed in the central nervous system, although specific expression of either locus may also occur elsewhere. RasGrf1 is a paternally-expressed, imprinted gene that is expressed only after birth. In contrast, RasGrf2 is not imprinted and shows a wider expression pattern. A variety of isoforms for both genes are also detectable in different cellular contexts. The RasGrf proteins exhibit modular structures composed by multiple domains including CDC25H and DHPH motifs responsible for promoting GDP/GTP exchange, respectively, on Ras or Rho GTPase targets. The various domains are essential to define their intrinsic exchanger activity and to modulate the specificity of their functional activity so as to connect different upstream signals to various downstream targets and cellular responses. Despite their homology, RasGrf1 and RasGrf2 display differing target specificities and non overlapping functional roles in a variety of signaling contexts related to cell growth and differentiation as well as neuronal excitability and response or synaptic plasticity. Whereas both RasGrfs are activatable by glutamate receptors, G-protein-coupled receptors or changes in intracellular calcium concentration, only RasGrf1 is reported to be activated by LPA, cAMP, or agonist-activated Trk and cannabinoid receptors. Analysis of various knockout mice strains has uncovered a specific functional contribution of RasGrf1 in processes of memory and learning, photoreception, control of post-natal growth and body size and pancreatic β-cell function and glucose homeostasis. For RasGrf2, specific roles in lymphocyte proliferation, T-cell signaling responses and lymphomagenesis have been described.
Collapse
|