1
|
Zhao Y, Qin C, Zhao B, Wang Y, Li Z, Li T, Yang X, Wang W. Pancreatic cancer stemness: dynamic status in malignant progression. J Exp Clin Cancer Res 2023; 42:122. [PMID: 37173787 PMCID: PMC10182699 DOI: 10.1186/s13046-023-02693-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most aggressive malignancies worldwide. Increasing evidence suggests that the capacity for self-renewal, proliferation, and differentiation of pancreatic cancer stem cells (PCSCs) contribute to major challenges with current PC therapies, causing metastasis and therapeutic resistance, leading to recurrence and death in patients. The concept that PCSCs are characterized by their high plasticity and self-renewal capacities is central to this review. We focused specifically on the regulation of PCSCs, such as stemness-related signaling pathways, stimuli in tumor cells and the tumor microenvironment (TME), as well as the development of innovative stemness-targeted therapies. Understanding the biological behavior of PCSCs with plasticity and the molecular mechanisms regulating PC stemness will help to identify new treatment strategies to treat this horrible disease.
Collapse
Affiliation(s)
- Yutong Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in, Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Cheng Qin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in, Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Bangbo Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in, Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Yuanyang Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in, Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Zeru Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in, Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Tianyu Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in, Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Xiaoying Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in, Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Weibin Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China.
- National Science and Technology Key Infrastructure On Translational Medicine in, Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China.
| |
Collapse
|
2
|
Guerrero EN, Vega S, Fu C, De León R, Beltran D, Solis MA. Increased proliferation and differentiation capacity of placenta-derived mesenchymal stem cells from women of median maternal age correlates with telomere shortening. Aging (Albany NY) 2021; 13:24542-24559. [PMID: 34845112 PMCID: PMC8660609 DOI: 10.18632/aging.203724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 11/22/2021] [Indexed: 01/09/2023]
Abstract
Mesenchymal stem cells (MSCs) experience functional decline with systemic aging, resulting in reduced proliferation, increased senescence, and lower differentiation potential. The placenta represents a valuable source of MSCs, but the possible effect of donor age on the properties of placenta-derived mesenchymal stem cells (PDMSCs) has not been thoroughly studied. Thus, the aim of this study was to underscore the effect of maternal age on the biological characteristics and stemness properties of PDMSCs. PDMSCs were isolated from 5 donor age groups (A: 18-21, B: 22-25, C: 26-30, D:31-35 and E: ≥36 years) for comparison of morphological, proliferative and differentiation properties. The pluripotency markers NANOG, OCT4, and SSEA4, as well as multipotency and differentiation markers, showed higher expression in PDMSCs from mothers aged 22-35 years, with up to a 7-fold increase in adipogenesis. Cumulative population doubling, cell growth curves, and colony-forming unit-fibroblast assays revealed higher self-renewal ability in donors 26-30 years old. An increase in the proliferative characteristics of PDMSCs correlated with increased telomere shortening, suggesting that shorter telomere lengths could be related to cellular division rather than aging. A clear understanding of the effect of maternal age on MSC regenerative potential will assist in increasing the effectiveness of future cell therapies.
Collapse
Affiliation(s)
- Erika N. Guerrero
- Stem Cell Research Group, Department of Research in Sexual and Reproductive Health, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama
- Universidad Latina de Panamá, Panama City, Republic of Panama
| | - Shantal Vega
- Stem Cell Research Group, Department of Research in Sexual and Reproductive Health, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama
- Universidad de Panamá, Panama City, Republic of Panama
| | - Cindy Fu
- Stem Cell Research Group, Department of Research in Sexual and Reproductive Health, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama
- Universidad Latina de Panamá, Panama City, Republic of Panama
| | - Ruth De León
- Stem Cell Research Group, Department of Research in Sexual and Reproductive Health, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama
| | - Davis Beltran
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama
| | - Mairim Alexandra Solis
- Stem Cell Research Group, Department of Research in Sexual and Reproductive Health, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama
- Universidad de Panamá, Panama City, Republic of Panama
- Sistema Nacional de Investigación, SENACYT, Panama City, Republic of Panama
| |
Collapse
|
3
|
Yaghobi Z, Movassaghpour A, Talebi M, Abdoli Shadbad M, Hajiasgharzadeh K, Pourvahdani S, Baradaran B. The role of CD44 in cancer chemoresistance: A concise review. Eur J Pharmacol 2021; 903:174147. [PMID: 33961871 DOI: 10.1016/j.ejphar.2021.174147] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022]
Abstract
CD44 is a cell surface adhesion molecule, which is overexpressed on cancer stem cells. The interaction of CD44 with hyaluronan is responsible for tumor development, metastasis, and expression of the chemoresistant phenotype. The overexpression of CD44 impedes the cytotoxic effect of chemotherapy medications in various cancers. Therefore, the high expression of CD44 is associated with a poor prognosis in affected patients. This high expression of CD44 in various cancers has provided an ample opportunity for the treatment of patients with chemoresistant malignancy. This review aims to demonstrate the various cross-talk between CD44 and intracellular and extracellular factors and highlight its role in developing chemoresistant tumors in some troublesome cancers.
Collapse
Affiliation(s)
- Zohreh Yaghobi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aliakbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Talebi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Shiva Pourvahdani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Dzobo K, Sinkala M. Cancer Stem Cell Marker CD44 Plays Multiple Key Roles in Human Cancers: Immune Suppression/Evasion, Drug Resistance, Epithelial-Mesenchymal Transition, and Metastasis. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:313-332. [PMID: 33961518 DOI: 10.1089/omi.2021.0025] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
One of the most frequently utilized cancer stem cell markers in human cancers, including colorectal cancer and breast cancer, is CD44. A glycoprotein, CD44, traverses the cell membrane and binds to many ligands, including hyaluronan, resulting in activation of signaling cascades. There are conflicting data, however, on expression of CD44 in relationship to subtypes of cancers. Moreover, the associations of CD44 expression with drug resistance, immune infiltration, epithelial-mesenchymal transition (EMT), metastasis, and clinical prognosis in several cancer types are not clear and call for further studies. We report here an original study on CD44 expression in several human cancers and its relationship with tumorigenesis. We harnessed data from the publicly available databases, including The Cancer Genome Atlas, Gene Expression Profiling Interactive Analysis, Oncomine, Genomics of Drug Sensitivity in Cancer, and the Tumor Immune Estimation Resource. Our analysis reveals that CD44 expression varies across cancer types and is significantly associated with cancer patients' survival, in gastric and pancreatic cancers (p < 0.05). In addition, CD44 expression is closely linked with immune infiltration and immune suppressive features in pancreatic, colon adenocarcinoma, and stomach cancer. High CD44 expression was significantly correlated with the expression of drug resistance, EMT, and metastasis associated genes. Tumors expressing high CD44 have higher mutation burden and afflict older patients compared to tumors expressing low CD44. Cell lines expressing high CD44 are more resistant to anticancer drugs compared to those expressing low CD44. Protein-protein interaction investigations and functional enrichment analysis showed that CD44 interacts with gene products related to cell-substrate adhesion, migration, platelet activation, and cellular response to stress. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that these genes play key roles in biological adhesion, cell component organization, locomotion, G-α-signaling, and the response to stimulus. In summary, these findings lend evidence for the multiple key roles played by CD44 in tumorigenesis and suggest that CD44 is considered further in future studies of cancer pathogenesis and the search for novel molecular targets and personalized medicine biomarkers in clinical oncology.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Musalula Sinkala
- Division of Computational Biology, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| |
Collapse
|
5
|
Agarwal G, Agiwal S, Srivastava A. Hyaluronic acid containing scaffolds ameliorate stem cell function for tissue repair and regeneration. Int J Biol Macromol 2020; 165:388-401. [PMID: 32961192 DOI: 10.1016/j.ijbiomac.2020.09.107] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/06/2020] [Accepted: 09/15/2020] [Indexed: 12/25/2022]
Abstract
Recent evidence based studies have proposed hyaluronic acid (HA) as an emerging biopolymer for various tissue engineering application. Meanwhile, stem cells (SCs) have also gained immense popularity for their tissue regenerative capacity. Thus, combining HA and stem cells for tissue engineering application have shown to foster tissue repair and regeneration process. HA possesses the ability to interact with SCs via cellular surface receptors along with the capacity to elicit the process of differentiation. The influence of HA on stem cells has been widely investigated in cartilage and bone repair but their properties of reducing inflammation has also been explored in various other tissue repair processes. In this review, we have provided an insight to the effect of crosslinked and non-crosslinked HA on various stem cells. Further, HA based scaffolds combined with stem cells have shown to have a synergistic effect in the regeneration capacity. Also, various chemically modified HA and biomolecules conjugated HA as a suitable carrier or matrix for stem cells delivery and the effect of HA in fine tuning the stem cells function is discussed.
Collapse
Affiliation(s)
- Gopal Agarwal
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Opp. Airforce Station, Palaj, Gandhinagar 382355, Gujarat, India
| | - Shubham Agiwal
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Opp. Airforce Station, Palaj, Gandhinagar 382355, Gujarat, India
| | - Akshay Srivastava
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Opp. Airforce Station, Palaj, Gandhinagar 382355, Gujarat, India.
| |
Collapse
|
6
|
Solis MA, Wei YH, Chang CH, Yu CH, Huang LLH. Hyaluronan Induces a Mitochondrial Functional Switch in Fast-Proliferating Human Mesenchymal Stem. Int J Stem Cells 2020; 13:151-162. [PMID: 31910510 PMCID: PMC7119204 DOI: 10.15283/ijsc19004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/23/2019] [Accepted: 04/21/2019] [Indexed: 11/09/2022] Open
Abstract
Background and Objectives Hyaluronan preserves the proliferation and differentiation potential of mesenchymal stem cells. Supplementation of low-concentration hyaluronan (SHA) in stem cells culture medium increases its proliferative rate, whereas coated-surface hyaluronan (CHA) maintains cells in a slow-proliferating mode. We have previously demonstrated that in CHA, the metabolic proliferative state of stem cells was influenced by upregulating mitochondrial biogenesis and function. However, the effect of SHA on stem cells' energetic status remains unknown. In this study, we demonstrate the effect that low-concentration SHA at 0.001 mg/ml (SHA0.001) and high-concentration SHA at 5 mg/ml (SHA5) exert on stem cells' mitochondrial function compared with CHA and noncoated tissue culture surface (control). Methods and Results Fast-proliferating human placenta-derived mesenchymal stem cells (PDMSCs) cultured on SHA0.001 exhibited reduced mitochondrial mass, lower mitochondrial DNA copy number, and lower oxygen consumption rate compared with slow-proliferating PDMSCs cultured on CHA at 5.0 (CHA5) or 30 μg/cm2 (CHA30). The reduced mitochondrial biogenesis observed in SHA0.001 was accompanied by a 2-fold increased ATP content and lactate production, suggesting that hyaluronan-induced fast-proliferating PDMSCs may rely less on mitochondrial function as an energy source and induce a mitochondrial functional switch to glycolysis. Conclusions PDMSCs cultured on both CHA and SHA exhibited a reduction in reactive oxygen species levels. The results from this study clarify our understandings on the effect of hyaluronan on stem cells and provide important insights into the effect of distinct supplementation methods used during cell therapies.
Collapse
Affiliation(s)
- Mairim Alexandra Solis
- Gorgas Memorial Institute for Health Studies, Panama, Panama.,Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Yau-Huei Wei
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Department of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Chiung-Hsin Chang
- Department of Obstetrics and Gynecology, National Cheng Kung University, Tainan, Taiwan
| | - Chen-Hsiang Yu
- Department of Obstetrics and Gynecology, National Cheng Kung University, Tainan, Taiwan
| | - Lynn L H Huang
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Research Center of Excellence in Regenerative Medicine, National Cheng Kung University, Tainan, Taiwan.,International Research Center of Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
7
|
Tammi MI, Oikari S, Pasonen-Seppänen S, Rilla K, Auvinen P, Tammi RH. Activated hyaluronan metabolism in the tumor matrix — Causes and consequences. Matrix Biol 2019; 78-79:147-164. [PMID: 29709595 DOI: 10.1016/j.matbio.2018.04.012] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/13/2018] [Accepted: 04/25/2018] [Indexed: 02/08/2023]
|
8
|
Solis MA, Moreno Velásquez I, Correa R, Huang LLH. Stem cells as a potential therapy for diabetes mellitus: a call-to-action in Latin America. Diabetol Metab Syndr 2019; 11:20. [PMID: 30820250 PMCID: PMC6380040 DOI: 10.1186/s13098-019-0415-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
Latin America is a fast-growing region that currently faces unique challenges in the treatment of all forms of diabetes mellitus. The burden of this disease will be even greater in the coming years due, in part, to the large proportion of young adults living in urban areas and engaging in unhealthy lifestyles. Unfortunately, the national health systems in Latin-American countries are unprepared and urgently need to reorganize their health care services to achieve diabetic therapeutic goals. Stem cell research is attracting increasing attention as a promising and fast-growing field in Latin America. As future healthcare systems will include the development of regenerative medicine through stem cell research, Latin America is urged to issue a call-to-action on stem cell research. Increased efforts are required in studies focused on stem cells for the treatment of diabetes. In this review, we aim to inform physicians, researchers, patients and funding sources about the advances in stem cell research for possible future applications in diabetes mellitus. Emerging studies are demonstrating the potential of stem cells for β cell differentiation and pancreatic regeneration. The major economic burden implicated in patients with diabetes complications suggests that stem cell research may relieve diabetic complications. Closer attention should be paid to stem cell research in the future as an alternative treatment for diabetes mellitus.
Collapse
Affiliation(s)
| | | | - Ricardo Correa
- Department of Medicine, Warren Alpert School of Medicine, Brown University, Rhode Island, USA
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ USA
| | - Lynn L. H. Huang
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Research Center of Excellence in Regenerative Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
9
|
Quiescent Human Mesenchymal Stem Cells Are More Resistant to Heat Stress than Cycling Cells. Stem Cells Int 2018; 2018:3753547. [PMID: 30675168 PMCID: PMC6323451 DOI: 10.1155/2018/3753547] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/20/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
Quiescence is the prevailing state of many cell types under homeostatic conditions. Yet, surprisingly, little is known about how quiescent cells respond to environmental challenges. The aim of the present study is to compare stress responses of cycling and quiescent mesenchymal stem cells (MSC). Human endometrial mesenchymal cells (eMSС) were employed as adult stem cells. eMSC quiescence was modeled by serum starvation. Sublethal heat shock (HS) was used as a stress factor. Both quiescent and cycling cells were heated at 45°C for 30 min and then returned to standard culture conditions for their recovery. HS response was monitored by DNA damage response, stress-induced premature senescence (SIPS), cell proliferation activity, and oxidative metabolism. It has been found that quiescent cells repair DNA more rapidly, resume proliferation, and undergo SIPS less than proliferating cells. HS-enforced ROS production in heated cycling cells was accompanied with increased expression of genes regulating redox-active proteins. Quiescent cells exposed to HS did not intensify the ROS production, and genes involved in antioxidant defense were mostly silent. Altogether, the results have shown that quiescent cells are more resistant to heat stress than cycling cells. Next-generation sequencing (NGS) demonstrates that HS-survived cells retain differentiation capacity and do not exhibit signs of spontaneous transformation.
Collapse
|
10
|
Ru nanoparticles coated with γ-Fe2O3 promoting and monitoring the differentiation of human mesenchymal stem cells via MRI tracking. Colloids Surf B Biointerfaces 2018; 170:701-711. [DOI: 10.1016/j.colsurfb.2018.05.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 01/13/2023]
|
11
|
The biology and role of CD44 in cancer progression: therapeutic implications. J Hematol Oncol 2018; 11:64. [PMID: 29747682 PMCID: PMC5946470 DOI: 10.1186/s13045-018-0605-5] [Citation(s) in RCA: 852] [Impact Index Per Article: 121.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023] Open
Abstract
CD44, a non-kinase transmembrane glycoprotein, is overexpressed in several cell types including cancer stem cells and frequently shows alternative spliced variants that are thought to play a role in cancer development and progression. Hyaluronan, the main ligand for CD44, binds to and activates CD44 resulting in activation of cell signaling pathways that induces cell proliferation, increases cell survival, modulates cytoskeletal changes, and enhances cellular motility. The different functional roles of CD44 standard (CD44s) and specific CD44 variant (CD44v) isoforms are not fully understood. CD44v contain additional peptide motifs that can interact with and sequester growth factors and cytokines at the cell surface thereby functioning as coreceptors to facilitate cell signaling. Moreover, CD44v were expressed in metastasized tumors, whereas switching between CD44v and CD44s may play a role in regulating epithelial to mesenchymal transition (EMT) and in the adaptive plasticity of cancer cells. Here, we review current data on the structural and functional properties of CD44, the known roles for CD44 in tumorigencity, the regulation of CD44 expression, and the potential for targeting CD44 for cancer therapy.
Collapse
|
12
|
Wang Z, Zhao K, Hackert T, Zöller M. CD44/CD44v6 a Reliable Companion in Cancer-Initiating Cell Maintenance and Tumor Progression. Front Cell Dev Biol 2018; 6:97. [PMID: 30211160 PMCID: PMC6122270 DOI: 10.3389/fcell.2018.00097] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/08/2018] [Indexed: 12/19/2022] Open
Abstract
Metastasis is the leading cause of cancer death, tumor progression proceeding through emigration from the primary tumor, gaining access to the circulation, leaving the circulation, settling in distant organs and growing in the foreign environment. The capacity of a tumor to metastasize relies on a small subpopulation of cells in the primary tumor, so called cancer-initiating cells (CIC). CIC are characterized by sets of markers, mostly membrane anchored adhesion molecules, CD44v6 being the most frequently recovered marker. Knockdown and knockout models accompanied by loss of tumor progression despite unaltered primary tumor growth unraveled that these markers are indispensable for CIC. The unexpected contribution of marker molecules to CIC-related activities prompted research on underlying molecular mechanisms. This review outlines the contribution of CD44, particularly CD44v6 to CIC activities. A first focus is given to the impact of CD44/CD44v6 to inherent CIC features, including the crosstalk with the niche, apoptosis-resistance, and epithelial mesenchymal transition. Following the steps of the metastatic cascade, we report on supporting activities of CD44/CD44v6 in migration and invasion. These CD44/CD44v6 activities rely on the association with membrane-integrated and cytosolic signaling molecules and proteases and transcriptional regulation. They are not restricted to, but most pronounced in CIC and are tightly regulated by feedback loops. Finally, we discuss on the engagement of CD44/CD44v6 in exosome biogenesis, loading and delivery. exosomes being the main acteurs in the long-distance crosstalk of CIC with the host. In brief, by supporting the communication with the niche and promoting apoptosis resistance CD44/CD44v6 plays an important role in CIC maintenance. The multifaceted interplay between CD44/CD44v6, signal transducing molecules and proteases facilitates the metastasizing tumor cell journey through the body. By its engagement in exosome biogenesis CD44/CD44v6 contributes to disseminated tumor cell settlement and growth in distant organs. Thus, CD44/CD44v6 likely is the most central CIC biomarker.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong, China
| | - Kun Zhao
- Pancreas Section, University Hospital of Surgery, Heidelberg, Germany
| | - Thilo Hackert
- Pancreas Section, University Hospital of Surgery, Heidelberg, Germany
| | - Margot Zöller
- Pancreas Section, University Hospital of Surgery, Heidelberg, Germany
- *Correspondence: Margot Zöller
| |
Collapse
|
13
|
Medical Applications of Collagen and Hyaluronan in Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1077:285-306. [PMID: 30357694 DOI: 10.1007/978-981-13-0947-2_15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In order to develop and commercialize for the regenerative medicinal products, smart biomaterials with biocompatibility must be needed. In this chapter, we introduce collagen and hyaluronic acid (HA) as extracellular matrix as well as deal with the molecular mechanism as microenvironment, mechanistic effects, and gene expression. Application of collagen and HA have been reviewed in the area of orthopedics, orthopedics, ophthalmology, dermatology and plastic surgery. Finally, the ongoing and commercial products of collagen and HA for regenerative medicine have been introduced.
Collapse
|
14
|
Wong TY, Chang CH, Yu CH, Huang LLH. Hyaluronan keeps mesenchymal stem cells quiescent and maintains the differentiation potential over time. Aging Cell 2017; 16:451-460. [PMID: 28474484 PMCID: PMC5418204 DOI: 10.1111/acel.12567] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2016] [Indexed: 12/13/2022] Open
Abstract
Hyaluronan (HA), an abundant polysaccharide found in human bodies, plays a role in the mesenchymal stem cells (MSCs) maintenance. We had previously found that HA prolonged the lifespan, and prevented the cellular aging of murine adipose-derived stromal cells. Recently, we had also summarized the potential pathways associated with HA regulation in human MSCs. In this study, we used the human placenta-derived MSCs (PDMSC) to investigate the effectiveness of HA in maintaining the PDMSC. We found that coating the culture surface coated with 30 μg cm-2 of HA (C) led to cluster growth of PDMSC, and maintained a higher number of PDMSC in quiescence compared to those grown on the normal tissue culture surface (T). PDMSC were treated for either 4 (short-term) or 19 (long-term) consecutive passages. PDMSC which were treated with HA for 19 consecutive passages had reduced cell enlargement, preserved MSCs biomarker expressions and osteogenic potential when compared to those grown only on T. The PDMSC transferred to T condition after long-term HA treatment showed preserved replicative capability compared to those on only T. The telomerase activity of the HA-treated PDMSC was also higher than that of untreated PDMSC. These data suggested a connection between HA and MSC maintenance. We suggest that HA might be regulating the distribution of cytoskeletal proteins on cell spreading in the event of quiescence to preserve MSC stemness. Maintenance of MSCs stemness delayed cellular aging, leading to the anti-aging phenotype of PDMSC.
Collapse
Affiliation(s)
- Tzyy Yue Wong
- Institute of Biotechnology; College of Bioscience and Biotechnology; National Cheng Kung University; Tainan Taiwan
| | - Chiung-Hsin Chang
- Department of Obstetrics and Gynecology; National Cheng Kung University; Tainan Taiwan
| | - Chen-Hsiang Yu
- Department of Obstetrics and Gynecology; National Cheng Kung University; Tainan Taiwan
| | - Lynn L. H. Huang
- Institute of Biotechnology; College of Bioscience and Biotechnology; National Cheng Kung University; Tainan Taiwan
- Department of Biotechnology and Bioindustry Sciences; College of Bioscience and Biotechnology; National Cheng Kung University; Tainan Taiwan
- Institute of Clinical Medicine; College of Medicine; National Cheng Kung University; Tainan Taiwan
- Research Center of Excellence in Regenerative Medicine; National Cheng Kung University; Tainan Taiwan
- Advanced Optoelectronic Technology Center; National Cheng Kung University; Tainan Taiwan
| |
Collapse
|
15
|
Vanneste M, Hanoux V, Bouakka M, Bonnamy PJ. Hyaluronate synthase-2 overexpression alters estrogen dependence and induces histone deacetylase inhibitor-like effects on ER-driven genes in MCF7 breast tumor cells. Mol Cell Endocrinol 2017; 444:48-58. [PMID: 28137613 DOI: 10.1016/j.mce.2017.01.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 01/26/2017] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
Abstract
In breast carcinoma cells, high levels of hyaluronan (HA) and its CD44 receptor are frequently associated with alteration in estrogen signaling. We demonstrate that stable hyaluronate synthase 2 (HAS2) overexpression in estrogen receptor α (ERα) -positive MCF7 cells oppositely altered estrogen dependence of cell growth and its sensitivity towards antiestrogens. Albeit without effect on ERα expression and estradiol binding properties, HAS2 overexpression increased ERα Ser118 phosphorylation as well as transcriptional activity of estrogen in an ERE-luciferase reporter gene assay. However, HAS2 overexpression induced partial silencing of E2 driven-genes without affecting the magnitude of regulation by estradiol. This effect was associated with half-reduction in the activity of nuclear histone deacetylases (HDACs) through a post-translational mechanism likely consecutive to the enhanced expression of the histone acetyl-transferase EP300. In conclusion, increase in HA/CD44 interactions may contribute, through an HDAC inhibitor-like and ER-independent mechanism, to the silencing of estrogen-driven genes in breast carcinoma.
Collapse
Affiliation(s)
| | | | - Mohammed Bouakka
- Université Mohammed Premier, Faculté des Sciences, Laboratoire de Biochimie, Oujda, Morocco
| | | |
Collapse
|
16
|
Quotti Tubi L, Canovas Nunes S, Brancalion A, Doriguzzi Breatta E, Manni S, Mandato E, Zaffino F, Macaccaro P, Carrino M, Gianesin K, Trentin L, Binotto G, Zambello R, Semenzato G, Gurrieri C, Piazza F. Protein kinase CK2 regulates AKT, NF-κB and STAT3 activation, stem cell viability and proliferation in acute myeloid leukemia. Leukemia 2016; 31:292-300. [PMID: 27479180 DOI: 10.1038/leu.2016.209] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 06/15/2016] [Accepted: 06/21/2016] [Indexed: 12/28/2022]
Abstract
Protein kinase CK2 sustains acute myeloid leukemia cell growth, but its role in leukemia stem cells is largely unknown. Here, we discovered that the CK2 catalytic α and regulatory β subunits are consistently expressed in leukemia stem cells isolated from acute myeloid leukemia patients and cell lines. CK2 inactivation with the selective inhibitor CX-4945 or RNA interference induced an accumulation of leukemia stem cells in the late S-G2-M phases of the cell cycle and triggered late-onset apoptosis. As a result, leukemia stem cells displayed an increased sensitivity to the chemotherapeutic agent doxorubicin. From a molecular standpoint, CK2 blockade was associated with a downmodulation of the stem cell-regulating protein BMI-1 and a marked impairment of AKT, nuclear factor-κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) activation, whereas FOXO3a nuclear activity was induced. Notably, combined CK2 and either NF-κB or STAT3 inhibition resulted in a superior cytotoxic effect on leukemia stem cells. This study suggests that CK2 blockade could be a rational approach to minimize the persistence of residual leukemia cells.
Collapse
Affiliation(s)
- L Quotti Tubi
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - S Canovas Nunes
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - A Brancalion
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - E Doriguzzi Breatta
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - S Manni
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - E Mandato
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - F Zaffino
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - P Macaccaro
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - M Carrino
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - K Gianesin
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - L Trentin
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - G Binotto
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy
| | - R Zambello
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - G Semenzato
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - C Gurrieri
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| | - F Piazza
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy.,Laboratory of Normal and Malignant Hematopoiesis, Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
17
|
Solis MA, Wei YH, Chang CH, Yu CH, Kuo PL, Huang LLH. Hyaluronan Upregulates Mitochondrial Biogenesis and Reduces Adenoside Triphosphate Production for Efficient Mitochondrial Function in Slow-Proliferating Human Mesenchymal Stem Cells. Stem Cells 2016; 34:2512-2524. [PMID: 27354288 DOI: 10.1002/stem.2404] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 05/05/2016] [Accepted: 05/14/2016] [Indexed: 12/28/2022]
Abstract
Hyaluronan-coated surfaces preserve the proliferation and differentiation potential of mesenchymal stem cells by prolonging their G1-phase transit, which maintains cells in a slow-proliferative mode. Mitochondria are known to play a crucial role in stem cell self-renewal and differentiation. In this study, for the first time, the metabolic mechanism underlying the hyaluronan-regulated slow-proliferative maintenance of stem cells was investigated by evaluating mitochondrial functions. Human placenta-derived mesenchymal stem cells (PDMSCs) cultured on hyaluronan-coated surfaces at 0.5, 3.0, 5.0, and 30 µg/cm2 were found to have an average 58% higher mitochondrial mass and an increase in mitochondrial DNA copy number compared to noncoated tissue culture surfaces (control), as well as a threefold increase in the gene expression of the mitochondrial biogenesis-related gene PGC-1α. Increase in mitochondrial biogenesis led to a hyaluronan dose-dependent increase in mitochondrial membrane potential, ATP content, and oxygen consumption rate, with reactive oxygen species levels shown to be at least three times lower compared to the control. Although hyaluronan seemed to favor mitochondrial function, cell entry into a hyaluronan-regulated slow-proliferative mode led to a fivefold reduction in ATP production and coupling efficiency levels. Together, these results suggest that hyaluronan-coated surfaces influence the metabolic proliferative state of stem cells by upregulating mitochondrial biogenesis and function with controlled ATP production. This more efficiently meets the energy requirements of slow-proliferating PDMSCs. A clear understanding of the metabolic mechanism induced by hyaluronan in stem cells will allow future applications that may overcome the current limitations faced in stem cell culture. Stem Cells 2016;34:2512-2524.
Collapse
Affiliation(s)
- Mairim Alexandra Solis
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Yau-Huei Wei
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Department of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Chiung-Hsin Chang
- Department of Obstetrics and Gynecology, National Cheng Kung University, Tainan, Taiwan
| | - Chen-Hsiang Yu
- Department of Obstetrics and Gynecology, National Cheng Kung University, Tainan, Taiwan
| | - Pao-Lin Kuo
- Department of Obstetrics and Gynecology, National Cheng Kung University, Tainan, Taiwan
| | - Lynn L H Huang
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan. .,Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan. .,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Research Center of Excellence in Regenerative Medicine, National Cheng Kung University, Tainan, Taiwan. .,Advanced Optoelectronic Technology Center, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
18
|
Elevated integrin α6β4 expression is associated with venous invasion and decreased overall survival in non-small cell lung cancer. Hum Pathol 2016; 54:174-83. [PMID: 27107458 DOI: 10.1016/j.humpath.2016.04.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/26/2016] [Accepted: 04/08/2016] [Indexed: 12/12/2022]
Abstract
Lung cancer carries a poor prognosis and is the most common cause of cancer-related death worldwide. The integrin α6β4, a laminin receptor, promotes carcinoma progression in part by cooperating with various growth factor receptors to facilitate invasion and metastasis. In carcinoma cells with mutant TP53, the integrin α6β4 promotes cell survival. TP53 mutations and integrin α6β4 overexpression co-occur in many aggressive malignancies. Because of the high frequency of TP53 mutations in lung squamous cell carcinoma (SCC), we sought to investigate the association of integrin β4 expression with clinicopathologic features and survival in non-small cell lung cancer (NSCLC). We constructed a lung cancer tissue microarray and stained sections for integrin β4 subunit expression using immunohistochemistry. We found that integrin β4 expression is elevated in SCC compared with adenocarcinoma (P<.0001), which was confirmed in external gene expression data sets (P<.0001). We also determined that integrin β4 overexpression associates with the presence of venous invasion (P=.0048) and with reduced overall patient survival (hazard ratio, 1.46; 95% confidence interval, 1.01-2.09; P=.0422). Elevated integrin β4 expression was also shown to associate with reduced overall survival in lung cancer gene expression data sets (hazard ratio, 1.49; 95% confidence interval, 1.31-1.69; P<.0001). Using cBioPortal, we generated a network map demonstrating the 50 most highly altered genes neighboring ITGB4 in SCC, which included laminins, collagens, CD151, genes in the EGFR and PI3K pathways, and other known signaling partners. In conclusion, we demonstrate that integrin β4 is overexpressed in NSCLC where it is an adverse prognostic marker.
Collapse
|
19
|
Simpson RML, Hong X, Wong MM, Karamariti E, Bhaloo SI, Warren D, Kong W, Hu Y, Xu Q. Hyaluronan Is Crucial for Stem Cell Differentiation into Smooth Muscle Lineage. Stem Cells 2016; 34:1225-38. [PMID: 26867148 PMCID: PMC4864761 DOI: 10.1002/stem.2328] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/09/2015] [Accepted: 12/09/2015] [Indexed: 01/16/2023]
Abstract
Deciphering the extracellular signals that regulate SMC differentiation from stem cells is vital to further our understanding of the pathogenesis of vascular disease and for development of cell-based therapies and tissue engineering. Hyaluronan (HA) has emerged as an important component of the stem cell niche, however its role during stem cell differentiation is a complicated and inadequately defined process. This study aimed to investigate the role of HA in embryonic stem cell (ESC) differentiation toward a SMC lineage. ESCs were seeded on collagen-IV in differentiation medium to generate ESC-derived SMCs (esSMCs). Differentiation coincided with increased HA synthase (HAS) 2 expression, accumulation of extracellular HA and its assembly into pericellular matrices. Inhibition of HA synthesis by 4-methylumbelliferone (4MU), removal of the HA coat by hyaluronidase (HYAL) or HAS2 knockdown led to abrogation of SMC gene expression. HA activates ERK1/2 and suppresses EGFR signaling pathways via its principle receptor, CD44. EGFR inactivation coincided with increased binding to CD44, which was further augmented by addition of high molecular weight (HMW)-HA either exogenously or via HAS2 overexpression through adenoviral gene transfer. HMW-HA-stimulated esSMCs displayed a functional role in vascular tissue engineering ex vivo, vasculogenesis in a matrigel plug model and SMC accumulation in neointimal lesions of vein grafts in mice. These findings demonstrate that HAS2-induced HA synthesis and organization drives ESC-SMC differentiation. Thus, remodeling of the HA microenvironment is a critical step in directing stem cell differentiation toward a vascular lineage, highlighting HA as a potential target for treatment of vascular diseases. Stem Cells 2016;34:1225-1238.
Collapse
Affiliation(s)
- Russell M L Simpson
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Xuechong Hong
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Mei Mei Wong
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Eirini Karamariti
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Shirin Issa Bhaloo
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Derek Warren
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Yanhua Hu
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Qingbo Xu
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| |
Collapse
|
20
|
Upregulation of CD44v6 contributes to acquired chemoresistance via the modulation of autophagy in colon cancer SW480 cells. Tumour Biol 2016; 37:8811-24. [PMID: 26747179 DOI: 10.1007/s13277-015-4755-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 12/29/2015] [Indexed: 12/17/2022] Open
Abstract
The CD44 isoform containing variant exon v6 (CD44v6) plays an important role in the progression, metastasis, and prognosis of colorectal cancer (CRC). Recently, it was found that CD44v6 is involved in acquired drug resistance. This study aimed to investigate the molecular mechanism of CD44v6 in the resistance of CRC cells to chemotherapy. A stable CD44v6 overexpression model in SW480 cells was established via lentiviral transduction. The chemosensitivity of cells to 5-fluorouracil (5-FU) and oxaliplatin (L-OHP) was determined by cell counting kit (CCK)-8, lactate dehydrogenase (LDH) release, and colony formation assays. Immunohistochemical staining of CD44v6 was performed in human CRC tissues. The key components in cell apoptosis, drug efflux and metabolism, mismatch repair, autophagy, epithelial-mesenchymal transition (EMT), and the PI3K-Akt and MAPK-Ras-Erk1/2 pathways were assessed using flow cytometry, quantitative real-time polymerase chain reaction (PCR), and western blot assays. The CD44v6 overexpression cells showed a higher viability, a lower LDH release rate, and an increased clonogenicity than the control cells under drug treatment. Moreover, overexpression of CD44v6 resulted in enhanced autophagy flux, EMT, and phosphorylation of Akt and Erk in the presence of drugs. Furthermore, high CD44v6 expression in the primary tumor was closely associated with an early recurrence in CRC patients who underwent curative surgery and adjuvant chemotherapy. In conclusion, overexpression of CD44v6 contributes to chemoresistance in SW480 cells under cytotoxic stress via the modulation of autophagy, EMT, and activation of the PI3K-Akt and MAPK-Ras-Erk pathways.
Collapse
|
21
|
Differential Proteomic Analysis of Human Placenta-Derived Mesenchymal Stem Cells Cultured on Normal Tissue Culture Surface and Hyaluronan-Coated Surface. Stem Cells Int 2015; 2016:2809192. [PMID: 27057169 PMCID: PMC4709773 DOI: 10.1155/2016/2809192] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 09/07/2015] [Accepted: 10/07/2015] [Indexed: 12/15/2022] Open
Abstract
Our previous results showed that hyaluronan (HA) preserved human placenta-derived mesenchymal stem cells (PDMSC) in a slow cell cycling mode similar to quiescence, the pristine state of stem cells in vivo, and HA was found to prevent murine adipose-derived mesenchymal stem cells from senescence. Here, stable isotope labeling by amino acid in cell culture (SILAC) proteomic profiling was used to evaluate the effects of HA on aging phenomenon in stem cells, comparing (1) old and young passage PDMSC cultured on normal tissue culture surface (TCS); (2) old passage on HA-coated surface (CHA) compared to TCS; (3) old and young passage on CHA. The results indicated that senescence-associated protein transgelin (TAGLN) was upregulated in old TCS. Protein CYR61, reportedly senescence-related, was downregulated in old CHA compared to old TCS. The SIRT1-interacting Nicotinamide phosphoribosyltransferase (NAMPT) increased by 2.23-fold in old CHA compared to old TCS, and is 0.48-fold lower in old TCS compared to young TCS. Results also indicated that components of endoplasmic reticulum associated degradation (ERAD) pathway were upregulated in old CHA compared to old TCS cells, potentially for overcoming stress to maintain cell function and suppress senescence. Our data points to pathways that may be targeted by HA to maintain stem cells youth.
Collapse
|
22
|
Kim HB, Lee SH, Um JH, Kim MJ, Hyun SK, Gong EJ, Oh WK, Kang CD, Kim SH. Sensitization of chemo-resistant human chronic myeloid leukemia stem-like cells to Hsp90 inhibitor by SIRT1 inhibition. Int J Biol Sci 2015; 11:923-34. [PMID: 26157347 PMCID: PMC4495410 DOI: 10.7150/ijbs.10896] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 05/15/2015] [Indexed: 12/12/2022] Open
Abstract
Development of effective therapeutic strategies to eliminate cancer stem-like cells (CSCs), which play a major role in drug resistance and disease recurrence, is critical to improve cancer treatment outcomes. The current investigation was undertaken to examine the effectiveness of the combination treatment of Hsp90 inhibitor and SIRT1 inhibitor in inhibiting the growth of chemo-resistant stem-like cells isolated from human chronic myeloid leukemia K562 cells. Inhibition of SIRT1 by use of SIRT1 siRNA or SIRT1 inhibitors (amurensin G and EX527) effectively potentiated sensitivity of Hsp90 inhibitors (17-AAG and AUY922) in CD44(high) K562 stem-like cells expressing high levels of CSC-related molecules including Oct4, CD34, β-catenin, c-Myc, mutant p53 (mut p53), BCRP and P-glycoprotein (P-gp) as well as CD44. SIRT1 depletion caused significant down-regulation of heat shock factor 1 (HSF1)/heat shock proteins (Hsps) as well as these CSC-related molecules, which led to the sensitization of CD44(high) K562 cells to Hsp90 inhibitor by SIRT1 inhibitor. Moreover, 17-AAG-mediated activation of HSF1/Hsps and P-gp-mediated efflux, major causes of Hsp90 inhibitor resistance, was suppressed by SIRT1 inhibitor in K562-CD44(high) cells. Our data suggest that combined treatment with Hsp90 inhibitor and SIRT1 inhibitor could be an effective therapeutic approach to target CSCs that are resistant to current therapies.
Collapse
Affiliation(s)
- Hak-Bong Kim
- 1. Department of Biochemistry, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Su-Hoon Lee
- 1. Department of Biochemistry, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Jee-Hyun Um
- 2. Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Korea
| | - Mi-Ju Kim
- 1. Department of Biochemistry, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Suh-Kyung Hyun
- 1. Department of Biochemistry, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Eun-Ji Gong
- 1. Department of Biochemistry, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Won Keun Oh
- 3. College of Pharmacy, Seoul National University, Seoul 151-818, Korea
| | - Chi-Dug Kang
- 1. Department of Biochemistry, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Sun-Hee Kim
- 1. Department of Biochemistry, Pusan National University School of Medicine, Yangsan 626-870, Korea
| |
Collapse
|
23
|
Zöller M. CD44, Hyaluronan, the Hematopoietic Stem Cell, and Leukemia-Initiating Cells. Front Immunol 2015; 6:235. [PMID: 26074915 PMCID: PMC4443741 DOI: 10.3389/fimmu.2015.00235] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 04/30/2015] [Indexed: 12/14/2022] Open
Abstract
CD44 is an adhesion molecule that varies in size due to glycosylation and insertion of so-called variant exon products. The CD44 standard isoform (CD44s) is highly expressed in many cells and most abundantly in cells of the hematopoietic system, whereas expression of CD44 variant isoforms (CD44v) is more restricted. CD44s and CD44v are known as stem cell markers, first described for hematopoietic stem cells and later on confirmed for cancer- and leukemia-initiating cells. Importantly, both abundantly expressed CD44s as well as CD44v actively contribute to the maintenance of stem cell features, like generating and embedding in a niche, homing into the niche, maintenance of quiescence, and relative apoptosis resistance. This is surprising, as CD44 is not a master stem cell gene. I here will discuss that the functional contribution of CD44 relies on its particular communication skills with neighboring molecules, adjacent cells and, last not least, the surrounding matrix. In fact, it is the interaction of the hyaluronan receptor CD44 with its prime ligand, which strongly assists stem cells to fulfill their special and demanding tasks. Recent fundamental progress in support of this “old” hypothesis, which may soon pave the way for most promising new therapeutics, is presented for both hematopoietic stem cell and leukemia-initiating cell. The contribution of CD44 to the generation of a stem cell niche, to homing of stem cells in their niche, to stem cell quiescence and apoptosis resistance will be in focus.
Collapse
Affiliation(s)
- Margot Zöller
- Department of Tumor Cell Biology, University Hospital of Surgery , Heidelberg , Germany
| |
Collapse
|
24
|
Misra S, Hascall VC, Markwald RR, Ghatak S. Interactions between Hyaluronan and Its Receptors (CD44, RHAMM) Regulate the Activities of Inflammation and Cancer. Front Immunol 2015; 6:201. [PMID: 25999946 PMCID: PMC4422082 DOI: 10.3389/fimmu.2015.00201] [Citation(s) in RCA: 585] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 04/13/2015] [Indexed: 01/04/2023] Open
Abstract
The glycosaminoglycan hyaluronan (HA), a major component of extracellular matrices, and cell surface receptors of HA have been proposed to have pivotal roles in cell proliferation, migration, and invasion, which are necessary for inflammation and cancer progression. CD44 and receptor for HA-mediated motility (RHAMM) are the two main HA-receptors whose biological functions in human and murine inflammations and tumor cells have been investigated comprehensively. HA was initially considered to be only an inert component of connective tissues, but is now known as a “dynamic” molecule with a constant turnover in many tissues through rapid metabolism that involves HA molecules of various sizes: high molecular weight HA (HMW HA), low molecular weight HA, and oligosaccharides. The intracellular signaling pathways initiated by HA interactions with CD44 and RHAMM that lead to inflammatory and tumorigenic responses are complex. Interestingly, these molecules have dual functions in inflammations and tumorigenesis. For example, the presence of CD44 is involved in initiation of arthritis, while the absence of CD44 by genetic deletion in an arthritis mouse model increases rather than decreases disease severity. Similar dual functions of CD44 exist in initiation and progression of cancer. RHAMM overexpression is most commonly linked to cancer progression, whereas loss of RHAMM is associated with malignant peripheral nerve sheath tumor growth. HA may similarly perform dual functions. An abundance of HMW HA can promote malignant cell proliferation and development of cancer, whereas antagonists to HA-CD44 signaling inhibit tumor cell growth in vitro and in vivo by interfering with HMW HA-CD44 interaction. This review describes the roles of HA interactions with CD44 and RHAMM in inflammatory responses and tumor development/progression, and how therapeutic strategies that block these key inflammatory/tumorigenic processes may be developed in rodent and human diseases.
Collapse
Affiliation(s)
- Suniti Misra
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina , Charleston, SC , USA
| | - Vincent C Hascall
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland , Ohio, OH , USA
| | - Roger R Markwald
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina , Charleston, SC , USA
| | - Shibnath Ghatak
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina , Charleston, SC , USA
| |
Collapse
|
25
|
Augustin F, Fiegl M, Schmid T, Pomme G, Sterlacci W, Tzankov A. Receptor for hyaluronic acid-mediated motility (RHAMM, CD168) expression is prognostically important in both nodal negative and nodal positive large cell lung cancer. J Clin Pathol 2015; 68:368-73. [DOI: 10.1136/jclinpath-2014-202819] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/03/2015] [Indexed: 12/17/2022]
|
26
|
The ability of hyaluronan fragments to reverse the resistance of C6 rat glioma cell line to temozolomide and carmustine. Contemp Oncol (Pozn) 2014; 18:323-8. [PMID: 25477754 PMCID: PMC4248052 DOI: 10.5114/wo.2014.43493] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/11/2013] [Accepted: 04/11/2014] [Indexed: 11/18/2022] Open
Abstract
Aim of the study Hyaluronan (HA) is an extracellular matrix (ECM) polymer that may contribute to the emergence of anti-cancer drug resistance. Attempts to reverse drug resistance using small hyaluronan oligomers (oHA) are being made. The initial reports suggest that the oHA fraction may effectively reverse anti-cancer drug resistance in glioma models. However, the reversal effects of oHA of defined molecular length on glioma cells have not been investigated yet. In this study, we examined HA fragments containing 2 disaccharide units (oHA-2), 5 disaccharide units (oHA-5), and 68 kDa hyaluronan polymer (HA-68k) as agents possibly reversing the resistance of a C6 rat glioma cell line to temozolomide (TMZ) and carmustine (BCNU). Material and methods A 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) viability assay was used to assess the cytotoxicity of TMZ and BCNU in the presence or absence of the hyaluronan fragments. By comparing viability of the cells, the reversal effects of HA fragments on TMZ and BCNU resistance in C6 glioma cells were assessed. Results We found statistically significant decreases in the viability of cells in the presence of TMZ+oHA-5 as compared to TMZ alone (51.2 ±4.5 vs. 74.2 ±5.8, p = 0.0031), BCNU+o-HA5 as compared to BCNU alone (49.3 ±4.4 vs. 65.6 ±5.7, p = 0.0119), and BCNU+HA-68k as compared to BCNU alone (55.2 ±2.3 vs. 65.6 ±5.7, p = 0.0496). Conclusions Conclusions: Hyaluronan oligomers of 5 disaccharide units (oHA-5) significantly reversed the resistance of C6 cells to TMZ and BCNU. The results are only preliminary and a more thorough follow-up investigation is required to assess their actual role.
Collapse
|
27
|
Passipieri JA, Kasai-Brunswick TH, Suhett G, Martins AB, Brasil GV, Campos DB, Rocha NN, Ramos IP, Mello DB, Rodrigues DC, Christie BB, Silva-Mendes BJ, Balduíno A, Sá RM, Lopes LM, Goldenberg RC, Campos de Carvalho AC, Carvalho AB. Improvement of cardiac function by placenta-derived mesenchymal stem cells does not require permanent engraftment and is independent of the insulin signaling pathway. Stem Cell Res Ther 2014; 5:102. [PMID: 25145631 PMCID: PMC4354978 DOI: 10.1186/scrt490] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/18/2014] [Accepted: 08/08/2014] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION The objective of this work was to evaluate the efficacy of placenta-derived mesenchymal stem cell (MSC) therapy in a mouse model of myocardial infarction (MI). Since MSCs can be obtained from two different regions of the human term placenta (chorionic plate or villi), cells obtained from both these regions were compared so that the best candidate for cell therapy could be selected. METHODS For the in vitro studies, chorionic plate MSCs (cp-MSCs) and chorionic villi MSCs (cv-MSCs) were extensively characterized for their genetic stability, clonogenic and differentiation potential, gene expression, and immunophenotype. For the in vivo studies, C57Bl/6 mice were submitted to MI and, after 21 days, received weekly intramyocardial injections of cp-MSCs for 3 weeks. Cells were also stably transduced with a viral construct expressing luciferase, under the control of the murine stem cell virus (MSCV) promoter, and were used in a bioluminescence assay. The expression of genes associated with the insulin signaling pathway was analyzed in the cardiac tissue from cp-MSCs and placebo groups. RESULTS Morphology, differentiation, immunophenotype, and proliferation were quite similar between these cells. However, cp-MSCs had a greater clonogenic potential and higher expression of genes related to cell cycle progression and genome stability. Therefore, we considered that the chorionic plate was preferable to the chorionic villi for the isolation of MSCs. Sixty days after MI, cell-treated mice had a significant increase in ejection fraction and a reduction in end-systolic volume. This improvement was not caused by a reduction in infarct size. In addition, tracking of cp-MSCs transduced with luciferase revealed that cells remained in the heart for 4 days after the first injection but that the survival period was reduced after the second and third injections. Quantitative reverse transcription-polymerase chain reaction revealed similar expression of genes involved in the insulin signaling pathway when comparing cell-treated and placebo groups. CONCLUSIONS Improvement of cardiac function by cp-MSCs did not require permanent engraftment and was not mediated by the insulin signaling pathway.
Collapse
Affiliation(s)
- Juliana A Passipieri
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Instituto Nacional de Cardiologia, Rua das Laranjeiras 374, Rio de Janeiro, 22240-006, Brazil.
| | - Tais H Kasai-Brunswick
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Instituto Nacional de Cardiologia, Rua das Laranjeiras 374, Rio de Janeiro, 22240-006, Brazil.
| | - Grazielle Suhett
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Rua Rodolpho Paulo Rocco 255, Rio de Janeiro, 21941-913, Brazil.
| | - Andreza B Martins
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Instituto Nacional de Cardiologia, Rua das Laranjeiras 374, Rio de Janeiro, 22240-006, Brazil.
| | - Guilherme V Brasil
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Dilza B Campos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Nazareth N Rocha
- Universidade Federal Fluminense, Rua Professor Hernani Melo 101, Niterói, 24210-130, Brazil.
| | - Isalira P Ramos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Rua Rodolpho Paulo Rocco 255, Rio de Janeiro, 21941-913, Brazil.
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Av Carlos Chagas Filho 373, Rio de Janeiro, 21941-902, Brazil.
| | - Debora B Mello
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Deivid C Rodrigues
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Beatriz B Christie
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Bernardo J Silva-Mendes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Alex Balduíno
- Centro de Pesquisa, Tecnologia e Inovação, Universidade Veiga de Almeida, Rua Ibituruna 108, Rio de Janeiro, 20271-020, Brazil.
| | - Renato M Sá
- Centro Pré-Natal de Diagnóstico e Tratamento, Clínica Perinatal, Rua das Laranjeiras 445, Rio de Janeiro, 22240-002, Brazil.
| | - Laudelino M Lopes
- Centro Pré-Natal de Diagnóstico e Tratamento, Clínica Perinatal, Rua das Laranjeiras 445, Rio de Janeiro, 22240-002, Brazil.
- Department of Obstetrics and Gynecology, Western University, London Health Sciences Centre-Victoria Hospital, B2-401, London, ON, N6H 5W9, Canada.
| | - Regina C Goldenberg
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Av Carlos Chagas Filho 373, Rio de Janeiro, 21941-902, Brazil.
| | - Antonio C Campos de Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Instituto Nacional de Cardiologia, Rua das Laranjeiras 374, Rio de Janeiro, 22240-006, Brazil.
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Av Carlos Chagas Filho 373, Rio de Janeiro, 21941-902, Brazil.
| | - Adriana B Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Av Carlos Chagas Filho 373, Rio de Janeiro, 21941-902, Brazil.
| |
Collapse
|
28
|
Megaptche AP, Erb U, Büchler MW, Zöller M. CD44v10, osteopontin and lymphoma growth retardation by a CD44v10-specific antibody. Immunol Cell Biol 2014; 92:709-20. [PMID: 24935458 DOI: 10.1038/icb.2014.47] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 05/04/2014] [Accepted: 05/05/2014] [Indexed: 02/07/2023]
Abstract
Blockade of CD44 is considered a therapeutic option for the elimination of leukemia-initiating cells. However, the application of anti-panCD44 can be burdened by severe side effects. We determined whether these side effects could be avoided by replacing anti-panCD44 with CD44 variant isoform (CD44v)-specific antibodies in CD44v-positive hematological malignancies using the EL4 thymoma and CD44v10-transfected EL4 (EL4-v10) as models. Subcutaneous growth of EL4 and EL4-v10 was equally well inhibited by the anti-panCD44 and anti-CD44v10 antibodies, respectively. Ex vivo analysis indicated that natural killer cytotoxicity and antibody-dependent cellular cytotoxicity were the main effector mechanisms. Under local inflammation, the efficacy of anti-CD44v10 prolonged the survival time twofold compared with untreated, EL4-v10 tumor-bearing mice, and this was due to inflammation-induced expression of osteopontin (OPN). A high level of OPN in EL4-v10 tumors supported leukocyte recruitment and tumor-infiltrating T-cell activation. Taken together, in hematological malignancies expressing CD44v, anti-panCD44 can be replaced by CD44v-specific antibodies without a loss in efficacy. Furthermore, CD44v10-specific antibodies appear particularly advantageous in cutaneous leukemia therapy, as CD44v10 binding of OPN drives leukocyte recruitment and activation.
Collapse
Affiliation(s)
| | - Ulrike Erb
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Markus Wolfgang Büchler
- Allgemeine, Viszeral- und Transplantationschirurgie, University Hospital of Surgery, Heidelberg, Germany
| | - Margot Zöller
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| |
Collapse
|
29
|
Sun D, Junger WG, Yuan C, Zhang W, Bao Y, Qin D, Wang C, Tan L, Qi B, Zhu D, Zhang X, Yu T. Shockwaves induce osteogenic differentiation of human mesenchymal stem cells through ATP release and activation of P2X7 receptors. Stem Cells 2014; 31:1170-80. [PMID: 23404811 DOI: 10.1002/stem.1356] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 01/13/2013] [Accepted: 01/29/2013] [Indexed: 12/11/2022]
Abstract
Shockwave treatment promotes bone healing of nonunion fractures. In this study, we investigated whether this effect could be due to adenosine 5'-triphosphate (ATP) release-induced differentiation of human mesenchymal stem cells (hMSCs) into osteoprogenitor cells. Cultured bone marrow-derived hMSCs were subjected to shockwave treatment and ATP release was assessed. Osteogenic differentiation and mineralization of hMSCs were evaluated by examining alkaline phosphatase activity, osteocalcin production, and calcium nodule formation. Expression of P2X7 receptors and c-fos and c-jun mRNA was determined with real-time reverse transcription polymerase chain reaction and Western blotting. P2X7-siRNA, apyrase, P2 receptor antagonists, and p38 MAPK inhibitors were used to evaluate the roles of ATP release, P2X7 receptors, and p38 MAPK signaling in shockwave-induced osteogenic hMSCs differentiation. Shockwave treatment released significant amounts (≈ 7 μM) of ATP from hMSCs. Shockwaves and exogenous ATP induced c-fos and c-jun mRNA transcription, p38 MAPK activation, and hMSC differentiation. Removal of ATP with apyrase, targeting of P2X7 receptors with P2X7-siRNA or selective antagonists, or blockade of p38 MAPK with SB203580 prevented osteogenic differentiation of hMSCs. Our findings indicate that shockwaves release cellular ATP that activates P2X7 receptors and downstream signaling events that caused osteogenic differentiation of hMSCs. We conclude that shockwave therapy promotes bone healing through P2X7 receptor signaling, which contributes to hMSC differentiation.
Collapse
Affiliation(s)
- Dahui Sun
- Department of Orthopedics, The First Norman Bethune Hospital of Jilin University, Changchun, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Qu C, Rilla K, Tammi R, Tammi M, Kröger H, Lammi MJ. Extensive CD44-dependent hyaluronan coats on human bone marrow-derived mesenchymal stem cells produced by hyaluronan synthases HAS1, HAS2 and HAS3. Int J Biochem Cell Biol 2014; 48:45-54. [DOI: 10.1016/j.biocel.2013.12.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 11/27/2013] [Accepted: 12/23/2013] [Indexed: 12/30/2022]
|
31
|
Marcucci F, Bellone M, Caserta CA, Corti A. Pushing tumor cells towards a malignant phenotype: stimuli from the microenvironment, intercellular communications and alternative roads. Int J Cancer 2013; 135:1265-76. [PMID: 24174383 DOI: 10.1002/ijc.28572] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 09/26/2013] [Accepted: 10/24/2013] [Indexed: 12/13/2022]
Abstract
The tumor microenvironment produces different types of stimuli capable of endowing tumor cells with an aggressive behavior that is characterized by increased motility, invasiveness and propensity to metastasize, gain of a tumor-initiating phenotype, and drug resistance. The following classes of stimuli have been reported to promote such a malignant phenotype: (i) solid- or fluid-induced stress; (ii) altered composition of the extracellular matrix; (iii) hypoxia and low pH; (iv) innate and adaptive immune responses; (v) antitumor drugs. The simultaneous presence of more than one of these stimuli, as likely occurs in vivo, may lead to synergistic interactions in the induction of malignant traits. In many cases, the gain of a malignant phenotype is not the result of a direct effect of the stimuli on tumor cells but, rather, a stimulus-promoted cross-talk between tumor cells and other cell types within the tumor microenvironment. This cross-talk is mainly mediated by two classes of molecules: paracrine factors and adhesion receptors. Stimuli that promote a malignant phenotype can promote additional outcomes in tumor cells, including autophagy and cell death. We summarize here the available evidence about the variables that induce tumor cells to take one or the other of these roads in response to the same stimuli. At the end of this review, we address some unanswered questions in this domain and indicate future directions of research.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Centro Nazionale di Epidemiologia Sorveglianza e Promozione della Salute (CNESPS), Istituto Superiore di Sanita' (ISS), Roma, Italy; Hepatology Association of Calabria (ACE), Reggio Calabria, Italy
| | | | | | | |
Collapse
|
32
|
Ricciardelli C, Ween MP, Lokman NA, Tan IA, Pyragius CE, Oehler MK. Chemotherapy-induced hyaluronan production: a novel chemoresistance mechanism in ovarian cancer. BMC Cancer 2013; 13:476. [PMID: 24124770 PMCID: PMC3852938 DOI: 10.1186/1471-2407-13-476] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 10/01/2013] [Indexed: 12/15/2022] Open
Abstract
Background Hyaluronan (HA) an important component of the extracellular matrix, has been linked to tumor progression and drug resistance in several malignancies. However, limited data is available for ovarian cancer. This study investigated the role of hyaluronan (HA) and a potential link between the HA-CD44 pathway and membrane ATP binding cassette (ABC) transporter proteins in ovarian cancer chemoresistance. Methods We investigated the ability of HA to block the cytotoxic effects of the chemotherapy drug carboplatin, and to regulate the expression of ABC transporters in ovarian cancer cells. We also examined HA serum levels in ovarian cancer patients prior to and following chemotherapy and assessed its prognostic relevance. Results HA increased the survival of carboplatin treated ovarian cancer cells expressing the HA receptor, CD44 (OVCAR-5 and OV-90). Carboplatin significantly increased expression of HAS2, HAS3 and ABCC2 and HA secretion in ovarian cancer cell conditioned media. Serum HA levels were significantly increased in patients following platinum based chemotherapy and at both 1st and 2nd recurrence when compared with HA levels prior to treatment. High serum HA levels (>50 μg/ml) prior to chemotherapy treatment were associated with significantly reduced progression-free (P = 0.014) and overall survival (P = 0.036). HA production in ovarian cancer cells was increased in cancer tissues collected following chemotherapy treatment and at recurrence. Furthermore HA treatment significantly increased the expression of ABC drug transporters (ABCB3, ABCC1, ABCC2, and ABCC3), but only in ovarian cancer cells expressing CD44. The effects of HA and carboplatin on ABC transporter expression in ovarian cancer cells could be abrogated by HA oligomer treatment. Importantly, HA oligomers increased the sensitivity of chemoresistant SKOV3 cells to carboplatin. Conclusions Our findings indicate that carboplatin chemotherapy induces HA production which can contribute to chemoresistance by regulating ABC transporter expression. The HA-CD44 signaling pathway is therefore a promising target in platinum resistant ovarian cancer.
Collapse
Affiliation(s)
- Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, School of Paediatrics and Reproductive Health, Research Centre for Reproductive Health, Robinson Institute, University of Adelaide, Adelaide 5005, South Australia, Australia.
| | | | | | | | | | | |
Collapse
|
33
|
CD44 is associated with the aggressive phenotype of nasopharyngeal carcinoma through redox regulation. Int J Mol Sci 2013; 14:13266-81. [PMID: 23803658 PMCID: PMC3742186 DOI: 10.3390/ijms140713266] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 06/18/2013] [Accepted: 06/19/2013] [Indexed: 12/19/2022] Open
Abstract
Recent studies have shown that cancer stem-like cells (CSCs) within a tumor have the capacity for self-renewal and differentiation, and are associated with an aggressive phenotype and therapeutic resistance. Studies have also associated tumor progression with alterations in the levels of intracellular reactive oxygen species (ROS). In this study, we cultured nasopharyngeal carcinoma (NPC) CSCs in conditions that allowed sphere formation. The resulting sphere cells displayed stemness properties, characteristics of the epithelial–mesenchymal transition (EMT), and increased expression of the CSC surface marker CD44. We further evaluated the association between CD44 expression and EMT marker expression, and any correlation with redox status, in these CSCs. We showed that the EMT in sphere cells is associated with the upregulation of CD44 expression and increased ROS generation, which might promote NPC aggressiveness. We also identified the coexpression of CD44 with the EMT marker N-cadherin in sphere cells, and downregulated CD44 expression after the addition of the antioxidant N-acetyl cysteine. Our results indicate that CD44 plays a role in the EMT phenotype of CSCs in NPC, and suggest its involvement in EMT-associated ROS production. These findings might facilitate the development of a novel therapy for the prevention of NPC recurrence and metastasis.
Collapse
|
34
|
Hyaluronan regulates cell behavior: a potential niche matrix for stem cells. Biochem Res Int 2012; 2012:346972. [PMID: 22400115 PMCID: PMC3287012 DOI: 10.1155/2012/346972] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 09/24/2011] [Accepted: 11/18/2011] [Indexed: 12/20/2022] Open
Abstract
Hyaluronan is a linear glycosaminoglycan that has received special attention in the last few decades due to its extraordinary physiological functions. This highly viscous polysaccharide is not only a lubricator, but also a significant regulator of cellular behaviors during embryogenesis, morphogenesis, migration, proliferation, and drug resistance in many cell types, including stem cells. Most hyaluronan functions require binding to its cellular receptors CD44, LYVE-1, HARE, layilin, and RHAMM. After binding, proteins are recruited and messages are sent to alter cellular activities. When low concentrations of hyaluronan are applied to stem cells, the proliferative activity is enhanced. However, at high concentrations, stem cells acquire a dormant state and induce a multidrug resistance phenotype. Due to the influence of hyaluronan on cells and tissue morphogenesis, with regards to cardiogenesis, chondrogenesis, osteogenesis, and neurogenesis, it is now been utilized as a biomaterial for tissue regeneration. This paper summarizes the most important and recent findings regarding the regulation of hyaluronan in cells.
Collapse
|
35
|
Abstract
Can an abundantly expressed molecule be a reliable marker for the cancer-initiating cells (CICs; also known as cancer stem cells), which constitute the minority of cells within the mass of a tumour? CD44 has been implicated as a CIC marker in several malignancies of haematopoietic and epithelial origin. Is this a fortuitous coincidence owing to the widespread expression of the molecule or is CD44 expression advantageous as it fulfils some of the special properties that are displayed by CICs, such as self-renewal, niche preparation, epithelial-mesenchymal transition and resistance to apoptosis?
Collapse
Affiliation(s)
- Margot Zöller
- Department of Tumour Cell Biology, University Hospital of Surgery and German Cancer Research Centre, D69120 Heidelberg, Germany.
| |
Collapse
|
36
|
Sarkar D, Ankrum JA, Teo GSL, Carman CV, Karp JM. Cellular and extracellular programming of cell fate through engineered intracrine-, paracrine-, and endocrine-like mechanisms. Biomaterials 2011; 32:3053-61. [PMID: 21262537 PMCID: PMC3043463 DOI: 10.1016/j.biomaterials.2010.12.036] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 12/26/2010] [Indexed: 12/11/2022]
Abstract
A cell's fate is tightly controlled by its microenvironment. Key factors contributing to this microenvironment include physical contacts with the extracellular matrix and neighboring cells, in addition to soluble factors produced locally or distally. Alterations to these cues can drive homeostatic processes, such as tissue regeneration/wound healing, or may lead to pathologic tissue dysfunction. In vitro models of cell and tissue microenvironments are desirable for enhanced understanding of the biology and ultimately for improved treatment. However, mechanisms to exert specific control over cellular microenvironments remains a significant challenge. Genetic modification has been used but is limited to products that can be manufactured by cells and release kinetics of therapeutics cannot easily be controlled. Herein we describe a non-genetic approach to engineer cells with an intracellular depot of phenotype altering agent/s that can be used for altering cell fate via intracrine-, paracrine-, and endocrine-like mechanisms. Specifically, we show that human mesenchymal stem cells (MSCs) can be engineered with poly lactide-co-glycolic acid (PLGA) particles containing dexamethasone, which acts on cytoplasmic receptors. The controlled release properties of these particles allowed for sustained intracellular and extracellular delivery of agent to promote differentiation of particle-carrying cells, as well as neighboring cells and distant cells that do not contain particles.
Collapse
Affiliation(s)
- Debanjan Sarkar
- Center for Regenerative Therapeutics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard-MIT Division of Health Sciences & Technology, Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - James A. Ankrum
- Center for Regenerative Therapeutics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard-MIT Division of Health Sciences & Technology, Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Grace S. L. Teo
- Center for Regenerative Therapeutics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard-MIT Division of Health Sciences & Technology, Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Christopher V. Carman
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02215, USA
| | - Jeffrey M. Karp
- Center for Regenerative Therapeutics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard-MIT Division of Health Sciences & Technology, Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, MA 02139, USA
| |
Collapse
|