1
|
Allert C, Müller-Tidow C, Blank MF. The relevance of the hematopoietic niche for therapy resistance in acute myeloid leukemia. Int J Cancer 2024; 154:197-209. [PMID: 37565773 DOI: 10.1002/ijc.34684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023]
Abstract
The expansion of acute myeloid leukemia (AML) blasts not only suppresses normal hematopoiesis, but also alters the microenvironment. The interplay of different components of the bone marrow gives rise to altered metabolic states and activates signaling pathways which lead to resistance and impede effective therapy. Therefore, the underlying processes and mechanisms represent attractive therapeutic leverage points for overcoming therapy resistance in AML. Here, we briefly discuss resistance mechanisms based on cell interactions and secreted soluble factors in the hematopoietic niche and provide an overview of niche-related therapeutic targets currently undergoing preclinical and clinical investigation which may help improve the outcome in AML therapy.
Collapse
Affiliation(s)
- Catana Allert
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- University of Heidelberg Medical Faculty, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- University of Heidelberg Medical Faculty, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Maximilian Felix Blank
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Division Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
2
|
Present and Future Role of Immune Targets in Acute Myeloid Leukemia. Cancers (Basel) 2022; 15:cancers15010253. [PMID: 36612249 PMCID: PMC9818182 DOI: 10.3390/cancers15010253] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
It is now well known that the bone marrow (BM) cell niche contributes to leukemogenesis, but emerging data support the role of the complex crosstalk between AML cells and the BM microenvironment to induce a permissive immune setting that protects leukemic stem cells (LSCs) from therapy-induced death, thus favoring disease persistence and eventual relapse. The identification of potential immune targets on AML cells and the modulation of the BM environment could lead to enhanced anti-leukemic effects of drugs, immune system reactivation, and the restoration of AML surveillance. Potential targets and effectors of this immune-based therapy could be monoclonal antibodies directed against LSC antigens such as CD33, CD123, and CLL-1 (either as direct targets or via several bispecific T-cell engagers), immune checkpoint inhibitors acting on different co-inhibitory axes (alone or in combination with conventional AML drugs), and novel cellular therapies such as chimeric antigen receptor (CAR) T-cells designed against AML-specific antigens. Though dozens of clinical trials, mostly in phases I and II, are ongoing worldwide, results have still been negatively affected by difficulties in the identification of the optimal targets on LSCs.
Collapse
|
3
|
Protein tyrosine kinase 2b inhibition reverts niche-associated resistance to tyrosine kinase inhibitors in AML. Leukemia 2022; 36:2418-2429. [PMID: 36056084 PMCID: PMC9522596 DOI: 10.1038/s41375-022-01687-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/08/2022]
Abstract
FLT3 tyrosine kinase inhibitor (TKI) therapy evolved into a standard therapy in FLT3-mutated AML. TKI resistance, however, develops frequently with poor outcomes. We analyzed acquired TKI resistance in AML cell lines by multilayered proteome analyses. Leupaxin (LPXN), a regulator of cell migration and adhesion, was induced during early resistance development, alongside the tyrosine kinase PTK2B which phosphorylated LPXN. Resistant cells differed in cell adhesion and migration, indicating altered niche interactions. PTK2B and LPXN were highly expressed in leukemic stem cells in FLT3-ITD patients. PTK2B/FAK inhibition abrogated resistance-associated phenotypes, such as enhanced cell migration. Altered pathways in resistant cells, assessed by nascent proteomics, were largely reverted upon PTK2B/FAK inhibition. PTK2B/FAK inhibitors PF-431396 and defactinib synergized with different TKIs or daunorubicin in FLT3-mutated AML. Midostaurin-resistant and AML cells co-cultured with mesenchymal stroma cells responded particularly well to PTK2B/FAK inhibitor addition. Xenograft mouse models showed significant longer time to leukemia symptom-related endpoint upon gilteritinib/defactinib combination treatment in comparison to treatment with either drug alone. Our data suggest that the leupaxin-PTK2B axis plays an important role in acquired TKI resistance in AML. PTK2B/FAK inhibitors act synergistically with currently used therapeutics and may overcome emerging TKI resistance in FLT3-mutated AML at an early timepoint.
Collapse
|
4
|
Marchand T, Pinho S. Leukemic Stem Cells: From Leukemic Niche Biology to Treatment Opportunities. Front Immunol 2021; 12:775128. [PMID: 34721441 PMCID: PMC8554324 DOI: 10.3389/fimmu.2021.775128] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/28/2021] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is one of the most common types of leukemia in adults. While complete remission can be obtained with intensive chemotherapy in young and fit patients, relapse is frequent and prognosis remains poor. Leukemic cells are thought to arise from a pool of leukemic stem cells (LSCs) which sit at the top of the hierarchy. Since their discovery, more than 30 years ago, LSCs have been a topic of intense research and their identification paved the way for cancer stem cell research. LSCs are defined by their ability to self-renew, to engraft into recipient mice and to give rise to leukemia. Compared to healthy hematopoietic stem cells (HSCs), LSCs display specific mutations, epigenetic modifications, and a specific metabolic profile. LSCs are usually considered resistant to chemotherapy and are therefore the drivers of relapse. Similar to their HSC counterpart, LSCs reside in a highly specialized microenvironment referred to as the “niche”. Bidirectional interactions between leukemic cells and the microenvironment favor leukemic progression at the expense of healthy hematopoiesis. Within the niche, LSCs are thought to be protected from genotoxic insults. Improvement in our understanding of LSC gene expression profile and phenotype has led to the development of prognosis signatures and the identification of potential therapeutic targets. In this review, we will discuss LSC biology in the context of their specific microenvironment and how a better understanding of LSC niche biology could pave the way for new therapies that target AML.
Collapse
Affiliation(s)
- Tony Marchand
- Service d'Hématologie Clinique, Centre Hospitalier Universitaire de Rennes, Rennes, France.,Faculté de médecine, Université Rennes 1, Rennes, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U1236, Rennes, France
| | - Sandra Pinho
- Department of Pharmacology & Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
5
|
Kim HN, Ruan Y, Ogana H, Kim YM. Cadherins, Selectins, and Integrins in CAM-DR in Leukemia. Front Oncol 2020; 10:592733. [PMID: 33425742 PMCID: PMC7793796 DOI: 10.3389/fonc.2020.592733] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
The interaction between leukemia cells and the bone microenvironment is known to provide drug resistance in leukemia cells. This phenomenon, called cell adhesion-mediated drug resistance (CAM-DR), has been demonstrated in many subsets of leukemia including B- and T-acute lymphoblastic leukemia (B- and T-ALL) and acute myeloid leukemia (AML). Cell adhesion molecules (CAMs) are surface molecules that allow cell-cell or cell-extracellular matrix (ECM) adhesion. CAMs not only recognize ligands for binding but also initiate the intracellular signaling pathways that are associated with cell proliferation, survival, and drug resistance upon binding to their ligands. Cadherins, selectins, and integrins are well-known cell adhesion molecules that allow binding to neighboring cells, ECM proteins, and soluble factors. The expression of cadherin, selectin, and integrin correlates with the increased drug resistance of leukemia cells. This paper will review the role of cadherins, selectins, and integrins in CAM-DR and the results of clinical trials targeting these molecules.
Collapse
Affiliation(s)
- Hye Na Kim
- Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Cancer and Blood Disease Institute, Los Angeles, CA, United States
| | - Yongsheng Ruan
- Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Cancer and Blood Disease Institute, Los Angeles, CA, United States.,Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Heather Ogana
- Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Cancer and Blood Disease Institute, Los Angeles, CA, United States
| | - Yong-Mi Kim
- Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Cancer and Blood Disease Institute, Los Angeles, CA, United States
| |
Collapse
|
6
|
Villatoro A, Konieczny J, Cuminetti V, Arranz L. Leukemia Stem Cell Release From the Stem Cell Niche to Treat Acute Myeloid Leukemia. Front Cell Dev Biol 2020; 8:607. [PMID: 32754595 PMCID: PMC7367216 DOI: 10.3389/fcell.2020.00607] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/19/2020] [Indexed: 01/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous, complex, and deadly disease, whose treatment has hardly evolved for decades and grounds on the use of intensive chemotherapy regimens. Chemotherapy helps reduce AML bulk, but promotes relapse in the long-run by selection of chemoresistant leukemia stem cells (LSC). These may diversify and result in progression to more aggressive forms of AML. In vivo models suggest that the bone marrow stem cell niche helps LSC stay dormant and protected from chemotherapy. Here, we summarize relevant changes in stem cell niche homing and adhesion of AML LSC vs. healthy hematopoietic stem cells, and provide an overview of clinical trials aiming at targeting these processes for AML treatment and future directions within this field. Promising results with various non-mutation-targeted novel therapies directed to LSC eradication via interference with their anchoring to the stem cell niche have encouraged on-going or future advanced phase III clinical trials. In the coming years, we may see a shift in the focus of AML treatment to LSC-directed therapies if the prospect of improved cure rates holds true. In the future, AML treatment should lean toward personalized therapies using combinations of these compounds plus mutation-targeted agents and/or targeted delivery of chemotherapy, aiming at LSC eradication with reduced side effects.
Collapse
Affiliation(s)
- Alicia Villatoro
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Joanna Konieczny
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Vincent Cuminetti
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Lorena Arranz
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway.,Norwegian Center for Molecular Medicine (NCMM), University of Oslo, Oslo, Norway
| |
Collapse
|
7
|
Basati G, Khaksarian M, Abbaszadeh S, Lashgarian HE, Marzban A. Cancer stem cells and nanotechnological approaches for eradication. Stem Cell Investig 2019; 6:38. [PMID: 31853454 DOI: 10.21037/sci.2019.10.07] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022]
Abstract
Cancer stem cells (CSCs) are currently known as the main cause of tumor recurrence. After chemotherapy is completed, CSCs proliferate and then differentiate to generate new tumor tissues. Similar to normal stem cells, this non-uniformly distributed cell population in the tumor tissue has self-renewal capacity and is responsible for survival of the tumor and difference in its genetic and metabolic characteristics. Followed by gene instability in CSCs, new phenotypic markers are aberrantly expressed in CSCs subpopulation. Hence, some of the surface markers and metabolic pathways that are upregulated in CSCs may be applied as specific targets for development of diagnostic and therapeutic approaches. In this review article, the distinctive properties of CSCs including signal pathways implicated in self-renewal and surface markers were discussed. Moreover, targeting CSCs based on their specific properties using nanodrugs was reviewed.
Collapse
Affiliation(s)
- Gholam Basati
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Mojtaba Khaksarian
- Razi Herbal Medicine Research Center & Department of Physiology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Saber Abbaszadeh
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hamed Esmaeil Lashgarian
- Department of Biotechnology, School of Medicine, Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Abdolrazagh Marzban
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
8
|
Mastelaro de Rezende M, Ferreira AT, Paredes-Gamero EJ. Leukemia stem cell immunophenotyping tool for diagnostic, prognosis, and therapeutics. J Cell Physiol 2019; 235:4989-4998. [PMID: 31709540 DOI: 10.1002/jcp.29394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 10/25/2019] [Indexed: 12/15/2022]
Abstract
The existence of cancer stem cells is debatable in numerous solid tumors, yet in leukemia, there is compelling evidence of this cell population. Leukemic stem cells (LSCs) are altered cells in which accumulating genetic and/or epigenetic alterations occur, resulting in the transition between the normal, preleukemic, and leukemic status. These cells do not follow the normal differentiation program; they are arrested in a primitive state but with high proliferation potential, generating undifferentiated blast accumulation and a lack of a mature cell population. The identification of LSCs might guide stem cell biology research and provide key points of distinction between these cells and their normal counterparts. The identification and characterization of the main features of LSCs can be useful as tools for diagnosis and treatment. In this context, the aim of the present review was to connect immunophenotype data in the main types of leukemia to further guide technical improvements.
Collapse
Affiliation(s)
| | - Alice T Ferreira
- Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Edgar J Paredes-Gamero
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.,Division - Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| |
Collapse
|
9
|
Chattopadhyay S, Chaklader M, Law S. Aberrant Wnt Signaling Pathway in the Hematopoietic Stem/Progenitor Compartment in Experimental Leukemic Animal. J Cell Commun Signal 2019; 13:39-52. [PMID: 29978347 PMCID: PMC6381377 DOI: 10.1007/s12079-018-0470-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/22/2018] [Indexed: 10/28/2022] Open
Abstract
The evolutionarily conserved Wnt signaling pathway regulates physiological hematopoiesis, a process of formation of blood cells and has been shown to play crucial role in the development of both myeloid and lymphoid malignancies. The Wnt signaling pathway can be broadly divided into canonical and non-canonical pathways. In the present study, we investigated the pathobiology of leukemia by studying the expression profile of Wnt proteins, receptors, key signaling intermediates and endogenous Wnt antagonist involved in canonical and non-canonical pathways in the bone marrow (BM) hematopoietic stem/progenitor cell (HSPC) compartment of experimental leukemic mice. Cell adhesion molecule N-Cadherin and leukemic BM microenvironment with reference to Wnt were also studied. We used ENU, a potent carcinogen, to induce leukemia in wild type Swiss albino mice and malignant transformation was cofirmed by peripheral blood and BM studies. Flow cytometric expression analysis revealed profound up-regulation of canonical Wnt3a/β-catenin/CyclinD1 signaling axis along with N-Cadherin whereas down-regulation of non-canonical Wnt5a/Ca2+/CaMKII signaling axis in the leukemic HSPC compartment. Subsequent use of anti-Wnt3a antibody in the in vitro clonogenicity assay uncovered that anti-Wnt3a antibody preferentially inhibited the growth and number of the primitive leukemic hematopoietic CFU-GEMM and BFU-E colonies. Stromal cells derived from the leukemic BM also exhibited aberrant Wnt3a and Wnt5a protein expression. Taken together, alteration of canonical and non-canonical Wnt signaling pathways in the HSPC compartment along with classical Wnt protein expression pattern in the leukemic stromal microenvironment resulted in progression of leukemia.
Collapse
Affiliation(s)
- Sukalpa Chattopadhyay
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108, C.R. Avenue, Kolkata, West Bengal, 700 073, India
| | - Malay Chaklader
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108, C.R. Avenue, Kolkata, West Bengal, 700 073, India
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, Box 81313, 3000, Leuven, Belgium
| | - Sujata Law
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108, C.R. Avenue, Kolkata, West Bengal, 700 073, India.
| |
Collapse
|
10
|
Fonseca ICCFE, da Luz FAC, Uehara IA, Silva MJB. Cell-adhesion molecules and their soluble forms: Promising predictors of "tumor progression" and relapse in leukemia. Tumour Biol 2018; 40:1010428318811525. [PMID: 30486756 DOI: 10.1177/1010428318811525] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Some surface markers are used to discriminate certain leukemic subpopulations that retain a greater oncogenic potential than others, and, for this reason, they were termed as leukemic stem cells, similar to the concept of cancer stem cells in carcinoma. Among these surface markers are proteins involved in cell-cell adhesion or cell-matrix adhesion, and they may play a role in the relapse of leukemia, similar to metastasis in carcinomas. The most important are epithelial cadherin, neural cadherin, epithelial cell-adhesion molecule, and CD44, which can be cleaved and released, and their soluble forms were found increased in serum levels of cancer patients, being implicated, in some cases, with progression, metastases, and relapse. In this review, we highlighted the role of these four adhesion molecules in carcinomas and hematological malignancies, mainly leukemia, and discuss if the serum levels of soluble forms can be correlated with the surface protein status on the leukemic cells. Accession of the soluble forms looks attractive, but their use as markers in cancer must be studied in association with other parameters, as there are significant changes in levels in other pathological conditions besides cancer. Studies correlating the levels of the forms with the status of the membrane-bound proteins in leukemic (stem) cells and correlating those parameters with relapse in leukemia may afford important knowledge and applicability of those serum markers in clinical practice. For instance, the expression of the membrane-bound forms of these adhesion proteins may have promising clinical use in leukemia and other hematological malignancies.
Collapse
Affiliation(s)
| | - Felipe Andrés Cordero da Luz
- 1 Laboratory of Tumor Biomarkers and Osteoimmunology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
- 2 Nucleus of Cancer Prevention and Research, Cancer Hospital, Federal University of Uberlândia, Uberlândia, Brazil
| | - Isadora Akemi Uehara
- 1 Laboratory of Tumor Biomarkers and Osteoimmunology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Marcelo José Barbosa Silva
- 1 Laboratory of Tumor Biomarkers and Osteoimmunology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| |
Collapse
|
11
|
Mrozik KM, Blaschuk OW, Cheong CM, Zannettino ACW, Vandyke K. N-cadherin in cancer metastasis, its emerging role in haematological malignancies and potential as a therapeutic target in cancer. BMC Cancer 2018; 18:939. [PMID: 30285678 PMCID: PMC6167798 DOI: 10.1186/s12885-018-4845-0] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022] Open
Abstract
In many types of solid tumours, the aberrant expression of the cell adhesion molecule N-cadherin is a hallmark of epithelial-to-mesenchymal transition, resulting in the acquisition of an aggressive tumour phenotype. This transition endows tumour cells with the capacity to escape from the confines of the primary tumour and metastasise to secondary sites. In this review, we will discuss how N-cadherin actively promotes the metastatic behaviour of tumour cells, including its involvement in critical signalling pathways which mediate these events. In addition, we will explore the emerging role of N-cadherin in haematological malignancies, including bone marrow homing and microenvironmental protection to anti-cancer agents. Finally, we will discuss the evidence that N-cadherin may be a viable therapeutic target to inhibit cancer metastasis and increase tumour cell sensitivity to existing anti-cancer therapies.
Collapse
Affiliation(s)
- Krzysztof Marek Mrozik
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | | | - Chee Man Cheong
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Andrew Christopher William Zannettino
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.,Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Kate Vandyke
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia. .,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.
| |
Collapse
|
12
|
Zhang Y, Xia F, Liu X, Yu Z, Xie L, Liu L, Chen C, Jiang H, Hao X, He X, Zhang F, Gu H, Zhu J, Bai H, Zhang CC, Chen GQ, Zheng J. JAM3 maintains leukemia-initiating cell self-renewal through LRP5/AKT/β-catenin/CCND1 signaling. J Clin Invest 2018; 128:1737-1751. [PMID: 29584620 DOI: 10.1172/jci93198] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 02/08/2018] [Indexed: 12/14/2022] Open
Abstract
Leukemia-initiating cells (LICs) are responsible for the initiation, development, and relapse of leukemia. The identification of novel therapeutic LIC targets is critical to curing leukemia. In this report, we reveal that junctional adhesion molecule 3 (JAM3) is highly enriched in both mouse and human LICs. Leukemogenesis is almost completely abrogated upon Jam3 deletion during serial transplantations in an MLL-AF9-induced murine acute myeloid leukemia model. In contrast, Jam3 deletion does not affect the functions of mouse hematopoietic stem cells. Moreover, knockdown of JAM3 leads to a dramatic decrease in the proliferation of both human leukemia cell lines and primary LICs. JAM3 directly associates with LRP5 to activate the downstream PDK1/AKT pathway, followed by the downregulation of GSK3β and activation of β-catenin/CCND1 signaling, to maintain the self-renewal ability and cell cycle entry of LICs. Thus, JAM3 may serve as a functional LIC marker and play an important role in the maintenance of LIC stemness through unexpected LRP5/PDK1/AKT/GSK3β/β-catenin/CCND1 signaling pathways but not via its canonical role in cell junctions and migration. JAM3 may be an ideal therapeutic target for the eradication of LICs without influencing normal hematopoiesis.
Collapse
Affiliation(s)
- Yaping Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangzhen Xia
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoye Liu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuo Yu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Xie
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ligen Liu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chiqi Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haishan Jiang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxin Hao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxiao He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feifei Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Gu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Zhu
- Department of Hematology, First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haitao Bai
- Department of Hematology, First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng Cheng Zhang
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Guo-Qiang Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junke Zheng
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Chen J, Wei H, Cheng J, Xie B, Wang B, Yi J, Tian B, Liu Z, Wang F, Zhang Z. Characteristics of doxorubicin-selected multidrug-resistant human leukemia HL-60 cells with tolerance to arsenic trioxide and contribution of leukemia stem cells. Oncol Lett 2017; 15:1255-1262. [PMID: 29399180 DOI: 10.3892/ol.2017.7353] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 06/09/2017] [Indexed: 01/26/2023] Open
Abstract
The present study selected and characterized a multidrug-resistant HL-60 human acute promyelocytic leukemia cell line, HL-60/RS, by exposure to stepwise incremental doses of doxorubicin. The drug-resistant HL-60/RS cells exhibited 85.68-fold resistance to doxorubicin and were cross-resistant to other chemotherapeutics, including cisplatin, daunorubicin, cytarabine, vincristine and etoposide. The cells over-expressed the transporters P-glycoprotein, multidrug-resistance-related protein 1 and breast-cancer-resistance protein, encoded by the adenosine triphosphate-binding cassette (ABC)B1, ABCC1 and ABCG2 genes, respectively. Unlike other recognized chemoresistant leukemia cell lines, HL-60/RS cells were also strongly cross-resistant to arsenic trioxide. The proportion of leukemia stem cells (LSCs) increased synchronously with increased of drug resistance in the doxorubicin-induced HL-60 cell population. The present study confirmed that doxorubicin-induced HL-60 cells exhibited multidrug-resistance and high arsenic-trioxide resistance. Drug-resistance in these cells may be due to surviving chemoresistant LSCs in the HL-60 population, which have been subjected to long and consecutive selection by doxorubicin.
Collapse
Affiliation(s)
- Jing Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Hulai Wei
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jie Cheng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Bei Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Bei Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Juan Yi
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Baoying Tian
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zhuan Liu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Feifei Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zhewen Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
14
|
Barwe SP, Quagliano A, Gopalakrishnapillai A. Eviction from the sanctuary: Development of targeted therapy against cell adhesion molecules in acute lymphoblastic leukemia. Semin Oncol 2017; 44:101-112. [PMID: 28923207 DOI: 10.1053/j.seminoncol.2017.06.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 05/10/2017] [Accepted: 06/29/2017] [Indexed: 02/04/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is a malignant hematological disease afflicting hematopoiesis in the bone marrow. While 80%-90% of patients diagnosed with ALL will achieve complete remission at some point during treatment, ALL is associated with high relapse rate, with a 5-year overall survival rate of 68%. The initial remission failure and the high rate of relapse can be attributed to intrinsic chemoprotective mechanisms that allow persistence of ALL cells despite therapy. These mechanisms are mediated, at least in part, through the engagement of cell adhesion molecules (CAMs) within the bone marrow microenvironment. This review assembles CAMs implicated in protection of leukemic cells from chemotherapy. Such studies are limited in ALL. Therefore, CAMs that are associated with poor outcomes or are overexpressed in ALL and have been shown to be involved in chemoprotection in other hematological cancers are also included. It is likely that these molecules play parallel roles in ALL because the CAMs identified to be a factor in ALL chemoresistance also work similarly in other hematological malignancies. We review the signaling mechanisms activated by the engagement of CAMs that provide protection from chemotherapy. Development of targeted therapies against CAMs could improve outcome and raise the overall cure rate in ALL.
Collapse
Affiliation(s)
- Sonali P Barwe
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children, Wilmington, DE.
| | - Anthony Quagliano
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children, Wilmington, DE
| | | |
Collapse
|
15
|
Sudheer Shenoy P, Bose B. Identification, isolation, quantification and systems approach towards CD34, a biomarker present in the progenitor/stem cells from diverse lineages. Methods 2017; 131:147-156. [PMID: 28684339 DOI: 10.1016/j.ymeth.2017.06.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/28/2017] [Accepted: 06/30/2017] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) constitute the diverse progenitor populations in almost every tissue and are of immense importance in the field of regenerative medicine. CD34 is a cell surface glycoprotein identified first as a marker for the MSCs of hematopoietic origin. CD34 is now known to be expressed in cells of diverse lineages (tissues of non-hematopoietic origin) such as ectoderm, mesoderm and endoderm and is considered as a general marker for progenitor cells. Here, we present detailed protocols to obtain pure populations of MSCs from three diverse lineages such as skeletal muscle, skin, and liver from mouse tissues. We also present here the protocol for systems biology approach (proteomic analysis) of these purified cells. This proteomic approach can elucidate key signalling pathways and proteins utilized by these CD34 positive cells in undifferentiated and differentiated conditions. Furthermore in-depth proteomic analysis can also identify the altered proteome which is responsible for their function during non-clinical and clinical conditions.
Collapse
Affiliation(s)
- P Sudheer Shenoy
- Department of Stem Cell and Regenerative Medicine, Yenepoya Research Center, Yenepoya University, University Road, Mangalore 575018, Karnataka, India.
| | - Bipasha Bose
- Department of Stem Cell and Regenerative Medicine, Yenepoya Research Center, Yenepoya University, University Road, Mangalore 575018, Karnataka, India.
| |
Collapse
|
16
|
Switching CAR T cells on and off: a novel modular platform for retargeting of T cells to AML blasts. Blood Cancer J 2016; 6:e458. [PMID: 27518241 PMCID: PMC5022178 DOI: 10.1038/bcj.2016.61] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 05/27/2016] [Indexed: 12/17/2022] Open
Abstract
The adoptive transfer of CD19-specific chimeric antigen receptor engineered T cells (CAR T cells) resulted in encouraging clinical trials in indolent B-cell malignancies. However, they also show the limitations of this fascinating technology: CAR T cells can lead to even life-threatening off-tumor, on-target side effects if CAR T cells crossreact with healthy tissues. Here, we describe a novel modular universal CAR platform technology termed UniCAR that reduces the risk of on-target side effects by a rapid and reversible control of CAR T-cell reactivity. The UniCAR system consists of two components: (1) a CAR for an inert manipulation of T cells and (2) specific targeting modules (TMs) for redirecting UniCAR T cells in an individualized time- and target-dependent manner. UniCAR T cells can be armed against different tumor targets simply by replacement of the respective TM for (1) targeting more than one antigen simultaneously or subsequently to enhance efficacy and (2) reducing the risk for development of antigen-loss tumor variants under treatment. Here we provide ‘proof of concept' for retargeting of UniCAR T cells to CD33- and/or CD123-positive acute myeloid leukemia blasts in vitro and in vivo.
Collapse
|
17
|
Al-Mawali A, Gillis D, Lewis I. Immunoprofiling of leukemic stem cells CD34+/CD38-/CD123+ delineate FLT3/ITD-positive clones. J Hematol Oncol 2016; 9:61. [PMID: 27465508 PMCID: PMC4964068 DOI: 10.1186/s13045-016-0292-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 07/21/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a heterogeneous clonal disorder presenting with accumulation of proliferating undifferentiated blasts. Xenograft transplantation studies have demonstrated a rare population of leukemia-initiating cells called leukemic stem cells (LSCs) capable of propagating leukemia that are enriched in the CD34+/CD38- fraction. LSCs are quiescent, resistant to chemotherapy and likely responsible for relapse and therefore represent an ideal target for effective therapy. LSCs are reported to overexpress the alpha subunit of the IL-3 receptor (CD123) compared to normal CD34+/CD38- hematopoietic stem cells. It has not been demonstrated whether CD123-positive (CD34+/CD38-) subpopulation is enriched for any clonal markers of AML or any LSC properties. The aims of this study were to investigate whether FMS-like tyrosine kinase (FLT3)/internal tandem duplication (ITD) mutations are present at LSC level and whether FLT3/ITD mutation is confined to LSC as defined by CD34+/CD38-/CD123+ and not CD34+/CD38-/CD123-. METHODS Thirty-four AML cases were analyzed by five-color flow cytometry and sequential gating strategy to characterize of CD34+/CD38-/CD123+ cells. These cells were sorted, analyzed by PCR, and sequenced for FLT3/ITD. RESULTS In this study, we confirm significant expression of CD123 in 32/34 cases in the total blast population (median expression = 86 %). CD123 was also expressed in the CD34+/CD38- cells (96 ± 2 % positive) from 28/32 for CD123+ AML. CD123 was not expressed/low in normal bone marrow CD34+/CD38- cells (median expression = 0 %, range (0-.004 %). AML samples were tested for FLT3/ITD (10 positive/25). FLT3/ITD+ AML cases were sorted into two putative LSC populations according to the expression of CD123 and analyzed for FLT3/ITD again in the stem cell fractions CD34+/CD38-/CD123+ and CD34+/CD38-/CD123-. Interestingly, FLT3/ITD was only detected in CD34+/CD38-/CD123+ (7/7) and not in CD34+/CD38-/CD123- subpopulation (6/7). CONCLUSIONS This finding shows that FLT3/ITD are present at LSC level and may be a primary and not secondary event in leukemogenesis, and the oncogenic events of FLT3/ITD happen at a cell stage possessing CD123. It shows that CD123 immunoprofiling provides further delineation of FLT3+ LSC clone. This novel finding provides a rationale for treatment involving CD123-targeting antibodies with intracellular FLT3 inhibitors directed against CD34+/CD38-/CD123+. This may result in more effective anti-LSC eradication.
Collapse
Affiliation(s)
- Adhra Al-Mawali
- Division of Human Immunology and Haematology, SA Pathology, Hanson Institute, Frome Road, Adelaide, SA, 5000, Australia. .,Centre of Studies and Research, Ministry of Health, Muscat, Sultanate of Oman.
| | - David Gillis
- Division of Human Immunology and Haematology, SA Pathology, Hanson Institute, Frome Road, Adelaide, SA, 5000, Australia
| | - Ian Lewis
- Division of Human Immunology and Haematology, SA Pathology, Hanson Institute, Frome Road, Adelaide, SA, 5000, Australia
| |
Collapse
|
18
|
Galán-Díez M, Isa A, Ponzetti M, Nielsen MF, Kassem M, Kousteni S. Normal hematopoiesis and lack of β-catenin activation in osteoblasts of patients and mice harboring Lrp5 gain-of-function mutations. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1863:490-498. [PMID: 26681532 PMCID: PMC4924618 DOI: 10.1016/j.bbamcr.2015.11.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 10/29/2015] [Accepted: 11/30/2015] [Indexed: 10/22/2022]
Abstract
Osteoblasts are emerging regulators of myeloid malignancies since genetic alterations in them, such as constitutive activation of β-catenin, instigate their appearance. The LDL receptor-related protein 5 (LRP5), initially proposed to be a co-receptor for Wnt proteins, in fact favors bone formation by suppressing gut-serotonin synthesis. This function of Lrp5 occurring in the gut is independent of β-catenin activation in osteoblasts. However, it is unknown whether Lrp5 can act directly in osteoblast to influence other functions that require β-catenin signaling, particularly, the deregulation of hematopoiesis and leukemogenic properties of β-catenin activation in osteoblasts, that lead to development of acute myeloid leukemia (AML). Using mice with gain-of-function (GOF) Lrp5 alleles (Lrp5(A214V)) that recapitulate the human high bone mass (HBM) phenotype, as well as patients with the T253I HBM Lrp5 mutation, we show here that Lrp5 GOF mutations in both humans and mice do not activate β-catenin signaling in osteoblasts. Consistent with a lack of β-catenin activation in their osteoblasts, Lrp5(A214V) mice have normal trilinear hematopoiesis. In contrast to leukemic mice with constitutive activation of β-catenin in osteoblasts (Ctnnb1(CAosb)), accumulation of early myeloid progenitors, a characteristic of AML, myeloid-blasts in blood, and segmented neutrophils or dysplastic megakaryocytes in the bone marrow, are not observed in Lrp5(A214V) mice. Likewise, peripheral blood count analysis in HBM patients showed normal hematopoiesis, normal percentage of myeloid cells, and lack of anemia. We conclude that Lrp5 GOF mutations do not activate β-catenin signaling in osteoblasts. As a result, myeloid lineage differentiation is normal in HBM patients and mice. This article is part of a Special Issue entitled: Tumor Microenvironment Regulation of Cancer Cell Survival, Metastasis, Inflammation, and Immune Surveillance edited by Peter Ruvolo and Gregg L. Semenza.
Collapse
Affiliation(s)
- Marta Galán-Díez
- Department of Physiology & Cellular Biophysics, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Adiba Isa
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, DK-5000 Odense, Denmark
| | - Marco Ponzetti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Morten Frost Nielsen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, DK-5000 Odense, Denmark
| | - Moustapha Kassem
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, DK-5000 Odense, Denmark; The Danish Stem Cell Centre-DanStem, University of Copenhagen, 3B Blegdamsvej, DK-2200 Copenhagen, Denmark
| | - Stavroula Kousteni
- Department of Physiology & Cellular Biophysics, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
19
|
Marjon KD, Termini CM, Karlen KL, Saito-Reis C, Soria CE, Lidke KA, Gillette JM. Tetraspanin CD82 regulates bone marrow homing of acute myeloid leukemia by modulating the molecular organization of N-cadherin. Oncogene 2015; 35:4132-40. [PMID: 26592446 PMCID: PMC4877306 DOI: 10.1038/onc.2015.449] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/18/2015] [Accepted: 10/22/2015] [Indexed: 11/16/2022]
Abstract
Communication between acute myeloid leukemia (AML) and the bone marrow microenvironment is known to control disease progression. Therefore, regulation of AML cell trafficking and adhesion to the bone marrow is of significant interest. In this study, we demonstrate that differential expression of the membrane scaffold CD82 modulates the bone marrow homing of AML cells. By combining mutational analysis and super-resolution imaging, we identify membrane protein clustering by CD82 as a regulator of AML cell adhesion and bone marrow homing. Cluster analysis of super-resolution data indicates that N-linked glycosylation and palmitoylation of CD82 are both critical modifications that control the microdomain organization of CD82 as well as the nanoscale clustering of associated adhesion protein, N-cadherin. We demonstrate that inhibition of CD82 glycosylation increases the molecular packing of N-cadherin and promotes the bone marrow homing of AML cells. In contrast, we find that inhibition of CD82 palmitoylation disrupts the formation and organization of N-cadherin clusters and significantly diminishes bone marrow trafficking of AML. Taken together, these data establish a mechanism where the membrane organization of CD82, through specific post-translational modifications, regulates N-cadherin clustering and membrane density, which impacts the in vivo trafficking of AML cells. As such, these observations provide an alternative model for targeting AML where modulation of protein organization within the membrane may be an effective treatment therapy to disrupt the bone marrow homing potential of AML cells.
Collapse
Affiliation(s)
- K D Marjon
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - C M Termini
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - K L Karlen
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - C Saito-Reis
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - C E Soria
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - K A Lidke
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, USA
| | - J M Gillette
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
20
|
Yang P, Chen N, Jia JH, Gao XJ, Li SH, Cai J, Wang Z. Tie-1: A potential target for anti-angiogenesis therapy. ACTA ACUST UNITED AC 2015; 35:615-622. [PMID: 26489611 DOI: 10.1007/s11596-015-1479-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/19/2015] [Indexed: 01/30/2023]
Abstract
The tyrosine kinase system angiopoietin (Ang)/Tie interacts with vascular endothelial growth factor pathway and regulates vessel quiescence in adults as well as later steps of the angiogenic cascade related to vessel maturation. Since all Angs are able to bind to Tie-2 but none binds to Tie-1, the function of Tie-2 and its ligands have captured attention. However, emerging evidence indicates unique roles of the orphan receptor Tie-1 in angiogenesis under physiological and pathological conditions. It is required for maintaining vascular endothelial cell integrity and survival during murine embryo development and in adult and may be involved in modulating differentiation of hematopoietic cells in adult. Tie-1 exhibits poor tyrosine kinase activity and signals via forming heterodimers with Tie-2, inhibiting Tie-2 signaling mediated by Angs. This inhibition can be relieved by Tie-1 ectodomain cleavage mediated by tumor- and inflammatory-related factors, which causes destabilization of vessels and initiates vessel remodeling. Up-regulated Tie-1 expression has been found not only in some leukemia cells and tumor related endothelial cells but also in cytoplasm of carcinoma cells of a variety of human solid tumors, which is associated with tumor progression. In addition, it has pro-inflammatory functions in endothelial cells and is involved in some inflammatory diseases associated with angiogenesis. Recent research indicated that Tie-1 gene ablation exhibited significant effects on tumor blood- and lymph-angiogenesis and improved anti-Ang therapy, suggesting Tie-1 may be a potential target for tumor anti-angiogenesis treatment.
Collapse
MESH Headings
- Angiogenesis Inhibitors/therapeutic use
- Angiopoietins/genetics
- Angiopoietins/metabolism
- Animals
- Embryo, Mammalian
- Embryonic Development/genetics
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Gene Expression Regulation, Developmental
- Gene Expression Regulation, Neoplastic
- Humans
- Mice
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/metabolism
- Neoplasms/pathology
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Protein Binding
- Receptor, TIE-1/antagonists & inhibitors
- Receptor, TIE-1/genetics
- Receptor, TIE-1/metabolism
- Receptor, TIE-2/genetics
- Receptor, TIE-2/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Ping Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Obstetrics and Gynecology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832008, China
| | - Na Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing-Hui Jia
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xue-Jiao Gao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shi-Han Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
21
|
Mukherjee S, Saha S, Manna A, Mazumdar M, Chakraborty S, Paul S, Das T. Targeting Cancer Stem Cells by Phytochemicals: a Multimodal Approach to Colorectal Cancer. CURRENT COLORECTAL CANCER REPORTS 2014. [DOI: 10.1007/s11888-014-0251-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|