ijms-logo

Journal Browser

Journal Browser

Molecular Mechanism of Leukemia

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Biology".

Deadline for manuscript submissions: closed (31 December 2022) | Viewed by 21957

Special Issue Editor


E-Mail Website
Guest Editor
Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
Interests: DNA methylation; TET proteins; hematopoietic stem cells; hematopoiesis; leukemia; cancer epigenetics; cancer therapy; drug screen; metabolic diseases; obesity; diabetes; biosensor; signaling and gene expression
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Hematopoietic stem cells (HSCs) in the bone marrow ensure lifelong hematopoietic homeostasis by differentiating along the highly ordered differentiation pathways to give rise to the full repertoire of blood cells. They also undergo self-renewal to maintain a proper pool of HSCs in the bone marrow. A series of genetic and epigenetic abnormalities sometimes take place in HSPCs (hematopoietic stem and progenitors), some of which lead to disruption of the normal self-renewal, differentiation, proliferation, or survival of HSPCs, ultimately driving the neoplastic transformation of certain hematopoietic cell populations and an accumulation of these abnormal (also called ‘leukemic’) cells in the bone marrow and periphery, while substantially suppressing normal hematopoiesis. Changes in the cellular metabolism or altered intercellular interactions with non-hematopoietic cells within the bone marrow niche can also contribute to oncogenesis. Thus, understanding the fundamental molecular basis that governs normal hematopoiesis and leukemogenesis will facilitate the development of more rational and effective therapeutic interventions.

For this Special Issue, we invite original research articles or reviews that describe the molecular mechanisms of leukemia and their therapeutic applications. We will accept articles addressing how genetic or epigenetic factors modulate normal HSC self-renewal and differentiation and how their dysregulation affects the development of various types of leukemia. Furthermore, the consolidation of inputs from the HSC niche or intracellular metabolism with (epi)genetic programs to secure normal hematopoiesis and their perturbations in the pathogenesis of leukemia are also of great interest. Topics relevant to discoveries of novel genetic and epigenetic alterations, their impact on hematological oncogenesis, and advanced mechanism-based therapeutic strategies are also welcomed.

Dr. Myunggon Ko
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • epigenetics
  • chromatin modifiers
  • DNA (hydroxy)methylation
  • histone modification
  • hematopoietic
  • stem cells
  • self-renewal
  • differentiation
  • hematological malignancies
  • molecular mechanisms
  • epigenetic therapy

Related Special Issue

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

2 pages, 169 KiB  
Editorial
Editorial for the Special Issue “Molecular Mechanism of Leukemia”
by Jungeun An and Myunggon Ko
Int. J. Mol. Sci. 2023, 24(18), 13936; https://doi.org/10.3390/ijms241813936 - 11 Sep 2023
Viewed by 531
Abstract
Hematopoiesis is the intricate process responsible for all blood cell formation and maintenance, and is tightly regulated by a myriad of intrinsic and extrinsic factors [...] Full article
(This article belongs to the Special Issue Molecular Mechanism of Leukemia)

Research

Jump to: Editorial, Review

13 pages, 2688 KiB  
Article
Comprehensive Transcriptomic Analysis of VISTA in Acute Myeloid Leukemia: Insights into Its Prognostic Value
by Simona Pagliuca, Carmelo Gurnari, Keman Zhang, Tariq Kewan, Waled Bahaj, Minako Mori, Ishani Nautiyal, Marie Thérèse Rubio, Francesca Ferraro, Jaroslaw P. Maciejewski, Li Wang and Valeria Visconte
Int. J. Mol. Sci. 2022, 23(23), 14885; https://doi.org/10.3390/ijms232314885 - 28 Nov 2022
Cited by 6 | Viewed by 1982
Abstract
The V-domain Ig suppressor of T-cell activation (VISTA) has been recognized as a critical negative regulator of antitumor immune response and is gaining growing interest as a potential pharmacological target in immunotherapy. This molecule is highly expressed in hematopoietic stem cells and myeloid [...] Read more.
The V-domain Ig suppressor of T-cell activation (VISTA) has been recognized as a critical negative regulator of antitumor immune response and is gaining growing interest as a potential pharmacological target in immunotherapy. This molecule is highly expressed in hematopoietic stem cells and myeloid compartment, and it has been found upmodulated in acute myeloid leukemia (AML). However, VISTA-associated immune features are relatively unexplored in myeloid malignancies. Herein, we aimed to explore whether this immune checkpoint regulator could play a role in the generation of an immune escape environment in AML patients. We characterized VISTA mRNA expression levels in leukemia cell lines and in large publicly available cohorts of specimens from bone marrow of healthy individuals and AML patients at diagnosis by deploying bulk and single-cell RNA sequencing. We also defined the correlations with leukemia-associated burden using results of whole-exome sequencing of AML samples at disease onset. We showed that VISTA expression linearly increased across the myeloid differentiation tree in normal hematopoiesis. Accordingly, its transcript was highly enriched in AML cell lines as well as in AML patients at diagnosis presenting with myelomonocytic and monocytic differentiation. A strong correlation was seen with NPM1 mutations regardless of the presence of FLT3 lesions. Furthermore, VISTA expression levels at baseline correlated with disease recurrence in patients with normal karyotype and NPM1 mutations, a subgroup traditionally considered as favorable according to current diagnostic schemes. Indeed, when compared to patients with long-term remission (>5 years after standard chemotherapy regimens), cases relapsing within 2 years from diagnosis had increased VISTA expression in both leukemia and T cells. Our results suggest a rationale for developing VISTA-targeted therapeutic strategies to treat molecularly defined subgroups of AML patients to prevent disease recurrence and treatment resistance. Full article
(This article belongs to the Special Issue Molecular Mechanism of Leukemia)
Show Figures

Figure 1

Review

Jump to: Editorial, Research

15 pages, 2757 KiB  
Review
S100A8 and S100A9 in Hematologic Malignancies: From Development to Therapy
by Farnaz Razmkhah, Sena Kim, Sora Lim, Abdul-Jalil Dania and Jaebok Choi
Int. J. Mol. Sci. 2023, 24(17), 13382; https://doi.org/10.3390/ijms241713382 - 29 Aug 2023
Cited by 3 | Viewed by 1571
Abstract
S100A8 and S100A9 are multifunctional proteins that can initiate various signaling pathways and modulate cell function both inside and outside immune cells, depending on their receptors, mediators, and molecular environment. They have been reported as dysregulated genes and proteins in a wide range [...] Read more.
S100A8 and S100A9 are multifunctional proteins that can initiate various signaling pathways and modulate cell function both inside and outside immune cells, depending on their receptors, mediators, and molecular environment. They have been reported as dysregulated genes and proteins in a wide range of cancers, including hematologic malignancies, from diagnosis to response to therapy. The role of S100A8 and S100A9 in hematologic malignancies is highlighted due to their ability to work together or as antagonists to modify cell phenotype, including viability, differentiation, chemosensitivity, trafficking, and transcription strategies, which can lead to an oncogenic phase or reduced symptoms. In this review article, we discuss the critical roles of S100A8, S100A9, and calprotectin (heterodimer or heterotetramer forms of S100A8 and S100A9) in forming and promoting the malignant bone marrow microenvironment. We also focus on their potential roles as biomarkers and therapeutic targets in various stages of hematologic malignancies from diagnosis to treatment. Full article
(This article belongs to the Special Issue Molecular Mechanism of Leukemia)
Show Figures

Figure 1

32 pages, 1856 KiB  
Review
Metabolic Reprogramming and Potential Therapeutic Targets in Lymphoma
by Yuyang Pang, Tingxun Lu, Zijun Y. Xu-Monette and Ken H. Young
Int. J. Mol. Sci. 2023, 24(6), 5493; https://doi.org/10.3390/ijms24065493 - 13 Mar 2023
Cited by 4 | Viewed by 3308
Abstract
Lymphoma is a heterogeneous group of diseases that often require their metabolism program to fulfill the demand of cell proliferation. Features of metabolism in lymphoma cells include high glucose uptake, deregulated expression of enzymes related to glycolysis, dual capacity for glycolytic and oxidative [...] Read more.
Lymphoma is a heterogeneous group of diseases that often require their metabolism program to fulfill the demand of cell proliferation. Features of metabolism in lymphoma cells include high glucose uptake, deregulated expression of enzymes related to glycolysis, dual capacity for glycolytic and oxidative metabolism, elevated glutamine metabolism, and fatty acid synthesis. These aberrant metabolic changes lead to tumorigenesis, disease progression, and resistance to lymphoma chemotherapy. This metabolic reprogramming, including glucose, nucleic acid, fatty acid, and amino acid metabolism, is a dynamic process caused not only by genetic and epigenetic changes, but also by changes in the microenvironment affected by viral infections. Notably, some critical metabolic enzymes and metabolites may play vital roles in lymphomagenesis and progression. Recent studies have uncovered that metabolic pathways might have clinical impacts on the diagnosis, characterization, and treatment of lymphoma subtypes. However, determining the clinical relevance of biomarkers and therapeutic targets related to lymphoma metabolism is still challenging. In this review, we systematically summarize current studies on metabolism reprogramming in lymphoma, and we mainly focus on disorders of glucose, amino acids, and lipid metabolisms, as well as dysregulation of molecules in metabolic pathways, oncometabolites, and potential metabolic biomarkers. We then discuss strategies directly or indirectly for those potential therapeutic targets. Finally, we prospect the future directions of lymphoma treatment on metabolic reprogramming. Full article
(This article belongs to the Special Issue Molecular Mechanism of Leukemia)
Show Figures

Figure 1

22 pages, 1555 KiB  
Review
IKAROS in Acute Leukemia: A Positive Influencer or a Mean Hater?
by Maria Rosa Conserva, Immacolata Redavid, Luisa Anelli, Antonella Zagaria, Francesco Tarantini, Cosimo Cumbo, Giuseppina Tota, Elisa Parciante, Nicoletta Coccaro, Crescenzio Francesco Minervini, Angela Minervini, Giorgina Specchia, Pellegrino Musto and Francesco Albano
Int. J. Mol. Sci. 2023, 24(4), 3282; https://doi.org/10.3390/ijms24043282 - 07 Feb 2023
Cited by 2 | Viewed by 2434
Abstract
One key process that controls leukemogenesis is the regulation of oncogenic gene expression by transcription factors acting as tumor suppressors. Understanding this intricate mechanism is crucial to elucidating leukemia pathophysiology and discovering new targeted treatments. In this review, we make a brief overview [...] Read more.
One key process that controls leukemogenesis is the regulation of oncogenic gene expression by transcription factors acting as tumor suppressors. Understanding this intricate mechanism is crucial to elucidating leukemia pathophysiology and discovering new targeted treatments. In this review, we make a brief overview of the physiological role of IKAROS and the molecular pathway that contributes to acute leukemia pathogenesis through IKZF1 gene lesions. IKAROS is a zinc finger transcription factor of the Krüppel family that acts as the main character during hematopoiesis and leukemogenesis. It can activate or repress tumor suppressors or oncogenes, regulating the survival and proliferation of leukemic cells. More than 70% of Ph+ and Ph-like cases of acute lymphoblastic leukemia exhibit IKZF1 gene variants, which are linked to worse treatment outcomes in both childhood and adult B-cell precursor acute lymphoblastic leukemia. In the last few years, much evidence supporting IKAROS involvement in myeloid differentiation has been reported, suggesting that loss of IKZF1 might also be a determinant of oncogenesis in acute myeloid leukemia. Considering the complicated “social” network that IKAROS manages in hematopoietic cells, we aim to focus on its involvement and the numerous alterations of molecular pathways it can support in acute leukemias. Full article
(This article belongs to the Special Issue Molecular Mechanism of Leukemia)
Show Figures

Figure 1

23 pages, 2796 KiB  
Review
Targeting and Monitoring Acute Myeloid Leukaemia with Nucleophosmin-1 (NPM1) Mutation
by Lynn Chin, Chantelle Ye Gwen Wong and Harinder Gill
Int. J. Mol. Sci. 2023, 24(4), 3161; https://doi.org/10.3390/ijms24043161 - 05 Feb 2023
Cited by 6 | Viewed by 3366
Abstract
Mutations in NPM1, also known as nucleophosmin-1, B23, NO38, or numatrin, are seen in approximately one-third of patients with acute myeloid leukaemia (AML). A plethora of treatment strategies have been studied to determine the best possible approach to curing NPM1-mutated AML. Here, [...] Read more.
Mutations in NPM1, also known as nucleophosmin-1, B23, NO38, or numatrin, are seen in approximately one-third of patients with acute myeloid leukaemia (AML). A plethora of treatment strategies have been studied to determine the best possible approach to curing NPM1-mutated AML. Here, we introduce the structure and function of NPM1 and describe the application of minimal residual disease (MRD) monitoring using molecular methods by means of quantitative polymerase chain reaction (qPCR), droplet digital PCR (ddPCR), next-generation sequencing (NGS), and cytometry by time of flight (CyTOF) to target NPM1-mutated AML. Current drugs, now regarded as the standard of care for AML, as well as potential drugs still under development, will also be explored. This review will focus on the role of targeting aberrant NPM1 pathways such as BCL-2 and SYK; as well as epigenetic regulators (RNA polymerase), DNA intercalators (topoisomerase II), menin inhibitors, and hypomethylating agents. Aside from medication, the effects of stress on AML presentation have been reported, and some possible mechanisms outlined. Moreover, targeted strategies will be briefly discussed, not only for the prevention of abnormal trafficking and localisation of cytoplasmic NPM1 but also for the elimination of mutant NPM1 proteins. Lastly, the advancement of immunotherapy such as targeting CD33, CD123, and PD-1 will be mentioned. Full article
(This article belongs to the Special Issue Molecular Mechanism of Leukemia)
Show Figures

Graphical abstract

30 pages, 2552 KiB  
Review
Understanding the Roles of the Hedgehog Signaling Pathway during T-Cell Lymphopoiesis and in T-Cell Acute Lymphoblastic Leukemia (T-ALL)
by Alberto M. Martelli, Francesca Paganelli, Serena Truocchio, Carla Palumbo, Francesca Chiarini and James A. McCubrey
Int. J. Mol. Sci. 2023, 24(3), 2962; https://doi.org/10.3390/ijms24032962 - 03 Feb 2023
Cited by 4 | Viewed by 2126
Abstract
The Hedgehog (HH) signaling network is one of the main regulators of invertebrate and vertebrate embryonic development. Along with other networks, such as NOTCH and WNT, HH signaling specifies both the early patterning and the polarity events as well as the subsequent organ [...] Read more.
The Hedgehog (HH) signaling network is one of the main regulators of invertebrate and vertebrate embryonic development. Along with other networks, such as NOTCH and WNT, HH signaling specifies both the early patterning and the polarity events as well as the subsequent organ formation via the temporal and spatial regulation of cell proliferation and differentiation. However, aberrant activation of HH signaling has been identified in a broad range of malignant disorders, where it positively influences proliferation, survival, and therapeutic resistance of neoplastic cells. Inhibitors targeting the HH pathway have been tested in preclinical cancer models. The HH pathway is also overactive in other blood malignancies, including T-cell acute lymphoblastic leukemia (T-ALL). This review is intended to summarize our knowledge of the biological roles and pathophysiology of the HH pathway during normal T-cell lymphopoiesis and in T-ALL. In addition, we will discuss potential therapeutic strategies that might expand the clinical usefulness of drugs targeting the HH pathway in T-ALL. Full article
(This article belongs to the Special Issue Molecular Mechanism of Leukemia)
Show Figures

Figure 1

21 pages, 1360 KiB  
Review
Epigenetic Modification of Cytosines in Hematopoietic Differentiation and Malignant Transformation
by Jungeun An and Myunggon Ko
Int. J. Mol. Sci. 2023, 24(2), 1727; https://doi.org/10.3390/ijms24021727 - 15 Jan 2023
Cited by 5 | Viewed by 2699
Abstract
The mammalian DNA methylation landscape is established and maintained by the combined activities of the two key epigenetic modifiers, DNA methyltransferases (DNMT) and Ten-eleven-translocation (TET) enzymes. Once DNMTs produce 5-methylcytosine (5mC), TET proteins fine-tune the DNA methylation status by consecutively oxidizing 5mC to [...] Read more.
The mammalian DNA methylation landscape is established and maintained by the combined activities of the two key epigenetic modifiers, DNA methyltransferases (DNMT) and Ten-eleven-translocation (TET) enzymes. Once DNMTs produce 5-methylcytosine (5mC), TET proteins fine-tune the DNA methylation status by consecutively oxidizing 5mC to 5-hydroxymethylcytosine (5hmC) and further oxidized derivatives. The 5mC and oxidized methylcytosines are essential for the maintenance of cellular identity and function during differentiation. Cytosine modifications with DNMT and TET enzymes exert pleiotropic effects on various aspects of hematopoiesis, including self-renewal of hematopoietic stem/progenitor cells (HSPCs), lineage determination, differentiation, and function. Under pathological conditions, these enzymes are frequently dysregulated, leading to loss of function. In particular, the loss of DNMT3A and TET2 function is conspicuous in diverse hematological disorders, including myeloid and lymphoid malignancies, and causally related to clonal hematopoiesis and malignant transformation. Here, we update recent advances in understanding how the maintenance of DNA methylation homeostasis by DNMT and TET proteins influences normal hematopoiesis and malignant transformation, highlighting the potential impact of DNMT3A and TET2 dysregulation on clonal dominance and evolution of pre-leukemic stem cells to full-blown malignancies. Clarification of the normal and pathological functions of DNA-modifying epigenetic regulators will be crucial to future innovations in epigenetic therapies for treating hematological disorders. Full article
(This article belongs to the Special Issue Molecular Mechanism of Leukemia)
Show Figures

Figure 1

14 pages, 3564 KiB  
Review
A Focused Review of Ras Guanine Nucleotide-Releasing Protein 1 in Immune Cells and Cancer
by Tu Chun Hsu, Gisele O. L. Rodrigues, Hila Winer, Julie A. Hixon, Wenqing Li, Nadya I. Tarasova and Scott K. Durum
Int. J. Mol. Sci. 2023, 24(2), 1652; https://doi.org/10.3390/ijms24021652 - 13 Jan 2023
Cited by 5 | Viewed by 2896
Abstract
Four Ras guanine nucleotide-releasing proteins (RasGRP1 through 4) belong to the family of guanine nucleotide exchange factors (GEFs). RasGRPs catalyze the release of GDP from small GTPases Ras and Rap and facilitate their transition from an inactive GDP-bound to an active GTP-bound state. [...] Read more.
Four Ras guanine nucleotide-releasing proteins (RasGRP1 through 4) belong to the family of guanine nucleotide exchange factors (GEFs). RasGRPs catalyze the release of GDP from small GTPases Ras and Rap and facilitate their transition from an inactive GDP-bound to an active GTP-bound state. Thus, they regulate critical cellular responses via many downstream GTPase effectors. Similar to other RasGRPs, the catalytic module of RasGRP1 is composed of the Ras exchange motif (REM) and Cdc25 domain, and the EF hands and C1 domain contribute to its cellular localization and regulation. RasGRP1 can be activated by a diacylglycerol (DAG)-mediated membrane recruitment and protein kinase C (PKC)-mediated phosphorylation. RasGRP1 acts downstream of the T cell receptor (TCR), B cell receptors (BCR), and pre-TCR, and plays an important role in the thymocyte maturation and function of peripheral T cells, B cells, NK cells, mast cells, and neutrophils. The dysregulation of RasGRP1 is known to contribute to numerous disorders that range from autoimmune and inflammatory diseases and schizophrenia to neoplasia. Given its position at the crossroad of cell development, inflammation, and cancer, RASGRP1 has garnered interest from numerous disciplines. In this review, we outline the structure, function, and regulation of RasGRP1 and focus on the existing knowledge of the role of RasGRP1 in leukemia and other cancers. Full article
(This article belongs to the Special Issue Molecular Mechanism of Leukemia)
Show Figures

Figure 1

Back to TopTop