1
|
Martis ASA, Soundararajan L, Shetty P, Moin S, Vanje T, Jai Sankar Y, Parveen S. Chromosome number alterations cause apoptosis and cellular hypertrophy in induced pluripotent stem cell models of embryonic epiblast cells. Biol Open 2025; 14:BIO061814. [PMID: 39851179 PMCID: PMC11789280 DOI: 10.1242/bio.061814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 01/26/2025] Open
Abstract
Chromosomal aneuploidies are a major cause of developmental failure and pregnancy loss. To investigate the possible consequences of aneuploidy on early embryonic development in vitro, we focused on primed pluripotent stem cells that are relatable to the epiblast of post-implantation embryos in vivo. We used human induced pluripotent stem cells (iPSCs) as an epiblast model and altered chromosome numbers by treating with reversine, a small-molecule inhibitor of monopolar spindle 1 kinase (MSP1) that inactivates the spindle assembly checkpoint, which has been strongly implicated in chromosome mis-segregation and aneuploidy generation. Upon reversine treatment, we obtained cells with varied chromosomal content that retained pluripotency and potential to differentiate into cells of three germ lineages. However, these cells displayed lagging chromosomes, increased micronuclei content, high p53 expression and excessive apoptotic activity. Cell proliferation was not affected. Prolonged in vitro culture of these cells resulted in a selective pool of cells with supernumerary chromosomes, which exhibited cellular hypertrophy, enlarged nuclei, and overproduction of total RNAs and proteins. We conclude that increased DNA damage responses, apoptosis, and improper cellular mass and functions are possible mechanisms that contribute to abnormal epiblast development.
Collapse
Affiliation(s)
- Althea Stella Anil Martis
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| | - Loshini Soundararajan
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| | - Pallavi Shetty
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| | - Syed Moin
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| | - Tejashree Vanje
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| | - Yogeshwaran Jai Sankar
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| | - Shagufta Parveen
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
2
|
Bou Malhab LJ, Harb AA, Eldohaji L, Taneera J, Al‐Hroub HM, Abuhelwa A, Alzoubi KH, Abu‐Irmaileh B, Hudaib M, Almaliti J, Abdel‐Rahman WM, Shanableh A, Semreen MH, El‐Huneidi W, Abu‐Gharbieh E, Bustanji Y. Exploring the Anticancer Effect of Artemisia herba-alba on Colorectal Cancer: Insights From Eight Colorectal Cancer Cell Lines. Food Sci Nutr 2025; 13:e4715. [PMID: 39803277 PMCID: PMC11717013 DOI: 10.1002/fsn3.4715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/30/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Colorectal cancer (CRC) is a prevalent and deadly disease, necessitating the exploration of novel therapeutic strategies. Traditional chemotherapy often encounters drug resistance and adverse side effects, highlighting the need for alternative approaches. Artemisia herba-alba, a plant rich in phytochemical constituents, was investigated for its potential as an anticancer agent against colorectal cancer (CRC). The primary objective of this study was to investigate the cytotoxic effects of the methanolic extract of A. herba-alba on eight CRC cell lines including: Caco-2, DLD1, RKO+/+p53, RKO-/-p53, HCT+/+p53, HCT-/-p53, SW620, and SW480. Specifically, the study investigated the extract's impact on cell viability, apoptosis, cell cycle progression, and effects on the PI3K/AKT/mTOR signaling pathway. Chemical derivatization and Gas Chromatography-Mass Spectrometry (GC-MS) analysis revealed a diverse array of bioactive compounds, including ephedrine, hydroxyflavone, quinolinic acid, 4-hydroxybenzoic acid, borneol, β-eudesmol, and camphor, known for their cytotoxic properties. The methanolic extract of A. herba-alba exhibited varying degrees of cytotoxicity across a panel of CRC cell lines, with IC50 values indicating differential sensitivity. The extract triggered apoptosis in many cell lines, irrespective of p53 status. Importantly, A. herba-alba extract caused G2-M phase cell cycle arrest in CRC cells, accompanied by a decrease in Cyclin B1 and CDK1 expression. Furthermore, the extract demonstrated an inhibitory effect on the PI3K/AKT/mTOR pathway, crucial in cancer progression. These findings highlight the promising anticancer potential of Artemisia herba-alba as a valuable resource for innovative CRC treatments. Further research is warranted to elucidate its specific anticancer characteristics and explore its potential incorporation into future cancer therapy approaches.
Collapse
Affiliation(s)
| | - Amani A. Harb
- Department of Basic Sciences, Faculty of Arts and SciencesAl‐Ahliyya Amman UniversityAmmanJordan
| | - Leen Eldohaji
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
| | - Jalal Taneera
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of MedicineUniversity of SharjahSharjahUAE
| | - Hamza M. Al‐Hroub
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
| | - Ahmad Abuhelwa
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of PharmacyUniversity of SharjahSharjahUAE
| | - Karem H. Alzoubi
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of PharmacyUniversity of SharjahSharjahUAE
| | | | | | - Jehad Almaliti
- School of PharmacyThe University of JordanAmmanJordan
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Wael M. Abdel‐Rahman
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- Department of Medical Laboratory Sciences, College of Health SciencesUniversity of SharjahSharjahUAE
| | - Abdallah Shanableh
- Research Institute of Science and Engineering (RISE)University of SharjahSharjahUAE
| | - Mohammad H. Semreen
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of PharmacyUniversity of SharjahSharjahUAE
| | - Waseem El‐Huneidi
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of MedicineUniversity of SharjahSharjahUAE
| | - Eman Abu‐Gharbieh
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of MedicineUniversity of SharjahSharjahUAE
- School of PharmacyThe University of JordanAmmanJordan
| | - Yasser Bustanji
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of MedicineUniversity of SharjahSharjahUAE
- School of PharmacyThe University of JordanAmmanJordan
| |
Collapse
|
3
|
Kwon EJ, Lee H, Shin U, Kim ES, Myung K, Kim J, Park JH, Kim K, Lee Y, Oh CK, Kim YH. Ionizing radiation inhibits zebrafish embryo hatching through induction of tissue inhibitors of metalloproteinases (TIMPs) expression. FEBS J 2024; 291:5470-5485. [PMID: 39547957 DOI: 10.1111/febs.17318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/26/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
Ionizing radiation (IR) has garnered growing attention because of its biological effects on aquatic organisms and humans. Here, we identify the most impacted organs and uncover the molecular mechanisms causing the changes in the context of vertebrate development using single-cell RNA sequencing. Alterations in cellular composition and biological functions were explored using transcriptomic profiling of zebrafish embryos exposed to 5 Gy. Single-cell RNA sequencing analyses unveiled notable shifts in the proportions of brain/central nervous system and hatching gland clusters. Although IR exposure led to increased expression of hatching enzymes, a significant but mild delay in hatching was observed following 5 Gy IR exposure. Gene Ontology analysis showed an increased expression of tissue inhibitors of metalloproteinases (TIMPs), known as matrix metalloproteinase inhibitors, which was confirmed via whole-mount in situ hybridization. Correlation analysis linked TIMPs to transcription factors cebpb and cebpd, which were significantly correlated post-IR exposure. Although no morphological changes were observed in some organs, including the brain, the study reveals substantial alterations in developing vertebrates. Notably, despite increased hatching enzymes, elevated TIMPs in the hatching gland suggest a regulatory mechanism impacting hatching activity. This research contributes to comprehending the ecological repercussions of IR exposure, emphasizing the importance of safety measures for aquatic ecosystems and overall environmental health.
Collapse
Affiliation(s)
- Eun Jung Kwon
- Medical Research Institute, Pusan National University, Yangsan, Korea
| | - Hansong Lee
- Medical Research Institute, Pusan National University, Yangsan, Korea
| | - Unbum Shin
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Korea
| | - Eun-Sun Kim
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Korea
| | - Jeongmo Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Korea
| | - Jung-Hoon Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Korea
| | - Kihun Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Korea
| | - Yoonsung Lee
- Research Institute of Clinical Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Chang-Kyu Oh
- Department of Biochemistry, School of Medicine, Pusan National University, Yangsan, Korea
- Institute for Future Earth, Pusan National University, Busan, Korea
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Korea
| |
Collapse
|
4
|
Vallmajo-Martin Q, Ma Z, Srinivasan S, Murali D, Dravis C, Mukund K, Subramaniam S, Wahl GM, Lytle NK. The molecular chronology of mammary epithelial cell fate switching. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617155. [PMID: 39415993 PMCID: PMC11482796 DOI: 10.1101/2024.10.08.617155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The adult mammary gland is maintained by lineage-restricted progenitor cells through pregnancy, lactation, involution, and menopause. Injury resolution, transplantation-associated mammary gland reconstitution, and tumorigenesis are unique exceptions, wherein mammary basal cells gain the ability to reprogram to a luminal state. Here, we leverage newly developed cell-identity reporter mouse strains, and time-resolved single-cell epigenetic and transcriptomic analyses to decipher the molecular programs underlying basal-to-luminal fate switching in vivo. We demonstrate that basal cells rapidly reprogram toward plastic cycling intermediates that appear to hijack molecular programs we find in bipotent fetal mammary stem cells and puberty-associatiated cap cells. Loss of basal-cell specifiers early in dedifferentiation coincides with activation of Notch and BMP, among others. Pharmacologic blockade of each pathway disrupts basal-to-luminal transdifferentiation. Our studies provide a comprehensive map and resource for understanding the coordinated molecular changes enabling terminally differentiated epithelial cells to transition between cell lineages and highlights the stunning rapidity by which epigenetic reprogramming can occur in response to disruption of tissue structure.
Collapse
Affiliation(s)
- Queralt Vallmajo-Martin
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- These authors contributed equally
| | - Zhibo Ma
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- These authors contributed equally
| | - Sumana Srinivasan
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- These authors contributed equally
| | - Divya Murali
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- These authors contributed equally
| | - Christopher Dravis
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Kavitha Mukund
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- San Diego Supercomputer Center, University of California, San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Geoffrey M. Wahl
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Nikki K. Lytle
- Department of Surgery, Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
- These authors contributed equally
| |
Collapse
|
5
|
Zhang Y, Zheng Y, Zhang J, Xu C, Wu J. Apoptotic signaling pathways in bone metastatic lung cancer: a comprehensive analysis. Discov Oncol 2024; 15:310. [PMID: 39060849 PMCID: PMC11282049 DOI: 10.1007/s12672-024-01151-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
This review provides a comprehensive analysis of apoptotic signaling pathways in the context of bone metastatic lung cancer, emphasizing the intricate molecular mechanisms and microenvironmental influences. Beginning with an overview of apoptosis in cancer, the paper explores the specific molecular characteristics of bone metastatic lung cancer, highlighting alterations in apoptotic pathways. Focused discussions delve into key apoptotic signaling pathways, including the intrinsic and extrinsic pathways, and the roles of critical molecular players such as Bcl-2 family proteins and caspases. Microenvironmental factors, such as the tumor microenvironment, extracellular matrix interactions, and immune cell involvement, are examined in depth. The review also addresses experimental approaches and techniques employed in studying apoptotic signaling, paving the way for a discussion on current therapeutic strategies, their limitations, and future prospects. This synthesis contributes a holistic understanding of apoptosis in bone metastatic lung cancer, offering insights for potential therapeutic advancements.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Orthopedic Surgery, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Yi Zheng
- Department of Orthopedic Surgery, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Jiakai Zhang
- Department of Orthopedic Surgery, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Chaoyang Xu
- Hangzhou Medical College, Hangzhou, 310053, Zhejiang, China
| | - Junlong Wu
- Department of Orthopedic Surgery, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|
6
|
Roy A, Chakraborty AR, DePamphilis ML. PIKFYVE inhibitors trigger interleukin-24-dependent cell death of autophagy-dependent melanoma. Mol Oncol 2024; 18:988-1011. [PMID: 38414326 PMCID: PMC10994231 DOI: 10.1002/1878-0261.13607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/16/2024] [Accepted: 02/02/2024] [Indexed: 02/29/2024] Open
Abstract
Inhibitors specifically targeting the 1-phosphatidylinositol 3-phosphate 5-kinase (PIKFYVE) disrupt lysosome homeostasis, thereby selectively terminating autophagy-dependent human cancer cells in vivo as well as in vitro without harming the viability of nonmalignant cells. To elucidate the mechanism by which PIKFYVE inhibition induces cell death, autophagy-dependent melanoma cells were compared with normal foreskin fibroblasts. RNA sequence profiling suggested that PIKFYVE inhibitors upregulated an endoplasmic reticulum (ER) stress response involving interleukin-24 (IL24; also known as MDA7) selectively in melanoma cells. Subsequent biochemical and genetic analyses confirmed these results and extended them to tumor xenografts in which tumor formation and expansion were inhibited. IL24 expression was upregulated by the DDIT3/CHOP/CEBPz transcription factor, a component of the PERK-dependent ER-stress response. Ectopic expression of IL24-induced cell death in melanoma cells, but not in foreskin fibroblasts, whereas ablation of the IL24 gene in melanoma cells prevented death. IL24 upregulation was triggered specifically by PIKFYVE inhibition. Thus, unlike thapsigargin and tunicamycin, which induce ER-stress indiscriminately, PIKFYVE inhibitors selectively terminated PIKFYVE-sensitive melanoma by inducing IL24-dependent ER-stress. Moreover, induction of cell death by a PIKFYVE inhibitor together with ectopic expression of IL24 protein was cumulative, thereby confirming the therapeutic potential of PIKFYVE inhibitors in the treatment of melanoma.
Collapse
Affiliation(s)
- Ajit Roy
- National Institute of Child Health & Human DevelopmentNational Institutes of HealthBethesdaMDUSA
| | - Arup R. Chakraborty
- National Institute of Child Health & Human DevelopmentNational Institutes of HealthBethesdaMDUSA
| | - Melvin L. DePamphilis
- National Institute of Child Health & Human DevelopmentNational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
7
|
Valverde-Lopez JA, Li-Bao L, Sierra R, Santos E, Giovinazzo G, Díaz-Díaz C, Torres M. P53 and BCL-2 family proteins PUMA and NOXA define competitive fitness in pluripotent cell competition. PLoS Genet 2024; 20:e1011193. [PMID: 38489392 PMCID: PMC10971546 DOI: 10.1371/journal.pgen.1011193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 03/27/2024] [Accepted: 02/22/2024] [Indexed: 03/17/2024] Open
Abstract
Cell Competition is a process by which neighboring cells compare their fitness. As a result, viable but suboptimal cells are selectively eliminated in the presence of fitter cells. In the early mammalian embryo, epiblast pluripotent cells undergo extensive Cell Competition, which prevents suboptimal cells from contributing to the newly forming organism. While competitive ability is regulated by MYC in the epiblast, the mechanisms that contribute to competitive fitness in this context are largely unknown. Here, we report that P53 and its pro-apoptotic targets PUMA and NOXA regulate apoptosis susceptibility and competitive fitness in pluripotent cells. PUMA is widely expressed specifically in pluripotent cells in vitro and in vivo. We found that P53 regulates MYC levels in pluripotent cells, which connects these two Cell Competition pathways, however, MYC and PUMA/NOXA levels are independently regulated by P53. We propose a model that integrates a bifurcated P53 pathway regulating both MYC and PUMA/NOXA levels and determines competitive fitness.
Collapse
Affiliation(s)
- Jose A Valverde-Lopez
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Lin Li-Bao
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Rocío Sierra
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Elisa Santos
- Pluripotent Cell Technology Unit, Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain
| | - Giovanna Giovinazzo
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Pluripotent Cell Technology Unit, Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain
| | - Covadonga Díaz-Díaz
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Miguel Torres
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
8
|
Saini P, Anugula S, Fong YW. The Role of ATP-Binding Cassette Proteins in Stem Cell Pluripotency. Biomedicines 2023; 11:1868. [PMID: 37509507 PMCID: PMC10377311 DOI: 10.3390/biomedicines11071868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Pluripotent stem cells (PSCs) are highly proliferative cells that can self-renew indefinitely in vitro. Upon receiving appropriate signals, PSCs undergo differentiation and can generate every cell type in the body. These unique properties of PSCs require specific gene expression patterns that define stem cell identity and dynamic regulation of intracellular metabolism to support cell growth and cell fate transitions. PSCs are prone to DNA damage due to elevated replicative and transcriptional stress. Therefore, mechanisms to prevent deleterious mutations in PSCs that compromise stem cell function or increase the risk of tumor formation from becoming amplified and propagated to progenitor cells are essential for embryonic development and for using PSCs including induced PSCs (iPSCs) as a cell source for regenerative medicine. In this review, we discuss the role of the ATP-binding cassette (ABC) superfamily in maintaining PSC homeostasis, and propose how their activities can influence cellular signaling and stem cell fate decisions. Finally, we highlight recent discoveries that not all ABC family members perform only canonical metabolite and peptide transport functions in PSCs; rather, they can participate in diverse cellular processes from genome surveillance to gene transcription and mRNA translation, which are likely to maintain the pristine state of PSCs.
Collapse
Affiliation(s)
- Prince Saini
- Brigham Regenerative Medicine Center, Brigham and Women’s Hospital, Boston, MA 02115, USA; (P.S.); (S.A.)
- Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Sharath Anugula
- Brigham Regenerative Medicine Center, Brigham and Women’s Hospital, Boston, MA 02115, USA; (P.S.); (S.A.)
- Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Yick W. Fong
- Brigham Regenerative Medicine Center, Brigham and Women’s Hospital, Boston, MA 02115, USA; (P.S.); (S.A.)
- Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
9
|
Tichy ED. Specialized Circuitry of Embryonic Stem Cells Promotes Genomic Integrity. Crit Rev Oncog 2023; 27:1-15. [PMID: 36734869 DOI: 10.1615/critrevoncog.2022042332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Embryonic stem cells (ESCs) give rise to all cell types of the organism. Given the importance of these cells in this process, ESCs must employ robust mechanisms to protect genomic integrity or risk catastrophic propagation of mutations throughout the organism. Should such an event occur in daughter cells that will eventually contribute to the germline, the overall species health could dramatically decline. This review describes several key mechanisms employed by ESCs that are unique to these cells, in order to maintain their genomic integrity. Additionally, the contributions of cell cycle regulators in modulating ESC differentiation, after DNA damage exposure, are also examined. Where data are available, findings reported in ESCs are extended to include observations described in induced pluripotent stem cells (IPSCs).
Collapse
Affiliation(s)
- Elisia D Tichy
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, 371 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104-6081
| |
Collapse
|
10
|
Transcriptome Analysis in High Temperature Inhibiting Spermatogonial Stem Cell Differentiation In Vitro. Reprod Sci 2022; 30:1938-1951. [DOI: 10.1007/s43032-022-01133-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022]
|
11
|
Ayaz G, Yan H, Malik N, Huang J. An Updated View of the Roles of p53 in Embryonic Stem Cells. Stem Cells 2022; 40:883-891. [PMID: 35904997 PMCID: PMC9585900 DOI: 10.1093/stmcls/sxac051] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/14/2022] [Indexed: 11/12/2022]
Abstract
The TP53 gene is unarguably one of the most studied human genes. Its encoded protein, p53, is a tumor suppressor and is often called the "guardian of the genome" due to its pivotal role in maintaining genome stability. Historically, most studies of p53 have focused on its roles in somatic cells and tissues, but in the last two decades, its functions in embryonic stem cells (ESCs) and induced pluripotent stem cells have attracted increasing attention. Recent studies have identified p53 as a critical regulator of pluripotency, self-renewal, differentiation, proliferation, and genome stability in mouse and human embryonic stem cells. In this article, we systematically review the studies on the functions of p53 in ESCs, provide an updated overview, attempt to reconcile controversial results described in the literature, and discuss the relevance of these cellular functions of p53 to its roles in tumor suppression.
Collapse
Affiliation(s)
- Gamze Ayaz
- Cancer and Stem Cell Epigenetics, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hualong Yan
- Cancer and Stem Cell Epigenetics, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Navdeep Malik
- Cancer and Stem Cell Epigenetics, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jing Huang
- Cancer and Stem Cell Epigenetics, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
12
|
Laka K, Mbita Z. P53-Related Anticancer Activities of Drimia calcarata Bulb Extracts Against Lung Cancer. Front Mol Biosci 2022; 9:876213. [PMID: 35769912 PMCID: PMC9235921 DOI: 10.3389/fmolb.2022.876213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/02/2022] [Indexed: 01/18/2023] Open
Abstract
Current lung cancer treatment strategies are ineffective, and lung cancer cases continue to soar; thus, novel anticancer drugs and targets are needed, and medicinal plants are promising to offer better alternatives. This study was aimed at analysing two p53 splice variants during the potential anticancer activities of Drimia calcarata (Dc) methanol and water extracts against different human lung cancer cell lines of varying p53 mutation status, and these included mutant H1573 and mutant H1437 and p53-wild type (A549) cells. The anticancer activities of the Dc extracts were assessed by establishing the cytotoxic effect and the apoptosis-inducing capacity of these extracts, using the MTT assay and Annexin V analysis, respectively, with the latter confirmed using fluorescence microscopy. The molecular mechanisms induced by these extracts were further evaluated using cell cycle analysis and RT-PCR. Both extracts demonstrated safety against noncancerous lung MRC-5 fibroblasts and exhibited significant anticancer potency (p < 0.001) against the H1437 (IC50 values: 62.50 μg/ml methanol extract and 125 μg/ml WE), H1573 (IC50 value: 125 μg/ml for both extracts) and A549 (IC50 value: 500 μg/ml ME). The water extract had no effect on the viability of A549 cells. Treated H1437 cells underwent p53-dependent apoptosis and S-phase cell cycle arrest while H1573 treated cells underwent p53-independed apoptosis and G0/G1 cell cycle arrest through upregulation of p21 mRNA expression levels. The expression levels of STAT1, STAT3, STAT5A and STAT5B genes increased significantly (p < 0.001) following the treatment of H1573 cells with ME and WE. Treatment of H1437 cells with ME upregulated the STAT1, STAT3, STAT5A and STAT5B mRNAs. Our results indicate that the proliferative inhibitory effect of D. calcarata extracts on A549 and H1573 cells is correlated with the suppression of Bcl-2, STAT3 and STAT5B while that is not the case in H1437 cells. Thus, our results suggest that the dysregulation of anti-apoptotic molecules Bcl-2, STAT3, STAT5A and STAT5B in H1437 may play a role in cancer cell survival, which may consequently contribute to the development of p53-mutated non-small human lung cancer. Our results indicate that D. calcarata is a promising source of anticancer agents for the treatment of p53-mutant human non-small lung cancer cells than the p53-wild type human non-small lung cancer cells.
Collapse
|
13
|
Raj S, Jaiswal SK, DePamphilis ML. Cell Death and the p53 Enigma During Mammalian Embryonic Development. Stem Cells 2022; 40:227-238. [PMID: 35304609 PMCID: PMC9199838 DOI: 10.1093/stmcls/sxac003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/20/2021] [Indexed: 01/30/2023]
Abstract
Twelve forms of programmed cell death (PCD) have been described in mammalian cells, but which of them occurs during embryonic development and the role played by the p53 transcription factor and tumor suppressor remains enigmatic. Although p53 is not required for mouse embryonic development, some studies conclude that PCD in pluripotent embryonic stem cells from mice (mESCs) or humans (hESCs) is p53-dependent whereas others conclude that it is not. Given the importance of pluripotent stem cells as models of embryonic development and their applications in regenerative medicine, resolving this enigma is essential. This review reconciles contradictory results based on the facts that p53 cannot induce lethality in mice until gastrulation and that experimental conditions could account for differences in results with ESCs. Consequently, activation of the G2-checkpoint in mouse ESCs is p53-independent and generally, if not always, results in noncanonical apoptosis. Once initiated, PCD occurs at equivalent rates and to equivalent extents regardless of the presence or absence of p53. However, depending on experimental conditions, p53 can accelerate initiation of PCD in ESCs and late-stage blastocysts. In contrast, DNA damage following differentiation of ESCs in vitro or formation of embryonic fibroblasts in vivo induces p53-dependent cell cycle arrest and senescence.
Collapse
Affiliation(s)
- Sonam Raj
- National Cancer Institute, Bethesda, MD 20892, USA
| | - Sushil K Jaiswal
- National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Melvin L DePamphilis
- National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| |
Collapse
|
14
|
Chakraborty AR, Vassilev A, Jaiswal SK, O'Connell CE, Ahrens JF, Mallon BS, Pera MF, DePamphilis ML. Selective elimination of pluripotent stem cells by PIKfyve specific inhibitors. Stem Cell Reports 2022; 17:397-412. [PMID: 35063131 PMCID: PMC8828683 DOI: 10.1016/j.stemcr.2021.12.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 01/05/2023] Open
Abstract
Inhibition of PIKfyve phosphoinositide kinase selectively kills autophagy-dependent cancer cells by disrupting lysosome homeostasis. Here, we show that PIKfyve inhibitors can also selectively eliminate pluripotent embryonal carcinoma cells (ECCs), embryonic stem cells, and induced pluripotent stem cells under conditions where differentiated cells remain viable. PIKfyve inhibitors prevented lysosome fission, induced autophagosome accumulation, and reduced cell proliferation in both pluripotent and differentiated cells, but they induced death only in pluripotent cells. The ability of PIKfyve inhibitors to distinguish between pluripotent and differentiated cells was confirmed with xenografts derived from ECCs. Pretreatment of ECCs with the PIKfyve specific inhibitor WX8 suppressed their ability to form teratocarcinomas in mice, and intraperitoneal injections of WX8 into mice harboring teratocarcinoma xenografts selectively eliminated pluripotent cells. Differentiated cells continued to proliferate, but at a reduced rate. These results provide a proof of principle that PIKfyve specific inhibitors can selectively eliminate pluripotent stem cells in vivo as well as in vitro.
Collapse
Affiliation(s)
- Arup R Chakraborty
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - Alex Vassilev
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - Sushil K Jaiswal
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - Constandina E O'Connell
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - John F Ahrens
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - Barbara S Mallon
- NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Melvin L DePamphilis
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA.
| |
Collapse
|
15
|
Noncanonical roles of p53 in cancer stemness and their implications in sarcomas. Cancer Lett 2022; 525:131-145. [PMID: 34742870 DOI: 10.1016/j.canlet.2021.10.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 09/24/2021] [Accepted: 10/25/2021] [Indexed: 12/25/2022]
Abstract
Impairment of the prominent tumor suppressor p53, well known for its canonical role as the "guardian of the genome", is found in almost half of human cancers. More recently, p53 has been suggested to be a crucial regulator of stemness, orchestrating the differentiation of embryonal and adult stem cells, suppressing reprogramming into induced pluripotent stem cells, or inhibiting cancer stemness (i.e., cancer stem cells, CSCs), which underlies the development of therapy-resistant tumors. This review addresses these noncanonical roles of p53 and their implications in sarcoma initiation and progression. Indeed, dysregulation of p53 family proteins is a common event in sarcomas and is associated with poor survival. Additionally, emerging studies have demonstrated that loss of wild-type p53 activity hinders the terminal differentiation of mesenchymal stem cells and leads to the development of aggressive sarcomas. This review summarizes recent findings on the roles of aberrant p53 in sarcoma development and stemness and further describes therapeutic approaches to restore normal p53 activity as a promising anti-CSC strategy to treat refractory sarcomas.
Collapse
|
16
|
Jaiswal SK, Raj S, DePamphilis ML. Developmental Acquisition of p53 Functions. Genes (Basel) 2021; 12:genes12111675. [PMID: 34828285 PMCID: PMC8622856 DOI: 10.3390/genes12111675] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/14/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Remarkably, the p53 transcription factor, referred to as “the guardian of the genome”, is not essential for mammalian development. Moreover, efforts to identify p53-dependent developmental events have produced contradictory conclusions. Given the importance of pluripotent stem cells as models of mammalian development, and their applications in regenerative medicine and disease, resolving these conflicts is essential. Here we attempt to reconcile disparate data into justifiable conclusions predicated on reports that p53-dependent transcription is first detected in late mouse blastocysts, that p53 activity first becomes potentially lethal during gastrulation, and that apoptosis does not depend on p53. Furthermore, p53 does not regulate expression of genes required for pluripotency in embryonic stem cells (ESCs); it contributes to ESC genomic stability and differentiation. Depending on conditions, p53 accelerates initiation of apoptosis in ESCs in response to DNA damage, but cell cycle arrest as well as the rate and extent of apoptosis in ESCs are p53-independent. In embryonic fibroblasts, p53 induces cell cycle arrest to allow repair of DNA damage, and cell senescence to prevent proliferation of cells with extensive damage.
Collapse
Affiliation(s)
- Sushil K. Jaiswal
- National Institute of Child Health and Human Development, Bethesda, MD 20892, USA;
- National Human Genome Research Institute, Bethesda, MD 20892, USA
| | - Sonam Raj
- National Cancer Institute, Bethesda, MD 20892, USA;
| | - Melvin L. DePamphilis
- National Institute of Child Health and Human Development, Bethesda, MD 20892, USA;
- Correspondence:
| |
Collapse
|
17
|
Atkinson SP. A preview of selected articles. STEM CELLS (DAYTON, OHIO) 2021; 38:1051-1054. [PMID: 32853480 DOI: 10.1002/stem.3262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 11/09/2022]
|
18
|
Vessoni AT, Zhang T, Quinet A, Jeong HC, Munroe M, Wood M, Tedone E, Vindigni A, Shay JW, Greenberg RA, Batista LF. Telomere erosion in human pluripotent stem cells leads to ATR-mediated mitotic catastrophe. J Cell Biol 2021; 220:211982. [PMID: 33851958 PMCID: PMC8050844 DOI: 10.1083/jcb.202011014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/03/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022] Open
Abstract
It is well established that short telomeres activate an ATM-driven DNA damage response that leads to senescence in terminally differentiated cells. However, technical limitations have hampered our understanding of how telomere shortening is signaled in human stem cells. Here, we show that telomere attrition induces ssDNA accumulation (G-strand) at telomeres in human pluripotent stem cells (hPSCs), but not in their differentiated progeny. This led to a unique role for ATR in the response of hPSCs to telomere shortening that culminated in an extended S/G2 cell cycle phase and a longer period of mitosis, which was associated with aneuploidy and mitotic catastrophe. Loss of p53 increased resistance to death, at the expense of increased mitotic abnormalities in hPSCs. Taken together, our data reveal an unexpected dominant role of ATR in hPSCs, combined with unique cell cycle abnormalities and, ultimately, consequences distinct from those observed in their isogenic differentiated counterparts.
Collapse
Affiliation(s)
| | - Tianpeng Zhang
- Department of Cancer Biology, Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Annabel Quinet
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Ho-Chang Jeong
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Michael Munroe
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Matthew Wood
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Enzo Tedone
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX
| | | | - Jerry W. Shay
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX
| | - Roger A. Greenberg
- Department of Cancer Biology, Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Luis F.Z. Batista
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO
- Correspondence to Luis F.Z. Batista:
| |
Collapse
|
19
|
Toro A, Anselmino N, Solari C, Francia M, Oses C, Sanchis P, Bizzotto J, Vazquez Echegaray C, Petrone MV, Levi V, Vazquez E, Guberman A. Novel Interplay between p53 and HO-1 in Embryonic Stem Cells. Cells 2020; 10:cells10010035. [PMID: 33383653 PMCID: PMC7823265 DOI: 10.3390/cells10010035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 02/06/2023] Open
Abstract
Stem cells genome safeguarding requires strict oxidative stress control. Heme oxygenase-1 (HO-1) and p53 are relevant components of the cellular defense system. p53 controls cellular response to multiple types of harmful stimulus, including oxidative stress. Otherwise, besides having a protective role, HO-1 is also involved in embryo development and in embryonic stem (ES) cells differentiation. Although both proteins have been extensively studied, little is known about their relationship in stem cells. The aim of this work is to explore HO-1-p53 interplay in ES cells. We studied HO-1 expression in p53 knockout (KO) ES cells and we found that they have higher HO-1 protein levels but similar HO-1 mRNA levels than the wild type (WT) ES cell line. Furthermore, cycloheximide treatment increased HO-1 abundance in p53 KO cells suggesting that p53 modulates HO-1 protein stability. Notably, H2O2 treatment did not induce HO-1 expression in p53 KO ES cells. Finally, SOD2 protein levels are also increased while Sod2 transcripts are not in KO cells, further suggesting that the p53 null phenotype is associated with a reinforcement of the antioxidant machinery. Our results demonstrate the existence of a connection between p53 and HO-1 in ES cells, highlighting the relationship between these stress defense pathways.
Collapse
Affiliation(s)
- Ayelén Toro
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Nicolás Anselmino
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Claudia Solari
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Marcos Francia
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Camila Oses
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Pablo Sanchis
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Juan Bizzotto
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Camila Vazquez Echegaray
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - María Victoria Petrone
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Valeria Levi
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Elba Vazquez
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Correspondence: (E.V.); (A.G.); Tel.: +54-91144087796 (E.V.); +54-115-285-8683 (A.G.)
| | - Alejandra Guberman
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
- Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Correspondence: (E.V.); (A.G.); Tel.: +54-91144087796 (E.V.); +54-115-285-8683 (A.G.)
| |
Collapse
|